Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.098
Filter
1.
Nature ; 629(8012): 704-709, 2024 May.
Article in English | MEDLINE | ID: mdl-38693257

ABSTRACT

Choline is an essential nutrient that the human body needs in vast quantities for cell membrane synthesis, epigenetic modification and neurotransmission. The brain has a particularly high demand for choline, but how it enters the brain remains unknown1-3. The major facilitator superfamily transporter FLVCR1 (also known as MFSD7B or SLC49A1) was recently determined to be a choline transporter but is not highly expressed at the blood-brain barrier, whereas the related protein FLVCR2 (also known as MFSD7C or SLC49A2) is expressed in endothelial cells at the blood-brain barrier4-7. Previous studies have shown that mutations in human Flvcr2 cause cerebral vascular abnormalities, hydrocephalus and embryonic lethality, but the physiological role of FLVCR2 is unknown4,5. Here we demonstrate both in vivo and in vitro that FLVCR2 is a BBB choline transporter and is responsible for the majority of choline uptake into the brain. We also determine the structures of choline-bound FLVCR2 in both inward-facing and outward-facing states using cryo-electron microscopy. These results reveal how the brain obtains choline and provide molecular-level insights into how FLVCR2 binds choline in an aromatic cage and mediates its uptake. Our work could provide a novel framework for the targeted delivery of therapeutic agents into the brain.


Subject(s)
Blood-Brain Barrier , Brain , Choline , Cryoelectron Microscopy , Membrane Transport Proteins , Models, Molecular , Choline/metabolism , Animals , Humans , Brain/metabolism , Mice , Blood-Brain Barrier/metabolism , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Female , Male , Biological Transport
2.
Nature ; 629(8012): 710-716, 2024 May.
Article in English | MEDLINE | ID: mdl-38693265

ABSTRACT

Phosphatidylcholine and phosphatidylethanolamine, the two most abundant phospholipids in mammalian cells, are synthesized de novo by the Kennedy pathway from choline and ethanolamine, respectively1-6. Despite the essential roles of these lipids, the mechanisms that enable the cellular uptake of choline and ethanolamine remain unknown. Here we show that the protein encoded by FLVCR1, whose mutation leads to the neurodegenerative syndrome posterior column ataxia and retinitis pigmentosa7-9, transports extracellular choline and ethanolamine into cells for phosphorylation by downstream kinases to initiate the Kennedy pathway. Structures of FLVCR1 in the presence of choline and ethanolamine reveal that both metabolites bind to a common binding site comprising aromatic and polar residues. Despite binding to a common site, FLVCR1 interacts in different ways with the larger quaternary amine of choline in and with the primary amine of ethanolamine. Structure-guided mutagenesis identified residues that are crucial for the transport of ethanolamine, but dispensable for choline transport, enabling functional separation of the entry points into the two branches of the Kennedy pathway. Altogether, these studies reveal how FLVCR1 is a high-affinity metabolite transporter that serves as the common origin for phospholipid biosynthesis by two branches of the Kennedy pathway.


Subject(s)
Choline , Ethanolamine , Membrane Transport Proteins , Models, Molecular , Humans , Choline/metabolism , Binding Sites , Ethanolamine/metabolism , Ethanolamine/chemistry , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Phosphatidylethanolamines/metabolism , Biological Transport , Animals , Phosphatidylcholines/metabolism , Phosphatidylcholines/chemistry , Phosphorylation
3.
Commun Biol ; 7(1): 610, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773269

ABSTRACT

The processes of nutrient uptake and signal sensing are crucial for microbial survival and adaptation. Membrane-embedded proteins involved in these functions (transporters and receptors) are commonly regarded as unrelated in terms of sequence, structure, mechanism of action and evolutionary history. Here, we analyze the protein structural universe using recently developed artificial intelligence-based structure prediction tools, and find an unexpected link between prominent groups of microbial transporters and receptors. The so-called S-components of Energy-Coupling Factor (ECF) transporters, and the membrane domains of sensor histidine kinases of the 5TMR cluster share a structural fold. The discovery of their relatedness manifests a widespread case of prokaryotic "transceptors" (related proteins with transport or receptor function), showcases how artificial intelligence-based structure predictions reveal unchartered evolutionary connections between proteins, and provides new avenues for engineering transport and signaling functions in bacteria.


Subject(s)
Bacterial Proteins , Membrane Transport Proteins , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Histidine Kinase/metabolism , Histidine Kinase/chemistry , Histidine Kinase/genetics , Models, Molecular , Bacteria/metabolism , Bacteria/genetics , Signal Transduction , Protein Folding , Artificial Intelligence
4.
J Chem Inf Model ; 64(8): 3524-3536, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38564295

ABSTRACT

Understanding the conformational dynamics of proteins, such as the inward-facing (IF) and outward-facing (OF) transition observed in transporters, is vital for elucidating their functional mechanisms. Despite significant advances in protein structure prediction (PSP) over the past three decades, most efforts have been focused on single-state prediction, leaving multistate or alternative conformation prediction (ACP) relatively unexplored. This discrepancy has led to the development of highly accurate PSP methods such as AlphaFold, yet their capabilities for ACP remain limited. To investigate the performance of current PSP methods in ACP, we curated a data set, named IOMemP, consisting of 32 experimentally determined high-resolution IF and OF structures of 16 membrane proteins with substantial conformational changes. We benchmarked 12 representative PSP methods, along with two recent multistate methods based on AlphaFold, against this data set. Our findings reveal a remarkably consistent preference for specific states across various PSP methods. We elucidated how coevolution information in MSAs influences state preference. Moreover, we showed that AlphaFold, when excluding coevolution information, estimated similar energies between the experimental IF and OF conformations, indicating that the energy model learned by AlphaFold is not biased toward any particular state. Our IOMemP data set and benchmark results are anticipated to advance the development of robust ACP methods.


Subject(s)
Membrane Transport Proteins , Protein Conformation , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Models, Molecular , Databases, Protein
5.
Nature ; 625(7995): 572-577, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38172635

ABSTRACT

Gram-negative bacteria are extraordinarily difficult to kill because their cytoplasmic membrane is surrounded by an outer membrane that blocks the entry of most antibiotics. The impenetrable nature of the outer membrane is due to the presence of a large, amphipathic glycolipid called lipopolysaccharide (LPS) in its outer leaflet1. Assembly of the outer membrane requires transport of LPS across a protein bridge that spans from the cytoplasmic membrane to the cell surface. Maintaining outer membrane integrity is essential for bacterial cell viability, and its disruption can increase susceptibility to other antibiotics2-6. Thus, inhibitors of the seven lipopolysaccharide transport (Lpt) proteins that form this transenvelope transporter have long been sought. A new class of antibiotics that targets the LPS transport machine in Acinetobacter was recently identified. Here, using structural, biochemical and genetic approaches, we show that these antibiotics trap a substrate-bound conformation of the LPS transporter that stalls this machine. The inhibitors accomplish this by recognizing a composite binding site made up of both the Lpt transporter and its LPS substrate. Collectively, our findings identify an unusual mechanism of lipid transport inhibition, reveal a druggable conformation of the Lpt transporter and provide the foundation for extending this class of antibiotics to other Gram-negative pathogens.


Subject(s)
Anti-Bacterial Agents , Bacterial Outer Membrane Proteins , Lipopolysaccharides , Membrane Transport Proteins , Acinetobacter/chemistry , Acinetobacter/drug effects , Acinetobacter/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Binding Sites/drug effects , Biological Transport/drug effects , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Lipopolysaccharides/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Microbial Viability , Protein Conformation/drug effects , Substrate Specificity
6.
J Phys Chem Lett ; 15(3): 725-732, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38215403

ABSTRACT

Transporter proteins change their conformations to carry their substrate across the cell membrane. The conformational dynamics is vital to understanding the transport function. We have studied the oxalate transporter (OxlT), an oxalate:formate antiporter from Oxalobacter formigenes, significant in avoiding kidney stone formation. The atomic structure of OxlT has been recently solved in the outward-open and occluded states. However, the inward-open conformation is still missing, hindering a complete understanding of the transporter. Here, we performed a Gaussian accelerated molecular dynamics simulation to sample the extensive conformational space of OxlT and successfully predicted the inward-open conformation where cytoplasmic substrate formate binding was preferred over oxalate binding. We also identified critical interactions for the inward-open conformation. The results were complemented by an AlphaFold2 structure prediction. Although AlphaFold2 solely predicted OxlT in the outward-open conformation, mutation of the identified critical residues made it partly predict the inward-open conformation, identifying possible state-shifting mutations.


Subject(s)
Molecular Dynamics Simulation , Oxalates , Oxalates/chemistry , Oxalates/metabolism , Membrane Transport Proteins/chemistry , Antiporters/metabolism , Formates/metabolism , Protein Conformation
7.
J Chem Inf Model ; 64(5): 1433-1455, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38294194

ABSTRACT

Solute carrier transporters (SLCs) are a class of important transmembrane proteins that are involved in the transportation of diverse solute ions and small molecules into cells. There are approximately 450 SLCs within the human body, and more than a quarter of them are emerging as attractive therapeutic targets for multiple complex diseases, e.g., depression, cancer, and diabetes. However, only 44 unique transporters (∼9.8% of the SLC superfamily) with 3D structures and specific binding sites have been reported. To design innovative and effective drugs targeting diverse SLCs, there are a number of obstacles that need to be overcome. However, computational chemistry, including physics-based molecular modeling and machine learning- and deep learning-based artificial intelligence (AI), provides an alternative and complementary way to the classical drug discovery approach. Here, we present a comprehensive overview on recent advances and existing challenges of the computational techniques in structure-based drug design of SLCs from three main aspects: (i) characterizing multiple conformations of the proteins during the functional process of transportation, (ii) identifying druggability sites especially the cryptic allosteric ones on the transporters for substrates and drugs binding, and (iii) discovering diverse small molecules or synthetic protein binders targeting the binding sites. This work is expected to provide guidelines for a deep understanding of the structure and function of the SLC superfamily to facilitate rational design of novel modulators of the transporters with the aid of state-of-the-art computational chemistry technologies including artificial intelligence.


Subject(s)
Artificial Intelligence , Computational Chemistry , Humans , Membrane Transport Proteins/chemistry , Drug Design , Drug Discovery/methods
8.
J Phys Chem B ; 128(3): 622-634, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38047375

ABSTRACT

Efflux pumps are specialized transport proteins that play a key role in the bacterial defense against a wide spectrum of antibiotics. Hence, understanding the biophysical mechanism associated with this complex system of drug expulsion becomes crucial. This work deals with some vital aspects of the outer membrane factor (OMF) of MexAB-OprM. After being passed through MexB and MexA, efflux substrates have to go through OprM for their final judgment. Thus, it is very important to understand the periplasmic pore opening mechanism and the associated biophysical changes during this process. Our study captures a detailed analysis of the pore opening mechanism involving OprM. With powerful molecular dynamics (MD) techniques such as well-tempered metadynamics, the presence of metastable states in between open and closed states was confirmed. Also, upon mutating R376, the energy barrier for the conversion of the close to open conformation decreases, indicating an important role played by the residue. Further, constant pH MD was performed to capture the effect of pH in both conformations. OprM exhibits distinct conformational states at pH values greater than 5.5 and lower than 5.5, suggesting its pH-responsive characteristics. Overall, our study elucidates a crucial undertaking toward discovering potential inhibitors for MexAB-OprM efflux pumps.


Subject(s)
Bacterial Outer Membrane Proteins , Membrane Transport Proteins , Membrane Transport Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Carrier Proteins/metabolism , Hydrogen-Ion Concentration , Pseudomonas aeruginosa/metabolism , Microbial Sensitivity Tests
9.
Nat Chem Biol ; 20(1): 103-110, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37872402

ABSTRACT

Plants sense abscisic acid (ABA) using chemical-induced dimerization (CID) modules, including the receptor PYR1 and HAB1, a phosphatase inhibited by ligand-activated PYR1. This system is unique because of the relative ease with which ligand recognition can be reprogrammed. To expand the PYR1 system, we designed an orthogonal '*' module, which harbors a dimer interface salt bridge; X-ray crystallographic, biochemical and in vivo analyses confirm its orthogonality. We used this module to create PYR1*MANDI/HAB1* and PYR1*AZIN/HAB1*, which possess nanomolar sensitivities to their activating ligands mandipropamid and azinphos-ethyl. Experiments in Arabidopsis thaliana and Saccharomyces cerevisiae demonstrate the sensitive detection of banned organophosphate contaminants using living biosensors and the construction of multi-input/output genetic circuits. Our new modules enable ligand-programmable multi-channel CID systems for plant and eukaryotic synthetic biology that can empower new plant-based and microbe-based sensing modalities.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Abscisic Acid , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Dimerization , Ligands , Membrane Transport Proteins/chemistry
10.
Biochimie ; 220: 167-178, 2024 May.
Article in English | MEDLINE | ID: mdl-38158037

ABSTRACT

Candida albicans and C. glabrata express exporters of the ATP-binding cassette (ABC) superfamily and address them to their plasma membrane to expel azole antifungals, which cancels out their action and allows the yeast to become multidrug resistant (MDR). In a way to understand this mechanism of defense, we describe the purification and characterization of Cdr1, the membrane ABC exporter mainly responsible for such phenotype in both species. Cdr1 proteins were functionally expressed in the baker yeast, tagged at their C-terminal end with either a His-tag for the glabrata version, cgCdr1-His, or a green fluorescent protein (GFP) preceded by a proteolytic cleavage site for the albicans version, caCdr1-P-GFP. A membrane Cdr1-enriched fraction was then prepared to assay several detergents and stabilizers, probing their level of extraction and the ATPase activity of the proteins as a functional marker. Immobilized metal-affinity and size-exclusion chromatographies (IMAC, SEC) were then carried out to isolate homogenous samples. Overall, our data show that although topologically and phylogenetically close, both proteins display quite distinct behaviors during the extraction and purification steps, and qualify cgCdr1 as a good candidate to characterize this type of proteins for developing future inhibitors of their azole antifungal efflux activity.


Subject(s)
Antifungal Agents , Azoles , Candida albicans , Drug Resistance, Fungal , Fungal Proteins , Membrane Transport Proteins , Azoles/pharmacology , Azoles/chemistry , Azoles/metabolism , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/isolation & purification , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/isolation & purification , Candida albicans/drug effects , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Candida glabrata/drug effects , Candida glabrata/genetics , Candida glabrata/metabolism , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/chemistry
11.
Trends Biochem Sci ; 49(2): 134-144, 2024 02.
Article in English | MEDLINE | ID: mdl-38102017

ABSTRACT

Tripartite ATP-independent periplasmic (TRAP) transporters are nutrient-uptake systems found in bacteria and archaea. These evolutionary divergent transporter systems couple a substrate-binding protein (SBP) to an elevator-type secondary transporter, which is a first-of-its-kind mechanism of transport. Here, we highlight breakthrough TRAP transporter structures and recent functional data that probe the mechanism of transport. Furthermore, we discuss recent structural and biophysical studies of the ion transporter superfamily (ITS) members and highlight mechanistic principles that are relevant for further exploration of the TRAP transporter system.


Subject(s)
Bacterial Proteins , Membrane Transport Proteins , Bacterial Proteins/metabolism , Membrane Transport Proteins/chemistry , Carrier Proteins/metabolism , Bacteria/metabolism , Biological Transport
12.
Structure ; 31(11): 1419-1430.e5, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37708891

ABSTRACT

The insertion and folding of proteins into membranes is crucial for cell viability. Yet, the detailed contributions of insertases remain elusive. Here, we monitor how the insertase YidC guides the folding of the polytopic melibiose permease MelB into membranes. In vivo experiments using conditionally depleted E. coli strains show that MelB can insert in the absence of SecYEG if YidC resides in the cytoplasmic membrane. In vitro single-molecule force spectroscopy reveals that the MelB substrate itself forms two folding cores from which structural segments insert stepwise into the membrane. However, misfolding dominates, particularly in structural regions that interface the pseudo-symmetric α-helical domains of MelB. Here, YidC takes an important role in accelerating and chaperoning the stepwise insertion and folding process of both MelB folding cores. Our findings reveal a great flexibility of the chaperoning and insertase activity of YidC in the multifaceted folding processes of complex polytopic membrane proteins.


Subject(s)
Escherichia coli Proteins , Membrane Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Cell Membrane/metabolism
13.
Nat Struct Mol Biol ; 30(12): 1996-2008, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37696957

ABSTRACT

Over half of mitochondrial proteins are imported from the cytosol via the pre-sequence pathway, controlled by the TOM complex in the outer membrane and the TIM23 complex in the inner membrane. The mechanisms through which proteins are translocated via the TOM and TIM23 complexes remain unclear. Here we report the assembly of the active TOM-TIM23 supercomplex of Saccharomyces cerevisiae with translocating polypeptide substrates. Electron cryo-microscopy analyses reveal that the polypeptide substrates pass the TOM complex through the center of a Tom40 subunit, interacting with a glutamine-rich region. Structural and biochemical analyses show that the TIM23 complex contains a heterotrimer of the subunits Tim23, Tim17 and Mgr2. The polypeptide substrates are shielded from lipids by Mgr2 and Tim17, which creates a translocation pathway characterized by a negatively charged entrance and a central hydrophobic region. These findings reveal an unexpected pre-sequence pathway through the TOM-TIM23 supercomplex spanning the double membranes of mitochondria.


Subject(s)
Membrane Transport Proteins , Saccharomyces cerevisiae Proteins , Membrane Transport Proteins/chemistry , Mitochondrial Precursor Protein Import Complex Proteins , Carrier Proteins/metabolism , Mitochondrial Membrane Transport Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Protein Transport , Mitochondria/metabolism , Saccharomyces cerevisiae/metabolism , Mitochondrial Proteins/metabolism , Peptides/metabolism , Membrane Proteins/metabolism
14.
Nature ; 620(7973): 445-452, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37495693

ABSTRACT

To replicate inside macrophages and cause tuberculosis, Mycobacterium tuberculosis must scavenge a variety of nutrients from the host1,2. The mammalian cell entry (MCE) proteins are important virulence factors in M. tuberculosis1,3, where they are encoded by large gene clusters and have been implicated in the transport of fatty acids4-7 and cholesterol1,4,8 across the impermeable mycobacterial cell envelope. Very little is known about how cargos are transported across this barrier, and it remains unclear how the approximately ten proteins encoded by a mycobacterial mce gene cluster assemble to transport cargo across the cell envelope. Here we report the cryo-electron microscopy (cryo-EM) structure of the endogenous Mce1 lipid-import machine of Mycobacterium smegmatis-a non-pathogenic relative of M. tuberculosis. The structure reveals how the proteins of the Mce1 system assemble to form an elongated ABC transporter complex that is long enough to span the cell envelope. The Mce1 complex is dominated by a curved, needle-like domain that appears to be unrelated to previously described protein structures, and creates a protected hydrophobic pathway for lipid transport across the periplasm. Our structural data revealed the presence of a subunit of the Mce1 complex, which we identified using a combination of cryo-EM and AlphaFold2, and name LucB. Our data lead to a structural model for Mce1-mediated lipid import across the mycobacterial cell envelope.


Subject(s)
Bacterial Proteins , Cryoelectron Microscopy , Lipids , Membrane Transport Proteins , Mycobacterium tuberculosis , Virus Internalization , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/ultrastructure , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/ultrastructure , Tuberculosis/microbiology , Virulence Factors/chemistry , Virulence Factors/metabolism , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/ultrastructure , Periplasm/metabolism , Protein Domains , Hydrophobic and Hydrophilic Interactions , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure
15.
Microbiology (Reading) ; 169(7)2023 07.
Article in English | MEDLINE | ID: mdl-37450375

ABSTRACT

The exchange of ammonium across cellular membranes is a fundamental process in all domains of life. In plants, bacteria and fungi, ammonium represents a vital source of nitrogen, which is scavenged from the external environment. In contrast, in animal cells ammonium is a cytotoxic metabolic waste product and must be excreted to prevent cell death. Transport of ammonium is facilitated by the ubiquitous Amt/Mep/Rh transporter superfamily. In addition to their function as transporters, Amt/Mep/Rh proteins play roles in a diverse array of biological processes and human physiopathology. Despite this clear physiological importance and medical relevance, the molecular mechanism of Amt/Mep/Rh proteins has remained elusive. Crystal structures of bacterial Amt/Rh proteins suggest electroneutral transport, whilst functional evidence supports an electrogenic mechanism. Here, focusing on bacterial members of the family, we summarize the structure of Amt/Rh proteins and what three decades of research tells us concerning the general mechanisms of ammonium translocation, in particular the possibility that the transport mechanism might differ in various members of the Amt/Mep/Rh superfamily.


Subject(s)
Ammonium Compounds , Animals , Humans , Ammonium Compounds/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/chemistry , Bacteria/genetics , Bacteria/metabolism , Nitrogen/metabolism , Fungi/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
16.
EMBO J ; 42(16): e113418, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37458117

ABSTRACT

Efflux of antibacterial compounds is a major mechanism for developing antimicrobial resistance. In the Gram-positive pathogen Staphylococcus aureus, QacA, a 14 transmembrane helix containing major facilitator superfamily antiporter, mediates proton-coupled efflux of mono and divalent cationic antibacterial compounds. In this study, we report the cryo-EM structure of QacA, with a single mutation D411N that improves homogeneity and retains efflux activity against divalent cationic compounds like dequalinium and chlorhexidine. The structure of substrate-free QacA, complexed to two single-domain camelid antibodies, was elucidated to a resolution of 3.6 Å. The structure displays an outward-open conformation with an extracellular helical hairpin loop (EL7) between transmembrane helices 13 and 14, which is conserved in a subset of DHA2 transporters. Removal of the EL7 hairpin loop or disrupting the interface formed between EL7 and EL1 compromises efflux activity. Chimeric constructs of QacA with a helical hairpin and EL1 grafted from other DHA2 members, LfrA and SmvA, restore activity in the EL7 deleted QacA revealing the allosteric and vital role of EL7 hairpin in antibacterial efflux in QacA and related members.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Cryoelectron Microscopy , Bacterial Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/chemistry , Anti-Bacterial Agents/pharmacology
17.
Proteins ; 91(9): 1205-1221, 2023 09.
Article in English | MEDLINE | ID: mdl-37455426

ABSTRACT

Mastoparan B (MP-B) is an amphiphilic peptide with a potent antimicrobial activity against most Gram-negative bacteria. However, there is little information available on the inhibition of the Acinetobacter baumannii resistance-nodulation-cell-division (RND) efflux pump using this antimicrobial peptide. Here, we carried out a series of in-silico experiments to find the mechanisms underlying the anti-efflux activity of MP-B using a multi-drug resistant (MDR) strain of A. baumannii (AB). According to our findings, MP-B demonstrated a potent antibacterial activity against an MDR-AB (minimum inhibitory concentration [MIC] = 1 µg/mL) followed by a 20-fold reduction in the adeB gene expression in the presence of sub-MIC of this peptide. Using Groningen Machine for Chemicals Simulation (GROMACS) via PyMOL Graphical User Interface (GUI), (we observed that, the AdeB transporter had conserved helix-turn-helix regions and a tight pore rich in Phe and Ala residues. To understand how inhibition of the AdeB is achieved, we generated 20 apo-MP-B poses using the InterPep and SiteMap tools. The high-quality model was created by homology modeling and used for docking via AutoDock/Vina to identify the MP-B binding sites. We established that the most apo-MP-B formed H-bonds to the backbone of five amino acids in the Helix-5. As a result, the dihedral angles of the involved amino acids shift by 9.0-9.6 Ǻ, causing a change in the conformation of the AdeB protein. This led to helix conformation stereoisomerization and block the AdeB activity. MP-B presumably has dual mechanisms. (1) It blocks the AdeB transporter by changing its conformation. (2) MP-B influences the adeB gene expression by binding to G-protein which laterally controls efflux regulators like MarA, RamA, SoxS, and Rob proteins.


Subject(s)
Acinetobacter baumannii , Acinetobacter baumannii/genetics , Acinetobacter baumannii/metabolism , Bacterial Proteins/chemistry , Anti-Bacterial Agents/chemistry , Membrane Transport Proteins/chemistry , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Gene Expression
18.
Trends Biochem Sci ; 48(9): 801-814, 2023 09.
Article in English | MEDLINE | ID: mdl-37355450

ABSTRACT

Solute carrier (SLCs) transporters mediate the transport of a broad range of solutes across biological membranes. Dysregulation of SLCs has been associated with various pathologies, including metabolic and neurological disorders, as well as cancer and rare diseases. SLCs are therefore emerging as key targets for therapeutic intervention with several recently approved drugs targeting these proteins. Unlocking this large and complex group of proteins is essential to identifying unknown SLC targets and developing next-generation SLC therapeutics. Recent progress in experimental and computational techniques has significantly advanced SLC research, including drug discovery. Here, we review emerging topics in therapeutic discovery of SLCs, focusing on state-of-the-art approaches in structural, chemical, and computational biology, and discuss current challenges in transporter drug discovery.


Subject(s)
Neoplasms , Solute Carrier Proteins , Humans , Solute Carrier Proteins/chemistry , Solute Carrier Proteins/metabolism , Membrane Transport Proteins/chemistry , Biological Transport/physiology , Drug Discovery/methods , Neoplasms/metabolism
19.
Protein Sci ; 32(7): e4704, 2023 07.
Article in English | MEDLINE | ID: mdl-37312651

ABSTRACT

Pulse EPR measurements provide information on distances and distance distributions in proteins but require the incorporation of pairs of spin labels that are usually attached to engineered cysteine residues. In previous work, we demonstrated that efficient in vivo labeling of the Escherichia coli outer membrane vitamin B12 transporter, BtuB, could only be achieved using strains defective in the periplasmic disulfide bond formation (Dsb) system. Here, we extend these in vivo measurements to FecA, the E. coli ferric citrate transporter. As seen for BtuB, pairs of cysteines cannot be labeled when the protein is present in a standard expression strain. However, incorporating plasmids that permit an arabinose induced expression of FecA into a strain defective in the thiol disulfide oxidoreductase, DsbA, enables efficient spin-labeling and pulse EPR of FecA in cells. A comparison of the measurements made on FecA in cells with measurements made in reconstituted phospholipid bilayers suggests that the cellular environment alters the behavior of the extracellular loops of FecA. In addition to these in situ EPR measurements, the use of a DsbA minus strain for the expression of BtuB improves the EPR signals and pulse EPR data obtained in vitro from BtuB that is labeled, purified, and reconstituted into phospholipid bilayers. The in vitro data also indicate the presence of intermolecular BtuB-BtuB interactions, which had not previously been observed in a reconstituted bilayer system. This result suggests that in vitro EPR measurements on other outer membrane proteins would benefit from protein expression in a DsbA minus strain.


Subject(s)
Escherichia coli Proteins , Membrane Transport Proteins , Membrane Transport Proteins/chemistry , Escherichia coli/metabolism , Disulfides/metabolism , Bacterial Outer Membrane Proteins/chemistry , Escherichia coli Proteins/chemistry , Electron Spin Resonance Spectroscopy/methods , Spin Labels , Molecular Chaperones/metabolism , Receptors, Cell Surface/chemistry
20.
Chembiochem ; 24(15): e202300156, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37170829

ABSTRACT

Bacterial acquisition of metabolites is largely facilitated by transporters with unique substrate scopes. The tripartite ATP-independent periplasmic (TRAP) transporters comprise a large family of bacterial proteins that facilitate the uptake of a variety of small molecules. It has been reported that some TRAP systems encode a fourth protein, the T component. The T-component, or TatT, is predicted to be a periplasmic-facing lipoprotein that enables the uptake of metabolites from the outer membrane. However, no substrates were revealed for any TatT and their functional role(s) remained enigmatic. We recently identified a homolog in Methylococcus capsulatus that binds to sterols, and herein, we report two additional homologs that demonstrate a preference for long-chain fatty acids. Our bioinformatics, quantitative analyses of protein-ligand interactions, and high-resolution crystal structures suggest that TatTs might facilitate the trafficking of hydrophobic or lipophilic substrates and represent a new class of bacterial lipid and fatty acid transporters.


Subject(s)
Bacteria , Membrane Transport Proteins , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Bacteria/metabolism , Bacterial Proteins/metabolism , Biological Transport , Fatty Acids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...