1
|
Jung WH, Lee SY, Lee Y, Ahn DJ. Freezing-driven ionic charge imbalance leads to pore formation and osmotic injury of lipid membranes. Comput Biol Med 2025; 189:109960. [PMID: 40031106 DOI: 10.1016/j.compbiomed.2025.109960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
In this work, we reveal the fundamental mechanism controlling osmotic injury of lipid membranes under low-temperature preservation using an in-vitro membrane system under freezing temperature and the molecular dynamic simulations. The freezing-driven ionic charge imbalance is the major factor affecting the membrane conformation and causing the osmotic injury. Under freezing temperature, the ionic charge imbalance, originating from the preferential incorporation of anions into the growing ice crystals, results in membrane poration with the directional penetration of ion molecules. Subsequently, the osmotic efflux of water molecules through the pore causes cell dehydration, eventually leading to the lethal osmotic injury of lipid membranes during freezing. Moreover, we find a stark difference in tolerance to freezing and the times required for pore formation in membranes with different lipid compositions. Membranes enriched with cholesterol and anionic lipids exhibit increased resistance to freezing-induced osmotic injury, as the addition of cholesterol and anionic lipids in membranes delays the pore formation under freezing temperature. These findings advance in depth the molecular-level understanding of freezing injury on lipid membranes and provide an opportunity to develop an alternative strategy to protect diverse cells during preservation at subzero temperatures by regulating the composition of lipid membranes.
Collapse
Affiliation(s)
- Woo Hyuk Jung
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, South Korea
| | - Sang Yup Lee
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Yedam Lee
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, South Korea
| | - Dong June Ahn
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, South Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea; Center for Theragnosis, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea.
| |
Collapse
|
2
|
Sardag I, Duvenci ZS, Belkaya S, Timucin E. Computational modeling of the anti-inflammatory complexes of IL37. J Mol Graph Model 2025; 136:108952. [PMID: 39854883 DOI: 10.1016/j.jmgm.2025.108952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Interleukin (IL) 37 is an anti-inflammatory cytokine belonging to the IL1 protein family. Owing to its pivotal role in modulating immune responses, elucidating the IL37 complex structures holds substantial therapeutic promise for various autoimmune disorders and cancers. However, none of the structures of IL37 complexes have been experimentally characterized. This computational study aims to address this gap through molecular modeling and classical molecular dynamics simulations. We modeled all protein-protein complexes of IL37 using a range of methods from homology modeling to AlphaFold2 multimer predictions. Models that successfully recapitulated experimental features underwent further analysis through molecular dynamics simulations. As positive controls, binary and ternary complexes of IL18 from PDB were included for comparison. Several key findings emerged from the comparative analysis of IL37 and IL18 complexes. IL37 complexes exhibited higher mobility than the IL18 complexes. Simulations of the IL37-IL18Rα complex revealed altered receptor conformations capable of accommodating a dimeric IL37, with the N-terminal loop of IL37 contributing significantly to complex mobility. Additionally, the glycosyl chain on N297 of IL18Rα, which contours one edge of the cytokine binding surface, acted as a steric block against the N-terminal loop of IL37. Further, investigations into interactions between IL37 and IL18BP suggested that a binding mode homologous to IL18 was unstable for IL37, indicating an alternative binding mechanism. Altogether, this study accesses to the structure and dynamics of IL37 complexes, revealing the structural underpinnings of the IL37's modulatory effect on the IL18 signaling pathway.
Collapse
Affiliation(s)
- Inci Sardag
- Bogazici University, Department of Molecular Biology and Genetics, Istanbul 34342, Turkey
| | - Zeynep Sevval Duvenci
- Acibadem University, Institute of Health Sciences Department of Biostatistics and Bioinformatics, Istanbul 34752, Turkey
| | - Serkan Belkaya
- Bilkent University, Department of Molecular Biology and Genetics, Ankara 06800, Turkey
| | - Emel Timucin
- Acibadem University, Institute of Health Sciences Department of Biostatistics and Bioinformatics, Istanbul 34752, Turkey; Acibadem University, School of Medicine Biostatistics and Medical Informatics, Istanbul 34752, Turkey.
| |
Collapse
|
3
|
Liu Y, Brown CM, Borges N, Nobre RN, Erramilli S, Belcher Dufrisne M, Kloss B, Giacometti S, Esteves AM, Timóteo CG, Tokarz P, Cater RJ, Lowary TL, Morita YS, Kossiakoff AA, Santos H, Stansfeld PJ, Nygaard R, Mancia F. Mechanistic studies of mycobacterial glycolipid biosynthesis by the mannosyltransferase PimE. Nat Commun 2025; 16:3974. [PMID: 40301322 DOI: 10.1038/s41467-025-57843-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/05/2025] [Indexed: 05/01/2025] Open
Abstract
Tuberculosis (TB), a leading cause of death among infectious diseases globally, is caused by Mycobacterium tuberculosis (Mtb). The pathogenicity of Mtb is largely attributed to its complex cell envelope, which includes a class of glycolipids called phosphatidyl-myo-inositol mannosides (PIMs). These glycolipids maintain the integrity of the cell envelope, regulate permeability, and mediate host-pathogen interactions. PIMs comprise a phosphatidyl-myo-inositol core decorated with one to six mannose residues and up to four acyl chains. The mannosyltransferase PimE catalyzes the transfer of the fifth PIM mannose residue from a polyprenyl phosphate-mannose (PPM) donor. This step contributes to the proper assembly and function of the mycobacterial cell envelope; however, the structural basis for substrate recognition and the catalytic mechanism of PimE remain poorly understood. Here, we present the cryo-electron microscopy (cryo-EM) structures of PimE from Mycobacterium abscessus in its apo and product-bound form. The structures reveal a distinctive binding cavity that accommodates both donor and acceptor substrates/products. Key residues involved in substrate coordination and catalysis were identified and validated via in vitro assays and in vivo complementation, while molecular dynamics simulations delineated access pathways and binding dynamics. Our integrated approach provides comprehensive insights into PimE function and informs potential strategies for anti-TB therapeutics.
Collapse
Affiliation(s)
- Yaqi Liu
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Chelsea M Brown
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry, UK
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh, The Netherlands
| | - Nuno Borges
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB NOVA, Universidade Nova de Lisboa, Oeiras, Portugal
- Marine and Environmental Sciences Centre, Escola Superior de Tecnologia, Instituto Politécnico de Setúbal, Setúbal, Portugal
| | - Rodrigo N Nobre
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB NOVA, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Satchal Erramilli
- Department of Biochemistry and Molecular Biophysics, University of Chicago, Chicago, IL, USA
| | - Meagan Belcher Dufrisne
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Brian Kloss
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Sabrina Giacometti
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- School of Medicine, New York University, New York, NY, USA
| | - Ana M Esteves
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB NOVA, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Cristina G Timóteo
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB NOVA, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Piotr Tokarz
- Department of Biochemistry and Molecular Biophysics, University of Chicago, Chicago, IL, USA
| | - Rosemary J Cater
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Todd L Lowary
- Institute of Biological Chemistry, Academia Sinica, Nangang, Taipei, Taiwan
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yasu S Morita
- Department of Microbiology, University of Massachusetts, Amherst, MA, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biophysics, University of Chicago, Chicago, IL, USA
| | - Helena Santos
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB NOVA, Universidade Nova de Lisboa, Oeiras, Portugal.
| | - Phillip J Stansfeld
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry, UK.
| | - Rie Nygaard
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA.
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
4
|
Jafari M, Sagresti L, Hu J, Merz KM. Ion-Induced Dipole Interactions Matter in Metadynamics Simulation of Transition Metal Ion Transporters. J Chem Theory Comput 2025; 21:4221-4235. [PMID: 40179291 PMCID: PMC12020355 DOI: 10.1021/acs.jctc.4c01535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 04/05/2025]
Abstract
Metal transporters play crucial roles in the homeostasis and detoxification of beneficial and toxic metals in the human body. Due to experimental limitations in studying some metal transporters, numerous simulation studies have been conducted to understand the mechanisms of metal transport. However, studying the transport of divalent metal ions across the plasma membrane by metal transporters has been challenging with traditional molecular dynamics (MD) simulations. The metal ions often become trapped inside the transporter due to encountering high energy barriers during the transport process. In this study, we combined a recently developed metadynamics setup, known as well-tempered (WT) volume-based MTD, with the 12-6-4 Lennard-Jones (LJ) model representing transition metal-His/Asp/Glu side chain interactions. We used this approach to investigate the mechanism of action of a Zrt-/Irt-like protein (ZIP) transporter and compared the results with simulations using standard 12-6 LJ parameters for the transition metal-His/Asp/Glu side chain interactions. Our results show that the 12-6-4 LJ model for transition metal-His/Asp/Glu side chain interactions samples conformational space more broadly than the standard 12-6 LJ model for the same interactions in MTD simulations, facilitating the sampling of states that are hard to reach with the standard 12-6 model within the same time scale. This is even more remarkable given the fact that the model is dominated by 12-6 LJ interactions for the majority of the system, while the transition metal-His/Asp/Glu side chain interactions are the only interactions using the 12-6-4 LJ model. Hence, a small subset of interactions significantly modifies the states sampled by the entire protein leading to a more frequent observation of the transport of the transition metal ion. Overall, using 12-6-4 LJ to model the transition metal-His/Asp/Glu side chain interactions increases the potential for discovering additional metastable states by enabling metal ions to traverse more freely along the defined transport pathways.
Collapse
Affiliation(s)
- Majid Jafari
- Department
of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Luca Sagresti
- Scuola
Normale Superiore, Piazza dei Cavalieri 7, I-56126 Pisa, Italy and CSGI
- Istituto
Nazionale di Fisica Nucleare (INFN) sezione di Pisa, Largo Bruno Pontecorvo 3, 56127 Pisa, Italy
| | - Jian Hu
- Department
of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Kenneth M. Merz
- Department
of Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
5
|
Prejanò M, Tonazzi A, Giangregorio N, Indiveri C, Marino T. Lanthanides Gd and Tm Can Inhibit Carnitine/Acylcarnitine Transporter: Insights from All-Atoms Simulations. Chembiochem 2025; 26:e202401018. [PMID: 39906981 DOI: 10.1002/cbic.202401018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 02/06/2025]
Abstract
Recent experimental evidence highlighted the inhibition of carnitine/acylcarnitine carrier (CAC), an important mitochondrial transmembrane protein for living organisms, by the early lanthanide Pr3+. A possible explanation of such a behaviour was found in the preference of the cation for amino acids like aspartate and glutamate containing a carboxylate in the side chain, laying in the inter-membrane space. Interaction of the cation with these residues can cause halt the transfer of the protein's substrates between the matrix and cytoplasm thus opening to new scenarios concerning the CAC-metal interactions and its relative inhibition. In the present work, the panel of metals binding the CAC protein is predictively expanded including Gd3+ and Tm3+, selected as representative species of middle and late lanthanides, respectively. A more realistic membrane-containing model of the protein was built and the comparative analysis of the molecular dynamics (MD) simulations of CAC apo-form with its complexed systems, named CAC-Pr, CAC-Gd and CAC-Tm, was performed. The analysis of the trajectories revealed that the inhibition is caused by the coordination of D132 and E179 to the cations and that such interactions generate a reorganization of important salt-bridges inside the framework of CAC. In detail, MD simulations highlighted that a spontaneous conformational change from cytoplasmatic-state (c-state) to matrix-state (m-state) induced by cations and that, in this condition, the protein channel is occluded, thus explaining the inhibition.
Collapse
Affiliation(s)
- Mario Prejanò
- Department of Chemistry and Chemical Technologies, University of Calabria, Via Pietro Bucci 14/C, 87036, Arcavacata di Rende, CS, Italy
| | - Annamaria Tonazzi
- National Research Council (CNR), Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126, Bari, Italy
| | - Nicola Giangregorio
- National Research Council (CNR), Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126, Bari, Italy
| | - Cesare Indiveri
- National Research Council (CNR), Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126, Bari, Italy
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4 C, 87036, Arcavacata di Rende, Italy
| | - Tiziana Marino
- Department of Chemistry and Chemical Technologies, University of Calabria, Via Pietro Bucci 14/C, 87036, Arcavacata di Rende, CS, Italy
| |
Collapse
|
6
|
Nieto-Giraldo DF, Rodas Rodríguez JM, Torres-Osorio JI. Analysis of the impact of protein conformational dynamics and intermolecular interactions on water flux through TIP3;1 aquaporins of Zea mays L. Phys Chem Chem Phys 2025; 27:7797-7804. [PMID: 40152806 DOI: 10.1039/d4cp04661g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The discovery of aquaporins (AQPs) in 1992 had a profound impact on our understanding of the mechanisms underlying the transport of substances across cell membranes. To further understand water mobilization through AQPs, this study focuses on the characterization of water flux through the TIP3;1 aquaporins of Zea mays L. using molecular dynamics. The primary objective is to elucidate how protein-water intermolecular interactions and protein conformational dynamics impact water mobility across the cell membrane. To conduct this analysis, the three-dimensional structure of TIP3;1 was modeled using AlphaFold2, from which the complete system was constructed. This system consisted of a homotetramer of TIP3;1 immersed in a fragment of cell membrane and solvated with water molecules and ions. Subsequently, molecular dynamics simulations were conducted for 90 ns, resulting in the determination of an osmotic permeability coefficient (pf) of 0.8172 ± 0.146 × 10-14 cm3 s-1. In general, the mobility of water along the single-file water channel is influenced by the complex interplay of protein conformational dynamics and hydrogen bonding. The conformational dynamics of the protein channel modify the pore radius available for the passage of water, which affects the frequency of protein-water interactions and consequently influences the mobility of water in the channel. This study contributes to our understanding of the molecular mechanisms by which AQP activity is modulated without involving changes in protein chemical composition.
Collapse
Affiliation(s)
| | | | - Javier Ignacio Torres-Osorio
- Grupo de investigación en Magnetobiología, Department of Physics, Universidad de Caldas, Calle 65 # 26-10, Manizales, Colombia
| |
Collapse
|
7
|
Radziunas-Salinas Y, Domínguez-Arca V, Pardo A, Cambón A, Taboada P, Prieto G. Long-chain cationic gemini surfactants as drug retention adjuvant on liposomes. A methodological approach with atorvastatin. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184419. [PMID: 40187473 DOI: 10.1016/j.bbamem.2025.184419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
This study delves into the development and characterization of dipalmitoyl phosphatidylcholine (DPPC) liposomes incorporated with gemini surfactant (tetradecamethylene-1,14 bis(dimethyl tetradecyl ammonium bromide); 14-14-14) and atorvastatin, aimed at enhancing drug delivery efficiency for cardiovascular diseases. The integration of gemini surfactants into liposomes is investigated for its potential to improve atorvastatin encapsulation and retention, addressing the drug's poor water solubility and the limitations of conventional liposomal systems. Through a combination of dynamic light scattering (DLS), differential scanning calorimetry (DSC), and molecular dynamics (MD) simulations, the study reveals that the presence of gemini surfactants significantly reduces liposome size and polydispersity, indicative of a more uniform and potentially unilamellar structure. DSC analysis highlights a decrease in transition temperatures and an alteration in transition symmetry, suggesting enhanced stability and a favourable drug release profile at physiological temperatures. MD simulations provide insight into the internalization mechanism of gemini surfactants and atorvastatin within the liposomal bilayer, demonstrating their mutual incorporation facilitated by polar interactions. Spectrophotometry-based retention studies further confirmed that liposomes containing gemini surfactants exhibit superior atorvastatin retention capabilities, nearly doubling the encapsulation efficiency compared to conventional liposomes. This research highlights the promising role of gemini surfactant-incorporated liposomes as an efficient drug delivery platform for cardiovascular therapeutics, offering insights into the molecular interactions and structural dynamics underlying their enhanced performance.
Collapse
Affiliation(s)
- Yago Radziunas-Salinas
- Applied Physics Department, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Vicente Domínguez-Arca
- Applied Physics Department, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Biosystems and Bioprocess Engineering (Bio2Eng) Group, Institute of Marine Research of Spanish Research Council, IIM-CSIC, 36208 Vigo, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Alberto Pardo
- Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Adriana Cambón
- Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Pablo Taboada
- Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Gerardo Prieto
- Applied Physics Department, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
8
|
Skog AE, Jones NC, Månsson LK, Morth JP, Vrønning Hoffmann S, Gerelli Y, Skepö M. Assessing the interaction between the N-terminal region of the membrane protein magnesium transporter A and a lipid bilayer. J Colloid Interface Sci 2025; 683:663-674. [PMID: 39706085 DOI: 10.1016/j.jcis.2024.12.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024]
Abstract
This study investigates the interaction of KEIF, the intrinsically disordered N-terminal region of the magnesium transporter MgtA, with lipid bilayers mimicking cell membranes. Combining experimental techniques such as neutron reflectometry (NR), quartz-crystal microbalance with dissipation monitoring (QCM-D), synchrotron radiation circular dichroism (SRCD), and oriented circular dichroism (OCD), with molecular dynamics (MD) simulations, we characterized KEIF's adsorption behavior. HYPOTHESIS KEIF undergoes conformational changes upon interacting with lipid bilayers, potentially influencing MgtA's function within the plasma membrane. EXPERIMENTS The study assessed KEIF's structural transitions and position within lipid bilayers under various conditions, including zwitterionic versus anionic bilayers and different salt concentrations. The techniques analyzed adsorption-induced structural shifts and peptide localization within the bilayer. FINDINGS KEIF transitions from a disordered to a more structured state, notably increasing α-helical content upon adsorption to lipid bilayers. The peptide resides primarily in the hydrophobic tail region of the bilayer, where it may displace lipids. Electrostatic interactions, modulated by bilayer charge and ionic strength, play a critical role. These results suggest that KEIF's conformational changes and bilayer interactions can be integral to its potential modulatory role in MgtA function within the plasma membrane. This research highlights the importance of surface-induced structural transitions in intrinsically disordered proteins and their implications for membrane protein modulation.
Collapse
Affiliation(s)
- Amanda Eriksson Skog
- Division of Computational Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund, 22100, Sweden
| | - Nykola C Jones
- ISA, Department of Physics & Astronomy, Aarhus University, Aarhus C, 8000, Denmark
| | - Linda K Månsson
- Division of Computational Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund, 22100, Sweden
| | - Jens Preben Morth
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs. Lyngby, 2800, Denmark
| | | | - Yuri Gerelli
- Institute for Complex Systems, Consiglio Nazionale delle Ricerche, Piazzale Aldo Moro 5, Roma, 00185, Italy; Department of Physics, Sapienza University of Rome,Piazzale Aldo Moro 2, Roma, 00185, Italy
| | - Marie Skepö
- Division of Computational Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund, 22100, Sweden.
| |
Collapse
|
9
|
Cheng KJ, Shastry S, Campolargo JD, Hallock MJ, Pogorelov TV. Charge, Hydrophobicity, and Lipid Type Drive Antimicrobial Peptides' Unique Perturbation Ensembles. Biochemistry 2025; 64:1484-1500. [PMID: 40105792 DOI: 10.1021/acs.biochem.4c00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Antimicrobial peptides (AMPs) have emerged as a promising solution to the escalating public health threat caused by multidrug-resistant bacteria. Although ongoing research efforts have established AMP's role in membrane permeabilization and leakage, the precise mechanisms driving these disruption patterns remain unclear. We leverage molecular dynamics (MD) simulations enhanced by membrane mimetic (HMMM) to systematically investigate how the physiochemical properties of magainin (+3) and pexiganan (+9) affect their localization, insertion, curvature perturbation, and membrane binding ensemble. Building on existing microbiology, NMR, circular dichroism, and fluorescence data, our analysis reveals that the lipid makeup is a key determinant in the binding dynamics and structural conformation of AMPs. We find that phospholipid type is crucial for peptide localization, demonstrated through magainin's predominant interaction with lipid tails and pexiganan's with polar headgroups in POPC/POPS membranes. The membrane curvature changes induced by pexiganan relative to magainin suggest that AMPs with larger charges have more potential in modulating bilayer bending. These insights advance our understanding of AMP-membrane interactions at the molecular level, offering guidance for the design of targeted antimicrobial therapies.
Collapse
Affiliation(s)
- Kevin J Cheng
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Shashank Shastry
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Juan David Campolargo
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Michael J Hallock
- School of Chemical Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Taras V Pogorelov
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- School of Chemical Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- National Center for Supercomputer Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
10
|
Rosa RSL, Leal da Silva M, Bernardi RC. Atomistic Insights into gp82 Binding: A Microsecond, Million-Atom Exploration of Trypanosoma cruzi Host-Cell Invasion. Biochemistry 2025. [PMID: 40152296 DOI: 10.1021/acs.biochem.4c00710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, affects millions globally, leading to severe cardiac and gastrointestinal complications in its chronic phase. The invasion of host cells by T. cruzi is mediated by the interaction between the parasite's glycoprotein gp82 and the human receptor lysosome-associated membrane protein 2 (LAMP2). While experimental studies have identified a few residues involved in this interaction, a comprehensive molecular-level understanding has been lacking. In this study, we present a 1.44-million-atom computational model of the gp82 complex, including over 3300 lipids, glycosylation sites, and full molecular representations of gp82 and LAMP2, making it the most complete model of a parasite-host interaction to date. Using microsecond-long molecular dynamics simulations and dynamic network analysis, we identified critical residue interactions, including novel regions of contact that were previously uncharacterized. Our findings also highlight the significance of the transmembrane domain of LAMP2 in stabilizing the complex. These insights extend beyond traditional hydrogen bond interactions, revealing a complex network of cooperative motions that facilitate T. cruzi invasion. This study not only confirms key experimental observations but also uncovers new molecular targets for therapeutic intervention, offering a potential pathway to disrupt T. cruzi infection and combat Chagas disease.
Collapse
Affiliation(s)
- Raissa S L Rosa
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, United States
- Programa de Pós-Graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ 21040-360, Brazil
| | - Manuela Leal da Silva
- Programa de Pós-Graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ 21040-360, Brazil
- Instituto de Biodiversidade e Sustentabilidade (NUPEM), Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil
| | - Rafael C Bernardi
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
11
|
Doktorova M, Symons JL, Zhang X, Wang HY, Schlegel J, Lorent JH, Heberle FA, Sezgin E, Lyman E, Levental KR, Levental I. Cell membranes sustain phospholipid imbalance via cholesterol asymmetry. Cell 2025:S0092-8674(25)00270-3. [PMID: 40179882 DOI: 10.1016/j.cell.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/05/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025]
Abstract
Membranes are molecular interfaces that compartmentalize cells to control the flow of nutrients and information. These functions are facilitated by diverse collections of lipids, nearly all of which are distributed asymmetrically between the two bilayer leaflets. Most models of biomembrane structure and function include the implicit assumption that these leaflets have similar abundances of phospholipids. Here, we show that this assumption is generally invalid and investigate the consequences of lipid abundance imbalances in mammalian plasma membranes (PMs). Using lipidomics, we report that cytoplasmic leaflets of human erythrocyte membranes have >50% overabundance of phospholipids compared with exoplasmic leaflets. This imbalance is enabled by an asymmetric interleaflet distribution of cholesterol, which regulates cellular cholesterol homeostasis. These features produce unique functional characteristics, including low PM permeability and resting tension in the cytoplasmic leaflet that regulates protein localization.
Collapse
Affiliation(s)
- Milka Doktorova
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA; Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 17165 Solna, Sweden.
| | - Jessica L Symons
- Department of Integrative Biology & Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoxuan Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Jan Schlegel
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| | - Joseph H Lorent
- Department of Cellular and Molecular Pharmacology, TFAR, LDRI, UCLouvain, Avenue Mounier 73, B1.73.05, 1200 Brussels, Belgium
| | - Frederick A Heberle
- Department of Chemistry, University of Tennessee Knoxville, Knoxville, TN 37916, USA
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| | - Edward Lyman
- Department of Physics and Astronomy, Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA.
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
12
|
Ransey E, Thomas GE, Wisdom E, Almoril-Porras A, Bowman R, Adamson E, Walder-Christensen KK, White JA, Hughes DN, Schwennesen H, Ferguson C, Tye KM, Mague SD, Niu L, Wang ZW, Colón-Ramos D, Hultman R, Bursac N, Dzirasa K. Long-term editing of brain circuits in mice using an engineered electrical synapse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645291. [PMID: 40196531 PMCID: PMC11974911 DOI: 10.1101/2025.03.25.645291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Electrical signaling across distinct populations of brain cells underpins cognitive and emotional function; however, approaches that selectively regulate electrical signaling between two cellular components of a mammalian neural circuit remain sparse. Here, we engineered an electrical synapse composed of two connexin proteins found in Morone americana (white perch fish) - connexin34.7 and connexin35 - to accomplish mammalian circuit modulation. By exploiting protein mutagenesis, devising a new in vitro system for assaying connexin hemichannel docking, and performing computational modeling of hemichannel interactions, we uncovered a structural motif that contributes to electrical synapse formation. Targeting these motifs, we designed connexin34.7 and connexin35 hemichannels that dock with each other to form an electrical synapse, but not with other major connexins expressed in the mammalian central nervous system. We validated this electrical synapse in vivo using C. elegans and mice, demonstrating that it can strengthen communication across neural circuits composed of pairs of distinct cell types and modify behavior accordingly. Thus, we establish 'Long-term integration of Circuits using connexins' (LinCx) for precision circuit-editing in mammals.
Collapse
Affiliation(s)
- Elizabeth Ransey
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Gwenaëlle E. Thomas
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Elias Wisdom
- Department of Neuroscience and Department of Cell Biology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Agustin Almoril-Porras
- Department of Neuroscience and Department of Cell Biology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Ryan Bowman
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Elise Adamson
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Biomedical Engineering, Duke University, Durham North Carolina 27708, USA
| | - Kathryn K. Walder-Christensen
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Jesse A. White
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Dalton N. Hughes
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Hannah Schwennesen
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Caly Ferguson
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Kay M. Tye
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Stephen D. Mague
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Longgang Niu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Daniel Colón-Ramos
- Department of Neuroscience and Department of Cell Biology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico, San Juan, Puerto Rico
| | - Rainbo Hultman
- Department of Molecular Physiology and Biophysics, Department of Psychiatry, University of Iowa, Iowa City, IA, 52242 USA
| | - Nenad Bursac
- Dept. of Biomedical Engineering, Duke University, Durham North Carolina 27708, USA
| | - Kafui Dzirasa
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
- Dept. of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, USA
- Dept. of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Dept. of Neurosurgery, Duke University Medical Center, Durham, North Carolina 27710, USA
- Dept. of Biomedical Engineering, Duke University, Durham North Carolina 27708, USA
| |
Collapse
|
13
|
Braza MKE, Dennis EA, Amaro RE. Conformational dynamics and activation of membrane-associated human Group IVA cytosolic phospholipase A 2 (cPLA 2 ). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.22.644760. [PMID: 40196679 PMCID: PMC11974688 DOI: 10.1101/2025.03.22.644760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cytosolic phospholipase A 2 (cPLA 2 ) associates with membranes where it hydrolyzes phospholipids containing arachidonic acid to initiate an inflammatory cascade. All-atom molecular dynamics simulations were employed to understand the activation process when cPLA 2 associates with the endoplasmic reticulum (ER) membrane of macrophages where it acts. We found that membrane association causes the lid region of cPLA 2 to undergo a closed-to-open state transition that is accompanied by the sideways movement of loop 495-540, allowing the exposure of a cluster of lysine residues (K488, K541, K543, and K544), which binds the allosteric activator PIP 2 in the membrane. The active site of the open form of cPLA 2 , containing the catalytic dyad residues S228 and D549, exhibited a three-fold larger cavity than the closed form of cPLA 2 in aqueous solution. These findings provide mechanistic insight as to how cPLA 2 ER membrane association promotes major transitions between conformational states critical to allosteric activation and enzymatic phospholipid hydrolysis.
Collapse
|
14
|
Tavanti F, Brancolini G, Perris R. Computational analysis of the structural-functional dynamics of a Co-receptor proteoglycan. Front Mol Biosci 2025; 12:1549177. [PMID: 40201239 PMCID: PMC11975855 DOI: 10.3389/fmolb.2025.1549177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Nerve-Glial Antigen 2/Chondroitin Sulphate Proteoglycan 4 (NG2/CSPG4) is the largest membrane-intercalated cell surface component of the human proteome known to date. NG2/CSPG4 is endowed with the capability of engaging a myriad of molecular interactions and exert co-receptor functions, of which primary ones are sequestering of growth factors and the anchoring of cells to the extracellular matrix. However, the nature of the interactive dynamics of the proteoglycan remains veiled because of its conspicuous size and structural complexity. By leveraging on a multi-scale in silico approach, we have pioneered a comprehensive computational analysis of the structural-functional traits of the NG2/CSPG4 ectodomain. The modelling highlights an intricate assembly of β-sheet motifs linked together by flexible loops. Furthermore, our in silico predictions highlight that the previously delineated D1 domain may consistently remain more accessible for molecular interplays with respect to the D2 and D3 domains. Based on these findings, we have simulated the structural mechanism through the proteoglycan may serve as a co-receptor for growth factor FGF-2, showing that NG2/CSPG4 bends towards the receptor FGFR-1 for this growth factor and confirming the previously hypothesized trimeric complex formation promoted by FGF-2 dimers bridging the FGFR-1-proteoglycan interaction. The Chondroitin Sulphate Proteoglycan 4 is a large multi-domain transmembrane protein involved in several biological processes including pathological conditions. Despite its importance, it has never been studied at the atomistic level due to its large size. Here, we employed a multi-scale computer simulations approach to study its three-dimensional structure, its movements and co-receptor properties, showing that it can serve as mediator in the growth factor signaling process.
Collapse
Affiliation(s)
- Francesco Tavanti
- COMT – Centre for Molecular and Translational Oncology, University of Parma, Parma, Italy
- Department of Chemical and Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Center S3, CNR Institute of Nanoscience – CNR-NANO, Modena, Italy
| | | | - Roberto Perris
- COMT – Centre for Molecular and Translational Oncology, University of Parma, Parma, Italy
- Department of Chemical and Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| |
Collapse
|
15
|
Majewska M, Hanić M, Bartölke R, Schmidt J, Bożek J, Gerhards L, Mouritsen H, Koch KW, Solov’yov IA, Brand I. European Robin Cryptochrome-4a Associates with Lipid Bilayers in an Ordered Manner, Fulfilling a Molecular-Level Condition for Magnetoreception. ACS Chem Biol 2025; 20:592-606. [PMID: 39982451 PMCID: PMC11934094 DOI: 10.1021/acschembio.4c00576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/22/2025]
Abstract
Since the middle of the 20th century, long-distance avian migration has been known to rely partly on geomagnetic field. However, the underlying sensory mechanism is still not fully understood. Cryptochrome-4a (ErCry4a), found in European robin (Erithacus rubecula), a night-migratory songbird, has been suggested to be a magnetic sensory molecule. It is sensitive to external magnetic fields via the so-called radical-pair mechanism. ErCry4a is primarily located in the outer segments of the double-cone photoreceptor cells in the eye, which contain stacked and highly ordered membranes that could facilitate the anisotropic attachment of ErCry4a needed for magnetic compass sensing. Here, we investigate possible interactions of ErCry4a with a model membrane that mimics the lipid composition of outer segments of vertebrate photoreceptor cells using experimental and computational approaches. Experimental results show that the attachment of ErCry4a to the membrane could be controlled by the physical state of lipid molecules (average area per lipid) in the outer leaflet of the lipid bilayer. Furthermore, polarization modulation infrared reflection absorption spectroscopy allowed us to determine the conformation, motional freedom, and average orientation of the α-helices in ErCry4a in a membrane-associated state. Atomistic molecular dynamics studies supported the experimental results. A ∼ 1000 kcal mol-1 decrease in the interaction energy as a result of ErCry4a membrane binding was determined compared to cases where no protein binding to the membrane occurred. At the molecular level, the binding seems to involve negatively charged carboxylate groups of the phosphoserine lipids and the C-terminal residues of ErCry4a. Our study reveals a potential direct interaction of ErCry4a with the lipid membrane and discusses how this binding could be an essential step for ErCry4a to propagate a magnetic signal further and thus fulfill a role as a magnetoreceptor.
Collapse
Affiliation(s)
- Marta Majewska
- Institute
of Chemistry, School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg 26111, Germany
| | - Maja Hanić
- Institute
of Physics, School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg 26111, Germany
| | - Rabea Bartölke
- Animal
Navigation, Institute of Biology and Environmental Sciences, School
of Mathematics and Science, Carl von Ossietzky
Universität Oldenburg, Oldenburg D-26111, Germany
| | - Jessica Schmidt
- Animal
Navigation, Institute of Biology and Environmental Sciences, School
of Mathematics and Science, Carl von Ossietzky
Universität Oldenburg, Oldenburg D-26111, Germany
| | - Justyna Bożek
- Institute
of Chemistry, School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg 26111, Germany
| | - Luca Gerhards
- Institute
of Physics, School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg 26111, Germany
| | - Henrik Mouritsen
- Animal
Navigation, Institute of Biology and Environmental Sciences, School
of Mathematics and Science, Carl von Ossietzky
Universität Oldenburg, Oldenburg D-26111, Germany
- Research
Center for Neurosensory Sciences, Carl von
Ossietzky Universität Oldenburg, Oldenburg D-26111, Germany
| | - Karl-Wilhelm Koch
- Research
Center for Neurosensory Sciences, Carl von
Ossietzky Universität Oldenburg, Oldenburg D-26111, Germany
- Division
of Biochemistry, Department of Neuroscience, Carl von Ossietzky Universität Oldenburg, Oldenburg D-26111, Germany
| | - Ilia A. Solov’yov
- Institute
of Physics, School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg 26111, Germany
- Research
Center for Neurosensory Sciences, Carl von
Ossietzky Universität Oldenburg, Oldenburg D-26111, Germany
- Institute
of Physics, Center for Nanoscale Dynamics (CENAD), Carl von Ossietzky Universität Oldenburg, Oldenburg 26129, Germany
| | - Izabella Brand
- Institute
of Chemistry, School of Mathematics and Science, Carl von Ossietzky Universität Oldenburg, Oldenburg 26111, Germany
- Research
Center for Neurosensory Sciences, Carl von
Ossietzky Universität Oldenburg, Oldenburg D-26111, Germany
| |
Collapse
|
16
|
Kozai D, Inoue M, Suzuki S, Kamegawa A, Nishikawa K, Suzuki H, Ekimoto T, Ikeguchi M, Fujiyoshi Y. Narrowed pore conformations of aquaglyceroporins AQP3 and GlpF. Nat Commun 2025; 16:2653. [PMID: 40113770 PMCID: PMC11926279 DOI: 10.1038/s41467-025-57728-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
Aquaglyceroporins such as aquaporin-3 (AQP3) and its bacterial homologue GlpF facilitate water and glycerol permeation across lipid bilayers. X-ray crystal structures of GlpF showed open pore conformations, and AQP3 has also been predicted to adopt this conformation. Here we present cryo-electron microscopy structures of rat AQP3 and GlpF in different narrowed pore conformations. In n-dodecyl-β-D-maltopyranoside detergent micelles, aromatic/arginine constriction filter residues of AQP3 containing Tyr212 form a 2.8-Å diameter pore, whereas in 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) nanodiscs, Tyr212 inserts into the pore. Molecular dynamics simulation shows the Tyr212-in conformation is stable and largely suppresses water permeability. AQP3 reconstituted in POPC liposomes exhibits water and glycerol permeability, suggesting that the Tyr212-in conformation may be altered during permeation. AQP3 Y212F and Y212T mutant structures suggest that the aromatic residue drives the pore-inserted conformation. The aromatic residue is conserved in AQP7 and GlpF, but neither structure exhibits the AQP3-like conformation in POPC nanodiscs. Unexpectedly, the GlpF pore is covered by an intracellular loop, but the loop is flexible and not primarily related to the GlpF permeability. Our findings illuminate the unique AQP3 conformation and structural diversity of aquaglyceroporins.
Collapse
Affiliation(s)
- Daisuke Kozai
- Cellular and Structural Physiology Laboratory (CeSPL), Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| | - Masao Inoue
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Shota Suzuki
- Cellular and Structural Physiology Laboratory (CeSPL), Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| | - Akiko Kamegawa
- Cellular and Structural Physiology Laboratory (CeSPL), Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan
- Joint Research Course for Advanced Biomolecular Characterization, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kouki Nishikawa
- Joint Research Course for Advanced Biomolecular Characterization, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hiroshi Suzuki
- Cellular and Structural Physiology Laboratory (CeSPL), Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| | - Toru Ekimoto
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Mitsunori Ikeguchi
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- HPC- and AI-driven Drug Development Platform Division, Center for Computational Science, RIKEN, Yokohama, Japan
| | - Yoshinori Fujiyoshi
- Cellular and Structural Physiology Laboratory (CeSPL), Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan.
- Joint Research Course for Advanced Biomolecular Characterization, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan.
| |
Collapse
|
17
|
Yarava J, Orwick-Rydmark M, Ryoo D, Hofstetter A, Gumbart JC, Habeck M, van Rossum BJ, Linke D, Oschkinat H. Probing the Dynamics of Yersinia Adhesin A (YadA) in Outer Membranes Hints at Requirements for β-Barrel Membrane Insertion. J Am Chem Soc 2025; 147:8618-8628. [PMID: 40014811 PMCID: PMC11912334 DOI: 10.1021/jacs.4c17726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
The vast majority of cells are protected and functionalized by a dense surface layer of glycans, proteoglycans, and glycolipids. This surface represents an underexplored space in structural biology that is exceedingly challenging to recreate in vitro. Here, we investigate β-barrel protein dynamics within an asymmetric outer membrane environment, with the trimeric autotransporter Yersinia adhesin A (YadA) as an example. Magic-angle spinning NMR relaxation data and a model-free approach reveal increased mobility in the second half of strand β2 after the conserved G72, which is responsible for membrane insertion and autotransport, and in the subsequent loop toward β3. In contrast, the protomer-protomer interaction sites (β1i-β4i-1) are rigid. Intriguingly, the mobility in the β-strand section following G72 is substantially elevated in the outer membrane and less so in the detergent environment of microcrystals. A possible source is revealed by molecular dynamics simulations that show the formation of a salt bridge involving E79 and R76 in competition with a dynamic interplay of calcium binding by E79 and the phosphate groups of the lipids. An estimation of overall barrel motion in the outer membrane and detergent-containing crystals yields values of around 41 ns for both. The global motion of YadA in the outer membrane has a stronger rotational component orthogonal to the symmetry axis of the trimeric porin than in the detergent-containing crystal. In summary, our investigation shows that the mobility in the second half of β2 and the loop to β3 required for membrane insertion and autotransport is maintained in the final folded form of YadA.
Collapse
Affiliation(s)
- Jayasubba
Reddy Yarava
- Leibniz-Forschungsinstitut
für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | | | - David Ryoo
- Interdisciplinary
Bioengineering Graduate Program, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
| | - Albert Hofstetter
- Department
of Chemistry and Applied Biosciences, ETH
Zurich, Vladimir-Prelog-Weg
2, 8093 Zurich, Switzerland
| | - James C. Gumbart
- School of
Physics, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Michael Habeck
- Microscopic
Image Analysis Group, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Barth-Jan van Rossum
- Leibniz-Forschungsinstitut
für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Dirk Linke
- Department
of Biosciences, University of Oslo, P.O.Box 1066 Blindern, 0316 Oslo, Norway
| | - Hartmut Oschkinat
- Leibniz-Forschungsinstitut
für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Freie
Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| |
Collapse
|
18
|
Krah A, Grover V, Fong TC, Bond PJ, Grüber G. Water occupancy in the Acinetobacter baumannii F-ATP synthase c-ring and its implications as a novel inhibitor target. Structure 2025; 33:594-600.e3. [PMID: 39892380 DOI: 10.1016/j.str.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/07/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
The Acinetobacter baumannii F1FO-ATP synthase is essential for the opportunistic human pathogen. Its membrane-embedded FO domain consists of the c-ring and subunit a. The c-ring translocates protons via a conserved carboxylate across the membrane via two half-channels in subunit a, and its revolution enables the F1 domain to carry out ATP formation. Here, we used molecular dynamics simulations, free energy calculations, and in vivo mutational experiments to assess the likely existence of water molecules in the binding site of the A. baumannii c-ring. We first predicted its binding site structure in the ion-locked conformation and extrapolated the presence of two water molecules in the ion-binding site. Based on our predictions, amino acid point mutations confirmed the critical role of key residues involved in the water-binding site upon ATP synthesis ability and cell growth. We discuss the implications of our findings in the context of rational drug design to target the A. baumannii FO domain.
Collapse
Affiliation(s)
- Alexander Krah
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore.
| | - Vandana Grover
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Tuck Choy Fong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Peter J Bond
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore; Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore.
| | - Gerhard Grüber
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore; School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
19
|
Paloncýová M, Valério M, Dos Santos RN, Kührová P, Šrejber M, Čechová P, Dobchev DA, Balsubramani A, Banáš P, Agarwal V, Souza PCT, Otyepka M. Computational Methods for Modeling Lipid-Mediated Active Pharmaceutical Ingredient Delivery. Mol Pharm 2025; 22:1110-1141. [PMID: 39879096 PMCID: PMC11881150 DOI: 10.1021/acs.molpharmaceut.4c00744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025]
Abstract
Lipid-mediated delivery of active pharmaceutical ingredients (API) opened new possibilities in advanced therapies. By encapsulating an API into a lipid nanocarrier (LNC), one can safely deliver APIs not soluble in water, those with otherwise strong adverse effects, or very fragile ones such as nucleic acids. However, for the rational design of LNCs, a detailed understanding of the composition-structure-function relationships is missing. This review presents currently available computational methods for LNC investigation, screening, and design. The state-of-the-art physics-based approaches are described, with the focus on molecular dynamics simulations in all-atom and coarse-grained resolution. Their strengths and weaknesses are discussed, highlighting the aspects necessary for obtaining reliable results in the simulations. Furthermore, a machine learning, i.e., data-based learning, approach to the design of lipid-mediated API delivery is introduced. The data produced by the experimental and theoretical approaches provide valuable insights. Processing these data can help optimize the design of LNCs for better performance. In the final section of this Review, state-of-the-art of computer simulations of LNCs are reviewed, specifically addressing the compatibility of experimental and computational insights.
Collapse
Affiliation(s)
- Markéta Paloncýová
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Mariana Valério
- Laboratoire
de Biologie et Modélisation de la Cellule, CNRS, UMR 5239,
Inserm, U1293, Université Claude Bernard Lyon 1, Ecole Normale
Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon, France
- Centre Blaise
Pascal de Simulation et de Modélisation Numérique, Ecole Normale Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon, France
| | | | - Petra Kührová
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Martin Šrejber
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Petra Čechová
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | | | - Akshay Balsubramani
- mRNA Center
of Excellence, Sanofi, Waltham, Massachusetts 02451, United States
| | - Pavel Banáš
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
| | - Vikram Agarwal
- mRNA Center
of Excellence, Sanofi, Waltham, Massachusetts 02451, United States
| | - Paulo C. T. Souza
- Laboratoire
de Biologie et Modélisation de la Cellule, CNRS, UMR 5239,
Inserm, U1293, Université Claude Bernard Lyon 1, Ecole Normale
Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon, France
- Centre Blaise
Pascal de Simulation et de Modélisation Numérique, Ecole Normale Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon, France
| | - Michal Otyepka
- Regional
Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký
University Olomouc, Šlechtitelů 27, 779 00 Olomouc, Czech Republic
- IT4Innovations,
VŠB − Technical University of Ostrava, 17. listopadu 2172/15, 708 00 Ostrava-Poruba, Czech Republic
| |
Collapse
|
20
|
Nivón-Ramirez D, Galván-García EA, Ponce-Pérez LD, Gómez-Balderas R. Virtual screening of potential inhibitors of the ATPase site in Acinetobacter baumannii DNA Gyrase. Comput Biol Med 2025; 186:109728. [PMID: 39862471 DOI: 10.1016/j.compbiomed.2025.109728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Bacterial resistance is a global public health problem because of the ineffectiveness of conventional antibiotics against super pathogens. To counter this situation, the search for or design of new molecules is essential to inhibit the key proteins involved in several stages of bacterial infection. One of these key proteins is DNA gyrase, which is responsible for packaging and unfolding of DNA chains during replication. Virtual screening calculations of 583,900 molecules against the ATPase site of DNA gyrase (PDB ID 7PQM) resulted in three promising molecules (Z927783420, Z4422201766, and Z2440107042) with significant binding modes at the active site, according to molecular docking studies. Additionally, they exhibited lower toxicological profiles than the previously reported 80S inhibitors. Molecular dynamics calculations revealed crucial interactions responsible for the inhibition process, with residues ASP87, GLU94, and ASN60 belonging to the ATPase site. On the other hand, the binding energy calculated using the MM/GBSA protocol highlighted Z2440107042 as the most promising inhibitor, with the best binding energy (-74.77 kcal/mol), suggesting that this molecule is a strong candidate for further biological studies.
Collapse
Affiliation(s)
- Didier Nivón-Ramirez
- Laboratorio de Fisicoquímica Analítica, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, 54714, Mexico
| | - Emir A Galván-García
- Laboratorio de Fisicoquímica Analítica, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, 54714, Mexico
| | - León D Ponce-Pérez
- Laboratorio de Fisicoquímica Analítica, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, 54714, Mexico
| | - Rodolfo Gómez-Balderas
- Laboratorio de Fisicoquímica Analítica, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, 54714, Mexico.
| |
Collapse
|
21
|
Liu Z, Liu W, Shen X, Jiang T, Li X, Liu H, Zheng Z. Molecular mechanism of type ib MET inhibitors and their potential for CNS tumors. Sci Rep 2025; 15:6926. [PMID: 40011494 DOI: 10.1038/s41598-025-85631-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/06/2025] [Indexed: 02/28/2025] Open
Abstract
The emergence of targeted therapies for MET exon 14 (METex14) skipping mutations has significantly changed the treatment landscape for NSCLC and other solid tumors. The skipping of METex14 results in activating the MET-HGF pathway and promoting tumor cell proliferation, migration, and preventing apoptosis. Type Ib MET inhibitors, designed to selectively target the "DFG-in" conformation of MET, characteristically bind to the ATP-binding pocket of MET in a U-shaped conformation, extending into the solvent-accessible region and interact strongly with residue Y1230 through π-π interactions, have shown remarkable efficacy in treating METex14-altered NSCLC, including cases with brain metastases (BMs). Notably, vebreltinib and capmatinib have demonstrated superior blood-brain barrier (BBB) permeability in both computational and experimental models, highlighting their potential for treating the central nervous system (CNS) metastases. P-glycoprotein (P-gp) is highly expressed in the BBB, which limits the brain uptake of many highly lipophilic drugs. Despite challenges posed by P-gp mediated efflux, vebreltinib has emerged as a promising candidate for CNS treatment due to its favorable pharmacokinetic profile and minimal susceptibility to P-gp efflux. This study underscores the importance of molecular dynamics simulations in predicting drug efficacy and BBB penetration, providing valuable insights for the development of CNS-targeted metastases therapies.
Collapse
Affiliation(s)
- Zhenhao Liu
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Wenlang Liu
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Xinyi Shen
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Tao Jiang
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Xionghao Li
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Hao Liu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, Hubei, People's Republic of China.
| | - Zheng Zheng
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
22
|
Avstrikova M, Milán Rodríguez P, Burke SM, Hibbs RE, Changeux JP, Cecchini M. Hidden complexity of α7 nicotinic acetylcholine receptor desensitization revealed by MD simulations and Markov state modeling. Proc Natl Acad Sci U S A 2025; 122:e2420993122. [PMID: 39946538 PMCID: PMC11848294 DOI: 10.1073/pnas.2420993122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
The α7 nicotinic acetylcholine receptor is a pentameric ligand-gated ion channel that plays an important role in neuronal signaling throughout the nervous system. Its implication in neurological disorders and inflammation has spurred the development of numerous compounds that enhance channel activation. However, the therapeutic potential of these compounds has been limited by the characteristically fast desensitization of the α7 receptor. Using recent high-resolution structures from cryo-EM, and all-atom molecular dynamic simulations augmented by Markov state modeling, here we explore the mechanism of α7 receptor desensitization and its implication on allosteric modulation. The results provide a precise characterization of the desensitization gate and illuminate the mechanism of ion-pore opening/closing with an agonist bound. In addition, the simulations reveal the existence of a short-lived, open-channel intermediate between the activated and desensitized states that rationalizes the paradoxical pharmacology of the L247T mutant and may be relevant to type-II allosteric modulation. This analysis provides an interpretation of the signal transduction mechanism and its regulation in α7 receptors.
Collapse
Affiliation(s)
- Mariia Avstrikova
- Institut de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, Strasbourg CedexF-67083, France
| | - Paula Milán Rodríguez
- Institut de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, Strasbourg CedexF-67083, France
| | - Sean M. Burke
- Molecular Biophysics Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Neurobiology, University of California, San Diego, La Jolla, CA92093
| | - Ryan E. Hibbs
- Department of Neurobiology, University of California, San Diego, La Jolla, CA92093
| | - Jean-Pierre Changeux
- Neuroscience Department, Institut Pasteur, Collège de France, ParisF-75005, France
| | - Marco Cecchini
- Institut de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, Strasbourg CedexF-67083, France
| |
Collapse
|
23
|
Hedger G, Yen HY. The Influence of Phosphoinositide Lipids in the Molecular Biology of Membrane Proteins: Recent Insights from Simulations. J Mol Biol 2025; 437:168937. [PMID: 39793883 PMCID: PMC7617384 DOI: 10.1016/j.jmb.2025.168937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/29/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
The phosphoinositide family of membrane lipids play diverse and critical roles in eukaryotic molecular biology. Much of this biological activity derives from interactions of phosphoinositide lipids with integral and peripheral membrane proteins, leading to modulation of protein structure, function, and cellular distribution. Since the discovery of phosphoinositides in the 1940s, combined molecular biology, biophysical, and structural approaches have made enormous progress in untangling this vast and diverse cellular network of interactions. More recently, in silico approaches such as molecular dynamics simulations have proven to be an asset in prospectively identifying, characterising, explaining the structural basis of these interactions, and in the best cases providing atomic level testable hypotheses on how such interactions control the function of a given membrane protein. This review details a number of recent seminal discoveries in phosphoinositide biology, enabled by advanced biomolecular simulation, and its integration with molecular biology, biophysical, and structural biology approaches. The results of the simulation studies agree well with experimental work, and in a number of notable cases have arrived at the key conclusion several years in advance of the experimental structures. SUMMARY: Hedger and Yen review developments in simulations of phosphoinositides and membrane proteins.
Collapse
Affiliation(s)
- George Hedger
- Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, SW7 2AZ, UK.
| | - Hsin-Yung Yen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Department of Chemistry, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK
| |
Collapse
|
24
|
Suhag K, Borkotoky S, Siddiqui SI, Kumar J, Kumar CS, Tatiya P, Ghosh S, Banerjee M. Mechanistic Insights into the Divergent Membrane Activities of a Viroporin from Chikungunya Virus and Its Transframe Variant. ACS Infect Dis 2025; 11:430-441. [PMID: 39745175 DOI: 10.1021/acsinfecdis.4c00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Alphaviruses, a genus of vector-borne viruses in the Togaviridae family, encode a small ion-channel-forming protein, 6K, and its transframe variant (TF) during infections. Although 6K/TF have vital roles in glycoprotein transport, virus assembly, and budding, there is no mechanistic explanation for these functions. We investigated the distinct biochemical functionalities of 6K and TF from the mosquito-borne alphavirus, Chikungunya Virus. We show that like 6K, TF is also capable of forming ion channels in bilayer membranes. The assemblies formed by 6K in membranes are structurally more complex and potentially more ion-restrictive than those formed by TF. Both 6K and TF show strong affinity toward the ER membranes, indicating that the localization of these components at the plasma membrane, as previously reported, is either linked to post-translational modification or mediated through interaction with binding partners. These structural and functional insights may elucidate the distinct roles of 6K and TF in the alphavirus life cycle.
Collapse
Affiliation(s)
- Kirti Suhag
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Subhomoi Borkotoky
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Shumaila Iqbal Siddiqui
- Department of Biophysics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Jitender Kumar
- Department of Physics and Astrophysics, Delhi University, New Delhi 110007, India
| | - Chandra Shekhar Kumar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Pushkar Tatiya
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Subhendu Ghosh
- Department of Biophysics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Manidipa Banerjee
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
25
|
Catalina-Hernandez E, Aguilella-Arzo M, Peralvarez-Marin A, Lopez-Martin M. Computational Insights into Membrane Disruption by Cell-Penetrating Peptides. J Chem Inf Model 2025; 65:1549-1559. [PMID: 39823544 DOI: 10.1021/acs.jcim.4c01940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Cell-penetrating peptides (CPPs) can translocate into cells without inducing cytotoxicity. The internalization process implies several steps at different time scales ranging from microseconds to minutes. We combine adaptive Steered Molecular Dynamics (aSMD) with conventional Molecular Dynamics (cMD) to observe nonequilibrium and equilibrium states to study the early mechanisms of peptide-bilayer interaction leading to CPPs internalization. We define three membrane compositions representing bilayer sections, neutral lipids (i.e., upper leaflet), neutral lipids with cholesterol (i.e., hydrophobic core), and neutral/negatively charged lipids with cholesterol (i.e., lower leaflet) to study the energy barriers and disruption mechanisms of Arg9, MAP, and TP2, representing cationic, amphiphilic, and hydrophobic CPPs, respectively. Cholesterol and negatively charged lipids increase the energetic barriers for the peptide-bilayer crossing. TP2 interacts with the bilayer by hydrophobic insertion, while Arg9 disrupts the bilayer by forming transient or stable pores. MAP has shown both behaviors. Collectively, these findings underscore the significance of innovative computational approaches in studying membrane-disruptive peptides and, more specifically, in harnessing their potential for cell penetration.
Collapse
Affiliation(s)
- Eric Catalina-Hernandez
- Unit of Biophysics, Department of Biochemistry and Molecular Biology, Facultat de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
- Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| | - Marcel Aguilella-Arzo
- Laboratory of Molecular Biophysics, Department of Physics, University Jaume I, 12071 Castellon, Spain
| | - Alex Peralvarez-Marin
- Unit of Biophysics, Department of Biochemistry and Molecular Biology, Facultat de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
- Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| | - Mario Lopez-Martin
- Unit of Biophysics, Department of Biochemistry and Molecular Biology, Facultat de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
- Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| |
Collapse
|
26
|
Savenko M, Vácha R, Ramseyer C, Rivel T. Role of Divalent Ions in Membrane Models of Polymyxin-Sensitive and Resistant Gram-Negative Bacteria. J Chem Inf Model 2025; 65:1476-1491. [PMID: 39825802 DOI: 10.1021/acs.jcim.4c01574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
Polymyxins, critical last-resort antibiotics, impact the distribution of membrane-bound divalent cations in the outer membrane of Gram-negative bacteria. We employed atomistic molecular dynamics simulations to model the effect of displacing these ions. Two polymyxin-sensitive and two polymyxin-resistant models of the outer membrane of Salmonella enterica were investigated. First, we found that the removal of all calcium ions induces global stress on the model membranes, leading to substantial membrane restructuring. Next, we used enhanced sampling methods to explore the effects of localized stress by displacing membrane-bound ions. Our findings indicate that creating defects in the membrane-bound ion network facilitates polymyxin permeation. Additionally, our study of polymyxin-resistant mutations revealed that divalent ions in resistant model membranes are less likely to be displaced, potentially contributing to the increased resistance associated with these mutations. Lastly, we compared results from all-atom molecular dynamics simulations with coarse-grained simulations, demonstrating that the choice of force field significantly influences the behavior of membrane-bound ions under stress.
Collapse
Affiliation(s)
- Mariia Savenko
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague16000, Czech Republic
- Laboratoire Chrono-Environnement UMR CNRS 6249, Université de Bourgogne Franche-Comté, Besançon25000, France
| | - Robert Vácha
- Central European Institute of Technology, Masaryk University, Brno60200, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno60200, Czech Republic
| | - Christophe Ramseyer
- Laboratoire Chrono-Environnement UMR CNRS 6249, Université de Bourgogne Franche-Comté, Besançon25000, France
| | - Timothée Rivel
- Central European Institute of Technology, Masaryk University, Brno60200, Czech Republic
| |
Collapse
|
27
|
Xu Z, Gan M, Guan W, Tian F, Wang Y, Zhang J, Cai L. In Slico Screening and In Vitro Identification of Hyperuricemia-Inhibiting Peptides from Trachurus japonicus. Foods 2025; 14:524. [PMID: 39942117 PMCID: PMC11817512 DOI: 10.3390/foods14030524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Hyperuricemia arises from imbalanced uric acid metabolism, contributing to gout and related chronic diseases. When traditional drugs are used to treat hyperuricemia, side effects are inevitable, which promotes the exploration of new bioactive compounds. Protein hydrolysates and peptides are gradually showing potential in the treatment of hyperuricemia. This study investigated the uric acid inhibitory activity of peptides extracted from Trachurus japonicus using in silico and in vitro methods. We employed in silico virtual enzymolysis and experimental validation to identify bioactive peptides from Trachurus japonicus proteins. Four peptides (DF, AGF, QPSF, and AGDDAPR) were comprehensively screened by molecular docking and database analysis. After solid-phase synthesis, the inhibitory effects of these peptides on hyperuricemia were further verified in vitro and at the cellular level. The results showed that all four peptides have good hyperuricemia-inhibiting activities. Molecular docking and molecular dynamics revealed that peptides DF and AGDDAPR affect the production of uric acid by binding to the active sites of urate transporter 1 (URAT1), glucose transporter 9 (GLUT9), and xanthine oxidase (XOD), while peptides QPSF and AGF mainly influence the XOD active site, confirming that it is feasible to rapidly screen hyperuricemia-inhibiting peptides by molecular docking.
Collapse
Affiliation(s)
- Zexuan Xu
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China;
| | - Miaoyu Gan
- Ningbo Innovation Center, College of Biosystems and Food Science, Zhejiang University, Ningbo 315100, China; (M.G.); (Y.W.)
| | - Weiliang Guan
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China;
| | - Fang Tian
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China;
| | - Yuxi Wang
- Ningbo Innovation Center, College of Biosystems and Food Science, Zhejiang University, Ningbo 315100, China; (M.G.); (Y.W.)
| | - Jinjie Zhang
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China;
| | - Luyun Cai
- Ningbo Innovation Center, College of Biosystems and Food Science, Zhejiang University, Ningbo 315100, China; (M.G.); (Y.W.)
| |
Collapse
|
28
|
Drabek M, Emmerich R, Djulic N, Bünemann M, Schlitzer M, Kolb P. Investigation of Muscarinic Acetylcholine Receptor M 3 Activation in Atomistic Detail: A Chemist's Viewpoint. ChemMedChem 2025; 20:e202400633. [PMID: 39462201 PMCID: PMC11793849 DOI: 10.1002/cmdc.202400633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/12/2024] [Accepted: 10/25/2024] [Indexed: 10/29/2024]
Abstract
We analyzed the precise ligand:receptor interactions required for activation of the muscarinic acetylcholine receptor M3, a prototypical G protein-coupled receptor and potential diabetes target. Starting from literature-known compounds and docking solutions, ligands were tailored for the modulation of this receptor's activation. Several aspects of the structure-activity relationship of agonists were investigated in atomistic detail, in order to delineate how the receptor can be activated via the orthosteric site. Such exquisitely precise knowledge is instrumental for designing potent and effiacious ligands. We put this strategy into practice and acquired or synthesized and measured a diverse set of 55 ligands ranging from small fragment-like amines coordinating D3.32 to bigger molecules extending towards helices 5 and 6 with diphenyl moieties. In the course of these investigations, we showed that the polarizability of the amine nitrogen and the rigidity and size of the moieties in the space delimited by helices 5 and 6 are the two key elements distinguishing potent and efficacious ligands from those that are not. The resulting data set will be highly useful in drug design and molecular machine learning alike.
Collapse
Affiliation(s)
- Matthäus Drabek
- Department of Pharmaceutical ChemistryUniversity of MarburgGermany
| | - Rolf Emmerich
- Department of Pharmaceutical ChemistryUniversity of MarburgGermany
| | - Nuri Djulic
- Department of Pharmaceutical ChemistryUniversity of MarburgGermany
| | - Moritz Bünemann
- Department of Pharmacology and Clinical PharmacyUniversity of MarburgGermany
| | - Martin Schlitzer
- Department of Pharmaceutical ChemistryUniversity of MarburgGermany
| | - Peter Kolb
- Department of Pharmaceutical ChemistryUniversity of MarburgGermany
| |
Collapse
|
29
|
Teskey G, Tiwari N, Butcko AJ, Kumar A, Yadav A, Huang YMM, Kelly CV, Granneman JG, Perfield JW, Mottillo EP. Lipid droplet targeting of the lipase coactivator ABHD5 and the fatty liver disease-causing variant PNPLA3 I148M is required to promote liver steatosis. J Biol Chem 2025; 301:108186. [PMID: 39814233 PMCID: PMC11849118 DOI: 10.1016/j.jbc.2025.108186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
The storage and release of triacylglycerol (TAG) in lipid droplets (LDs) is regulated by dynamic protein interactions. α/β Hydrolase domain-containing protein 5 (ABHD5; also known as CGI-58) is a membrane/LD-bound protein that functions as a co-activator of patatin-like phospholipase domain-containing 2 (PNPLA2; also known as adipose triglyceride lipase) the rate-limiting enzyme for TAG hydrolysis. The dysregulation of TAG hydrolysis is involved in various metabolic diseases such as metabolic dysfunction-associated steatotic liver disease (MASLD). We previously demonstrated that ABHD5 interacted with PNPLA3, a closely related family member to PNPLA2. Importantly, a common missense variant in PNPLA3 (I148M) is the greatest genetic risk factor for MASLD. PNPLA3 148M functions to sequester ABHD5 and prevent coactivation of PNPLA2, which has implications for initiating MASLD; however, the exact mechanisms involved are not understood. Here, we demonstrate that LD targeting of both ABHD5 and PNPLA3 I148M is required for the interaction. Molecular modeling demonstrates important residues in the C terminus of PNPLA3 for LD binding and fluorescence cross-correlation spectroscopy demonstrates that PNPLA3 I148M has greater association with ABHD5 than WT PNPLA3. Moreover, the C terminus of PNPLA3 is sufficient for functional targeting of PNPLAs to LD and the interaction with ABHD5. In addition, ABHD5 is a general binding partner of LD-bound PNPLAs. Finally, PNPLA3 I148M targeting to LD is required to promote steatosis in vitro and in the liver. Overall results suggest that the interaction of PNPLA3 I148M with ABHD5 on LD is required to promote liver steatosis.
Collapse
Affiliation(s)
- Grace Teskey
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - Nivedita Tiwari
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA
| | - Andrew J Butcko
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Amit Kumar
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, USA
| | - Anuradha Yadav
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, USA
| | - Yu-Ming M Huang
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, USA
| | - Christopher V Kelly
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, USA
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - James W Perfield
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis Indiana, USA
| | - Emilio P Mottillo
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA.
| |
Collapse
|
30
|
Haque A, Alenezi KM, Khan MWA, Soury R, Khan MS, Ahamad S, Ahmad S, Gupta D. In silico evaluation of 4-thiazolidinone-based inhibitors against the receptor for advanced glycation end products (RAGE). J Biomol Struct Dyn 2025; 43:985-996. [PMID: 38063048 DOI: 10.1080/07391102.2023.2290621] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/10/2023] [Indexed: 01/01/2025]
Abstract
Non-enzymatic glycation of biomolecules by reducing sugars led to several products, including the advanced glycation end products (AGEs), the accumulation of which has been linked to various life-threatening diseases. The binding of AGEs to their respective protein receptors for advanced glycation end products (RAGE) can initiate a cascade of reactions, which may alter physiological conditions. The present work investigates the potential of 4-thiazolidinones as RAGE inhibitors. We performed an extensive computational study to identify the structural requirements needed to act as RAGE inhibitors. To achieve this goal, 4-thiazolidinone-based compounds available in PubChem, ZINC15, ChEMBL, and ChEBI databases were screened against RAGE (PDB: 4LP5), leading to the identification of top five drug-like candidates with a high binding affinity to RAGE V-domain catalytic region. Drug likeness, absorption, distribution, metabolism, excretion, and toxicity (ADMET) of the top-scoring compounds have been studied and discussed. Global molecular descriptors, chemical reactivity, hardness, softness, etc., have been estimated. Finally, molecular dynamics (MD) simulations at 100 ns were carried out to check the stability and other properties. Overall, we believe that the identified compounds can potentially attenuate RAGE-AGE interactions.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ashanul Haque
- Department of Chemistry, College of Science, University of Ha'il, Ha'il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha'il, Hail, Saudi Arabia
| | - Khalaf M Alenezi
- Department of Chemistry, College of Science, University of Ha'il, Ha'il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha'il, Hail, Saudi Arabia
| | - Mohd Wajid A Khan
- Department of Chemistry, College of Science, University of Ha'il, Ha'il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha'il, Hail, Saudi Arabia
| | - Raoudha Soury
- Department of Chemistry, College of Science, University of Ha'il, Ha'il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha'il, Hail, Saudi Arabia
| | - Muhammad S Khan
- Department of Chemistry, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Shahzaib Ahamad
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Shahnawaz Ahmad
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| |
Collapse
|
31
|
Ho TH, Tran KG, Huynh LK, Nguyen TT. Fluoxetine Alters the Biophysics of DPPC and DPPG Bilayers through Phase-Dependent and Electrostatic Interactions. J Phys Chem B 2025; 129:1248-1259. [PMID: 39681524 DOI: 10.1021/acs.jpcb.4c04631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Lipid membranes can control the permeability of a pharmaceutical drug, whereas the drug can induce changes in the structural and biophysical properties of the membranes. Understanding this interplay of drug-lipid membrane interactions can be of great importance in drug design. Here, we present a molecular dynamics study to provide insights into the interactions between the antidepressant fluoxetine and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) or 1,2-dipalmitoyl-sn-glycero-3-phosphoglycerol (DPPG) bilayers. It was found that, due to the electrostatic interaction, the headgroup of the zwitterionic DPPC lipid is more stable than that of the negatively charged DPPG lipid, allowing the gel phase to persist even at the elevated temperature. At 25 °C, fluoxetine cannot penetrate into the gel-phase DPPC bilayer, while the electrostatic interaction between positively charged fluoxetine and negatively charged DPPG bilayer retains the drug within the lipid headgroup domain. When the temperature is increased to 45 °C, both neutral and charged forms of fluoxetine can partition into the DPPC and DPPG bilayers spontaneously. Analysis of the biophysical and structural changes in both DPPC and DPPG bilayers in the presence of fluoxetine revealed a phase-dependent effect. The binding of fluoxetine to the lipid bilayers limits the movement and orientation of the drug. These findings shed light on the interactions between a commonly prescribed antidepressant and lipid membranes, and such information can be beneficial to the development of potential therapeutic agents.
Collapse
Affiliation(s)
- Tho H Ho
- Vietnam National University, Ho Chi Minh City, Vietnam 700000
- School of Chemical and Environmental Engineering, International University, Quarter 6, Linh Trung Ward, Thu Duc City, Ho Chi Minh City, Vietnam 700000
| | - Khai G Tran
- Vietnam National University, Ho Chi Minh City, Vietnam 700000
- School of Chemical and Environmental Engineering, International University, Quarter 6, Linh Trung Ward, Thu Duc City, Ho Chi Minh City, Vietnam 700000
| | - Lam K Huynh
- Vietnam National University, Ho Chi Minh City, Vietnam 700000
- School of Chemical and Environmental Engineering, International University, Quarter 6, Linh Trung Ward, Thu Duc City, Ho Chi Minh City, Vietnam 700000
| | - Trang T Nguyen
- Vietnam National University, Ho Chi Minh City, Vietnam 700000
- School of Chemical and Environmental Engineering, International University, Quarter 6, Linh Trung Ward, Thu Duc City, Ho Chi Minh City, Vietnam 700000
| |
Collapse
|
32
|
Park SJ, Schnitzer KA, Kovalenko A, Cherepanov S, Patro LPP, Song Z, Pogozheva ID, Lomize AL, Im W. OPRLM: A Web Tool and a Database for Positioning and Simulations of Proteins in Realistic Lipid Membranes. J Mol Biol 2025:168966. [PMID: 40133776 DOI: 10.1016/j.jmb.2025.168966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 01/23/2025] [Indexed: 03/27/2025]
Abstract
Molecular dynamics (MD) simulations in explicit lipid bilayers enable modeling of protein-lipid interactions essential for membrane protein functions and regulation. The newly developed computational web tool, OPRLM (Orientations of Proteins in Realistic Lipid Membranes), automates the assembly of membrane protein structures with explicit lipids corresponding to 18 biological membrane types with symmetric or asymmetric lipid distributions, as well as 5 types of two-component lipid bilayers with varying cholesterol content. Built upon the CHARMM-GUI toolset and the PPM method, OPRLM simplifies the setup of complex simulation system involving integral and/or peripheral membrane proteins with explicit lipid mixtures and generates all necessary files for subsequent all-atom MD simulations. OPRLM has successfully generated protein-membrane systems for 286 tested protein structures in various biomembranes, including 138 structures containing ligands. The OPRLM database, an advanced successor of the OPM database, includes explicit protein-lipid systems for tested proteins in their native biomembranes. It provides coordinates of integral and peripheral membrane proteins from the Protein Data Bank embedded in planar or curved implicit lipid bilayers. Additionally, it includes the classification of proteins into types, superfamilies, and families, along with the information on intracellular localizations and membrane topology and visualization options. The OPRLM web tool and the database are publicly accessible at https://oprlm.org.
Collapse
Affiliation(s)
- Sang-Jun Park
- Department of Computer Science and Engineering, Lehigh University, 111 Research Drive, Bethlehem, PA 18015, United States
| | - Kyle A Schnitzer
- Department of Electrical Engineering and Computer Science, College of Engineering, University of Michigan, 1221 Beal Ave, Ann Arbor, MI 48109-2102, United States
| | - Alexey Kovalenko
- Department of Computer Science, College of Literature, Science, and the Arts, University of Michigan, 2260 Hayward Street, Ann Arbor, MI 48109-2121, United States
| | - Stanislav Cherepanov
- Biophysics Program, University of Michigan, 930 N. University Avenue, Ann Arbor, MI 48109-1065, United States
| | - L Ponoop Prasad Patro
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem, PA 18015, United States
| | - Zigang Song
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem, PA 18015, United States
| | - Irina D Pogozheva
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109-1065, United States
| | - Andrei L Lomize
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109-1065, United States.
| | - Wonpil Im
- Department of Computer Science and Engineering, Lehigh University, 111 Research Drive, Bethlehem, PA 18015, United States; Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem, PA 18015, United States.
| |
Collapse
|
33
|
Gouveia RP, Barreto CAV, Melo R, Carvalho AL, Moreira IS. Exploring the impact of the stargazin V143L mutation on the dynamics of the AMPA receptor: stargazin complex. Front Cell Neurosci 2025; 18:1505846. [PMID: 39895898 PMCID: PMC11782175 DOI: 10.3389/fncel.2024.1505846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/09/2024] [Indexed: 02/04/2025] Open
Abstract
Stargazin, a transmembrane AMPAR regulatory protein (TARP), plays a crucial role in facilitating the transport of AMPA receptors to the cell surface, stabilising their localisation at synapses and influencing their gating properties. The primary objective of this study was to investigate the effect of the V143L mutation in stargazin, previously linked to intellectual disability, on the interaction between stargazin and AMPA receptors. To achieve this, we conducted a thorough examination of eight distinct molecular dynamics simulations of AMPA receptor-stargazin complexes, each associated with different conductance levels. Through extensive analysis of complex interface structures and dynamics, we revealed that the stargazin V143L mutation had a more pronounced destabilising effect on complexes with lower conductance levels than on the conductive states of the receptor, suggesting a potential association with impaired synaptic transmission in individuals with this mutation.
Collapse
Affiliation(s)
- Raquel P. Gouveia
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Carlos A. V. Barreto
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIs—Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Rita Melo
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Bobadela LRS, Portugal
| | - Ana Luísa Carvalho
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Irina S. Moreira
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
34
|
Lloyd AR, Austin-Muttitt K, Mullins JGL. In silico drug repurposing at the cytoplasmic surface of human aquaporin 1. PLoS One 2025; 20:e0314151. [PMID: 39787482 PMCID: PMC11717375 DOI: 10.1371/journal.pone.0314151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/05/2024] [Indexed: 01/12/2025] Open
Abstract
Aquaporin 1 (AQP1) is a key channel for water transport in peritoneal dialysis. Inhibition of AQP1 could therefore impair water transport during peritoneal dialysis. It is not known whether inhibition of AQP1 occurs unintentionally due to off-target interactions of administered medications. A high-throughput virtual screening study has been performed to investigate the possible binding of licensed medications to the water pore of human AQP1. A complete model of human AQP1 based on its canonical sequence was assembled using I-TASSER and MODELLER. The model was refined via the incorporation of pore water molecules from a high-resolution yeast aquaporin structure. Docking studies were conducted for the cytoplasmic domain of the AQP1 monomer against a library of all compounds listed in the British National Formulary (BNF), using the PLANTS software with the ChemPLP scoring function. The stability of the best docked conformations within the intrinsic water pore was assessed via short 15 nanosecond molecular dynamics (MD) simulations using the GROMACS-on-Colab utility. Of the 1512 compounds tested, 1002 docking results were obtained, and 198 of these conformations occupied a position within the intrinsic water pore. 30 compounds with promising docking scores were assessed by MD. The docked conformations for dopamine, gabapentin, pregabalin, and methyldopa were stable in these short MD studies. For furosemide and pravastatin, the MD trajectory suggested a binding mode different to the docking result. A small set of compounds which could impede water transport through human AQP1 have been identified in this computational screening study.
Collapse
Affiliation(s)
- Aled R. Lloyd
- Genome and Structural Bioinformatics Group, Faculty of Medicine, Health and Life Science, Swansea University, Swansea, Wales, United Kingdom
| | - Karl Austin-Muttitt
- Genome and Structural Bioinformatics Group, Faculty of Medicine, Health and Life Science, Swansea University, Swansea, Wales, United Kingdom
| | - Jonathan G. L. Mullins
- Genome and Structural Bioinformatics Group, Faculty of Medicine, Health and Life Science, Swansea University, Swansea, Wales, United Kingdom
| |
Collapse
|
35
|
Giese APJ, Weng WH, Kindt KS, Chang HHV, Montgomery JS, Ratzan EM, Beirl AJ, Aponte Rivera R, Lotthammer JM, Walujkar S, Foster MP, Zobeiri OA, Holt JR, Riazuddin S, Cullen KE, Sotomayor M, Ahmed ZM. Complexes of vertebrate TMC1/2 and CIB2/3 proteins form hair-cell mechanotransduction cation channels. eLife 2025; 12:RP89719. [PMID: 39773557 PMCID: PMC11709434 DOI: 10.7554/elife.89719] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Calcium and integrin-binding protein 2 (CIB2) and CIB3 bind to transmembrane channel-like 1 (TMC1) and TMC2, the pore-forming subunits of the inner-ear mechano-electrical transduction (MET) apparatus. These interactions have been proposed to be functionally relevant across mechanosensory organs and vertebrate species. Here, we show that both CIB2 and CIB3 can form heteromeric complexes with TMC1 and TMC2 and are integral for MET function in mouse cochlea and vestibular end organs as well as in zebrafish inner ear and lateral line. Our AlphaFold 2 models suggest that vertebrate CIB proteins can simultaneously interact with at least two cytoplasmic domains of TMC1 and TMC2 as validated using nuclear magnetic resonance spectroscopy of TMC1 fragments interacting with CIB2 and CIB3. Molecular dynamics simulations of TMC1/2 complexes with CIB2/3 predict that TMCs are structurally stabilized by CIB proteins to form cation channels. Overall, our work demonstrates that intact CIB2/3 and TMC1/2 complexes are integral to hair-cell MET function in vertebrate mechanosensory epithelia.
Collapse
Affiliation(s)
- Arnaud PJ Giese
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | | | - Jonathan S Montgomery
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Evan M Ratzan
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Alisha J Beirl
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Roberto Aponte Rivera
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Jeffrey M Lotthammer
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
| | - Sanket Walujkar
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
| | - Mark P Foster
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Omid A Zobeiri
- Department of Biomedical Engineering, McGill UniversityMontrealCanada
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
| | - Kathleen E Cullen
- Departments of Biomedical Engineering, Neuroscience, and Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Ophthalmology and Visual Sciences, University of Maryland School of MedicineBaltimoreUnited States
| |
Collapse
|
36
|
Beaudoin CA, Kohli M, Salvage SC, Liu H, Arundel SJ, Hamaia SW, Lei M, Huang CLH, Jackson AP. Isoform-specific N-linked glycosylation of NaV channel α-subunits alters β-subunit binding sites. J Gen Physiol 2025; 157:e202413609. [PMID: 39680039 PMCID: PMC11666101 DOI: 10.1085/jgp.202413609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/19/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024] Open
Abstract
Voltage-gated sodium channel α-subunits (NaV1.1-1.9) initiate and propagate action potentials in neurons and myocytes. The NaV β-subunits (β1-4) have been shown to modulate α-subunit properties. Homo-oligomerization of β-subunits on neighboring or opposing plasma membranes has been suggested to facilitate cis or trans interactions, respectively. The interactions between several NaV channel isoforms and β-subunits have been determined using cryogenic electron microscopy (cryo-EM). Interestingly, the NaV cryo-EM structures reveal the presence of N-linked glycosylation sites. However, only the first glycan moieties are typically resolved at each site due to the flexibility of mature glycan trees. Thus, existing cryo-EM structures may risk de-emphasizing the structural implications of glycans on the NaV channels. Herein, molecular modeling and all-atom molecular dynamics simulations were applied to investigate the conformational landscape of N-linked glycans on NaV channel surfaces. The simulations revealed that negatively charged sialic acid residues of two glycan sites may interact with voltage-sensing domains. Notably, two NaV1.5 isoform-specific glycans extensively cover the α-subunit region that, in other NaV channel α-subunit isoforms, corresponds to the binding site for the β1- (and likely β3-) subunit immunoglobulin (Ig) domain. NaV1.8 contains a unique N-linked glycosylation site that likely prevents its interaction with the β2 and β4-subunit Ig-domain. These isoform-specific glycans may have evolved to facilitate specific functional interactions, for example, by redirecting β-subunit Ig-domains outward to permit cis or trans supraclustering within specialized cellular compartments such as the cardiomyocyte perinexal space. Further experimental work is necessary to validate these predictions.
Collapse
Affiliation(s)
| | - Manas Kohli
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Samantha C. Salvage
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Hengrui Liu
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Samuel J. Arundel
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Samir W. Hamaia
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Christopher L.-H. Huang
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Antony P. Jackson
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Şulea TA, Draga S, Mernea M, Corlan AD, Radu BM, Petrescu AJ, Amuzescu B. Differential Inhibition by Cenobamate of Canonical Human Nav1.5 Ion Channels and Several Point Mutants. Int J Mol Sci 2025; 26:358. [PMID: 39796214 PMCID: PMC11720074 DOI: 10.3390/ijms26010358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/21/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025] Open
Abstract
Cenobamate is a new and highly effective antiseizure compound used for the treatment of adults with focal onset seizures and particularly for epilepsy resistant to other antiepileptic drugs. It acts on multiple targets, as it is a positive allosteric activator of γ-aminobutyric acid type A (GABAA) receptors and an inhibitor of neuronal sodium channels, particularly of the late or persistent Na+ current. We recently evidenced the inhibitory effects of cenobamate on the peak and late current component of the human cardiac isoform hNav1.5. The determined apparent IC50 values of 87.6 µM (peak) and 46.5 µM (late current) are within a clinically relevant range of concentrations (the maximal plasma therapeutic effective concentration for a daily dose of 400 mg in humans is 170 µM). In this study, we built a 3D model of the canonical hNav1.5 channel (UniProt Q14524-1) in open conformation using AlphaFold2, embedded it in a DPPC lipid bilayer, corrected the residue protonation state (pH 7.2) with H++, and added 2 Na+ ions in the selectivity filter. By molecular docking, we found the cenobamate binding site in the central cavity. We identified 10-point mutant variants in the binding site region and explored them via docking and MD. Mutants N1462K/Y (rs1064795922, rs199473614) and M1765R (rs752476527) (by docking) and N932S (rs2061582195) (by MD) featured higher predicted affinity than wild-type.
Collapse
Affiliation(s)
- Teodor Asvadur Şulea
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry of the Romanian Academy, Splaiul Independentei 296, 060031 Bucharest, Romania; (T.A.Ş.); (A.-J.P.)
| | - Sorin Draga
- Biotehnos SA, Gorunului Str. 3-5, 075100 Otopeni, Romania;
- Non-Governmental Research Organization Biologic, 14 Schitului Str., 032044 Bucharest, Romania
| | - Maria Mernea
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania; (B.M.R.); (B.A.)
| | - Alexandru Dan Corlan
- Cardiology Research Unit, University and Emergency Hospital of Bucharest, Splaiul Independenței 169, 050098 Bucharest, Romania;
| | - Beatrice Mihaela Radu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania; (B.M.R.); (B.A.)
| | - Andrei-Jose Petrescu
- Department of Bioinformatics and Structural Biochemistry, Institute of Biochemistry of the Romanian Academy, Splaiul Independentei 296, 060031 Bucharest, Romania; (T.A.Ş.); (A.-J.P.)
| | - Bogdan Amuzescu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania; (B.M.R.); (B.A.)
| |
Collapse
|
38
|
Campbell O, Allsopp R, Klauda JB, Monje V. Atomistic Simulations and Analysis of Peripheral Membrane Proteins with Model Lipid Bilayers. Methods Mol Biol 2025; 2888:281-303. [PMID: 39699738 DOI: 10.1007/978-1-0716-4318-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
All-atom molecular dynamics (AAMD) is a computational technique that predicts the movement of particles based on the intermolecular forces acting on the system. It enables the study of biological systems at atomic detail, complements observations from experiments, and can help the selection of experimental targets. Here, we describe the applications of MD simulations to study the interaction between peripheral membrane proteins and lipid bilayers. Specifically, we provide step-by-step instructions to set up MD simulations to study the binding and interaction of ALPS, the amphipathic helix of the lipid transport protein Osh4, and Thanatin, an antimicrobial peptide with model membranes. We describe examples of systems built with fully atomistic lipid tails and those truncated with the highly-mobile-membrane-mimetic method to enhance conformational sampling. We also comment on the importance of lipid diversity, molecular resolution, and best practices for constructing, running, and analyzing protein-lipid simulation systems. In this second edition, we include a brief discussion on alternative approaches and software to construct protein-membrane coordinate systems, as well as analysis tools and practices that have become relevant to examining protein-lipid interactions since the first edition of this chapter.
Collapse
Affiliation(s)
- Oluwatoyin Campbell
- Chemical and Biological Engineering Department, School of Engineering and Applied Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Robert Allsopp
- Chemical and Biomolecular Engineering Department, A. James Clark School of Engineering, University of Maryland, College Park, MD, USA
| | - Jeffery B Klauda
- Chemical and Biomolecular Engineering Department, A. James Clark School of Engineering, University of Maryland, College Park, MD, USA
| | - Viviana Monje
- Chemical and Biological Engineering Department, School of Engineering and Applied Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
39
|
Nguyen SV, Planalp RP, Vashisth H. Role of sequence length and functionalization in interactions of bioconjugated peptides with mitomembranes. Biointerphases 2025; 20:011006. [PMID: 39998173 PMCID: PMC11906191 DOI: 10.1116/6.0004197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Cell-penetrating peptides are efficient tools for intracellular delivery of a variety of cargoes. In this study, we explored the effect of chain length, side chain chemistry, and the locations of conjugated molecules on the interaction between iron-chelating peptides and a mitochondrial-mimicking membrane. We report that a longer chain length enhanced peptide/membrane interactions, and conjugation at the N-terminus lowered the free-energy barrier for peptide translocation across the membrane. Peptides containing Phe side chains and those containing modified Phe (cyclohexane) side chains showed comparable peptide/membrane energetics and translocation energy barriers. Using steered molecular dynamics (SMD) simulations, we further probed the mechanistic details of translocation of each N-terminated peptide across the membrane and compared their metastable states. At a higher steering velocity, the peptide adopted a compact structure due to frequent π-π interactions among conjugated molecules, but at lower steering velocities, each N-terminated peptide adopted an extended structure. This structure allowed cationic residues to maximize their interactions with phosphate headgroups in the mitomembrane. The hydrophobic residues also formed interactions with the lipid acyl tails, facilitating the passage of peptides across the membrane with decreased free energy barriers. Our results highlight the significance of peptide chain length and conjugation in facilitating peptide transport across the membrane.
Collapse
Affiliation(s)
- Son V Nguyen
- Department of Chemistry, University of New Hampshire, Durham, New Hampshire 03824
| | - Roy P Planalp
- Department of Chemistry, University of New Hampshire, Durham, New Hampshire 03824
| | - Harish Vashisth
- Department of Chemistry, University of New Hampshire, Durham, New Hampshire 03824
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire 03824
- Integrated Applied Mathematics Program, University of New Hampshire, Durham, New Hampshire 03824
- Molecular and Cellular Biotechnology Program, University of New Hampshire, Durham, New Hampshire 03824
| |
Collapse
|
40
|
Dehury B, Mishra S, Panda S, Singh MK, Simha NL, Pati S. Structural Dynamics of Neutral Amino Acid Transporter SLC6A19 in Simple and Complex Lipid Bilayers. J Cell Biochem 2025; 126:e30693. [PMID: 39749651 PMCID: PMC11696832 DOI: 10.1002/jcb.30693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025]
Abstract
B0AT1 (SLC6A19) is a major sodium-coupled neutral amino acid transporter that relies on angiotensin converting enzyme 2 (ACE2) or collectrin for membrane trafficking. Despite its significant role in disorders associated with amino acid metabolism, there is a deficit of comprehensive structure-function understanding of B0AT1 in lipid environment. Herein, we have employed molecular dynamics (MD) simulations to explore the architectural characteristics of B0AT1 in two distinct environments: a simplified POPC bilayer and a complex lipid system replicating the native membrane composition. Notably, our B0AT1 analysis in terms of structural stability and regions of maximum flexibility shows consistency in both the systems with enhanced structural features in the case of complex lipid system. Our findings suggest that diacylglycerol phospholipids significantly alter the pore radius, hydrophobic index, and surface charge distribution of B0AT1, thereby affecting the flexibility of transmembrane helices TM7, TM12, and loop connecting TM7-TM8, crucial for ACE2-B0AT1 interaction. Pro41, Ser190, Arg214, Arg240, Ser413, Pro414, Cys463, and Val582 are among the most prominent lipid binding residues that might influence B0AT1 functionality. We also perceive notable lipid mediated deviation in the degree of tilt and loss of helicity in TM1 and TM6 which might affect the substrate binding sites S1 and S2 in B0AT1. Considerably, destabilization in the structure of B0AT1 in lipid environment was evident upon mutation in TM domain, associated with Hartnup disorder through various structure-based protein stability tools. Our two-tiered approach allowed us to validate the use of POPC as a baseline for initial analyses of SLC transporters. Altogether, our all-atoms MD study provides a platform for future investigations into the structure-function mechanism of B0AT1 in realistic lipid mimetic bilayers and offers a framework for developing new therapeutic agents targeting this transporter.
Collapse
Affiliation(s)
- Budheswar Dehury
- Department of Bioinformatics, Manipal School of Life SciencesManipal Academy of Higher EducationManipalIndia
- Bioinformatics Division I Microbiology DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| | - Sarbani Mishra
- Bioinformatics Division I Microbiology DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| | - Sunita Panda
- Bioinformatics Division I Microbiology DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| | | | - Nischal L. Simha
- Department of Bioinformatics, Manipal School of Life SciencesManipal Academy of Higher EducationManipalIndia
| | - Sanghamitra Pati
- Bioinformatics Division I Microbiology DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| |
Collapse
|
41
|
Lira AL, Taskin B, Puy C, Keshari RS, Silasi R, Pang J, Aslan JE, Shatzel JJ, Lorentz CU, Tucker EI, Schmaier AH, Gailani D, Lupu F, McCarty OJT. The physicochemical properties of lipopolysaccharide chemotypes regulate activation of the contact pathway of blood coagulation. J Biol Chem 2025; 301:108110. [PMID: 39706265 PMCID: PMC11773025 DOI: 10.1016/j.jbc.2024.108110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/15/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
Lipopolysaccharide (LPS) is the primary pathogenic factor in Gram-negative sepsis. While the presence of LPS in the bloodstream during infection is associated with disseminated intravascular coagulation, the mechanistic link between LPS and blood coagulation activation remains ill-defined. The contact pathway of coagulation-a series of biochemical reactions that initiates blood clotting when plasma factors XII (FXII) and XI (FXI), prekallikrein (PK), and high molecular weight kininogen interact with anionic surfaces-has been shown to be activated in Gram-negative septic patients. In this study, using an in vivo baboon model of Gram-negative Escherichia coli sepsis, we observed activation of the contact pathway including FXII, FXI, and PK. We examined whether E.coli LPS molecules could bind and activate contact pathway members by quantifying the interaction and activation of either FXII, FXI, or PK with each of the three chemotypes of LPS: O111:B4, O26:B6, or Rd2. The LPS chemotypes exhibited distinct physicochemical properties as aggregates and formed complexes with FXII, FXI, and PK. The LPS chemotype O26:B6 uniquely promoted the autoactivation of FXII to FXIIa and, in complex with FXIIa, promoted the cleavage of FXI and PK to generate FXIa and plasma kallikrein, respectively. Furthermore, in complex with the active forms of FXI or PK, LPS chemotypes were able to regulate the catalytic activity of FXIa and plasma kallikrein, respectively, despite the inability to promote the autoactivation of either zymogen. These data suggest that the procoagulant phenotype of E.coli is influenced by bacterial strain and the physicochemical properties of the LPS chemotypes.
Collapse
Affiliation(s)
- André L Lira
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.
| | - Berk Taskin
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Ravi S Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Division of Hematology and Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Christina U Lorentz
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Aronora, Inc, Portland, Oregon, USA
| | - Erik I Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Aronora, Inc, Portland, Oregon, USA
| | - Alvin H Schmaier
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - David Gailani
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
42
|
Zhao M, Lopes LJS, Sahni H, Yadav A, Do HN, Reddy T, López CA, Neale C, Gnanakaran S. Insertion and Anchoring of the HIV-1 Fusion Peptide into a Complex Membrane Mimicking the Human T-Cell. J Phys Chem B 2024; 128:12710-12727. [PMID: 39670799 DOI: 10.1021/acs.jpcb.4c05018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
A fundamental understanding of how the HIV-1 envelope (Env) protein facilitates fusion is still lacking. The HIV-1 fusion peptide, consisting of 15 to 22 residues, is the N-terminus of the gp41 subunit of the Env protein. Further, this peptide, a promising vaccine candidate, initiates viral entry into target cells by inserting and anchoring into human immune cells. The influence of membrane lipid reorganization and the conformational changes of the fusion peptide during the membrane insertion and anchoring processes, which can significantly affect HIV-1 cell entry, remains largely unexplored due to the limitations of experimental measurements. In this work, we investigate the insertion of the fusion peptide into an immune cell membrane mimic through multiscale molecular dynamics simulations. We mimic the native T-cell by constructing a nine-lipid asymmetric membrane, along with geometrical restraints accounting for insertion in the context of gp41. To account for the slow time scale of lipid mixing while enabling conformational changes, we implement a protocol to go back and forth between atomistic and coarse-grained simulations. Our study provides a molecular understanding of the interactions between the HIV-1 fusion peptide and the T-cell membrane, highlighting the importance of the conformational flexibility of fusion peptides and local lipid reorganization in stabilizing the anchoring of gp41 into the targeted host membrane during the early events of HIV-1 cell entry. Importantly, we identify a motif within the fusion peptide critical for fusion that can be further manipulated in future immunological studies.
Collapse
Affiliation(s)
- Mingfei Zhao
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Laura J S Lopes
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Harshita Sahni
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
- Department of Computer Science, University of New Mexico, Albuquerque, New Mexico 87106,United States
| | - Anju Yadav
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, Texas 79968,United States
| | - Hung N Do
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Tyler Reddy
- CCS-7 Applied Computer Science Group, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Cesar A López
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Chris Neale
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - S Gnanakaran
- T-6 Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| |
Collapse
|
43
|
Mei X, Xiang W, Pan W, Lin Q, Jia X, Zhang X, Tang X, Cheng X, Weng Y, Yang K, Lu N. Plasmalogens Reversed Oxidative Stress and Inflammatory Response Exacerbated by Damage to Cell Membrane Properties in Acute Liver Injury. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28280-28293. [PMID: 39576750 DOI: 10.1021/acs.jafc.4c06929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
BACKGROUND In acute liver injury (ALI), cell membrane damage could induce an inflammatory response and oxidative stress. As a membrane glycerophospholipid, plasmalogens (PLS) are crucial in regulating the cell membrane properties and exhibit beneficial effects in various liver diseases. However, the specific regulatory effects of PLS in the ALI remain unknown. METHODS We utilized CCl4 to induce ALI in AML12 hepatocytes and C57BL/6J mice and examined oxidative stress indicators and inflammatory cytokine levels. Our study further validated the effect of PLS on cell membrane integrity by Lactate Dehydrogenase (LDH) release assay and Dil/Calcein assay, and molecular dynamics (MD) simulations were employed to elucidate the molecular mechanisms by which PLS affected cell membranes. RESULTS PLS attenuated hepatocyte damage both in vivo and in vitro. Moreover, PLS increased levels of SOD, GSH, and CAT and inhibited the production of malondialdehyde. PLS succeeded in decreasing proinflammatory cytokines (TNF-α, IL-1β, and IL-6) while increasing anti-inflammatory cytokines (IL-10). Furthermore, PLS effectively maintained the cell membrane integrity. The MD simulations well explained the molecular mechanisms: a high level of PLS modulated the cell membrane properties, enabling them to be more flexible, elastic, and less prone to rupture. CONCLUSIONS Our study illustrated the effect and molecular mechanisms of PLS against ALI, potentially broadening its application in liver diseases.
Collapse
Affiliation(s)
- Xue Mei
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province 214122, People's Republic of China
| | - Wen Xiang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province 214125, People's Republic of China
| | - Wenyan Pan
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, Jiangsu Province 215006, People's Republic of China
| | - Quan Lin
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province 214122, People's Republic of China
| | - Xueyan Jia
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province 214122, People's Republic of China
| | - Xuan Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province 214122, People's Republic of China
| | - Xue Tang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province 214122, People's Republic of China
| | - Xiangrong Cheng
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province 214122, People's Republic of China
| | - Yuyan Weng
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, Jiangsu Province 215006, People's Republic of China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, Jiangsu Province 215006, People's Republic of China
| | - Naiyan Lu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province 214122, People's Republic of China
- National Laboratory of Solid State Microstructures, Nanjing University, Nanjing, Jiangsu Province 210093, People's Republic of China
| |
Collapse
|
44
|
Salam DSA, Gunasinghe KKJ, Hwang SS, Ginjom IRH, Chee Wezen X, Rahman T. In Silico Modeling and Characterization of Epstein-Barr Virus Latent Membrane Protein 1 Protein. ACS OMEGA 2024; 9:49422-49431. [PMID: 39713625 PMCID: PMC11656244 DOI: 10.1021/acsomega.4c06868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024]
Abstract
Latent membrane protein 1 (LMP1) plays a crucial role in Epstein-Barr virus (EBV)'s ability to establish latency and is involved in developing and progressing EBV-associated cancers. Additionally, EBV-infected cells affect the immune responses, making it challenging for the immune system to eliminate them. Due to the aforementioned reasons, it is crucial to understand the structural features of LMP1, which are essential for the development of novel cancer therapies that target its signaling pathways. To date, there is yet to be a complete LMP1 protein structure; therefore, in our work, we modeled the full-length LMP1 containing the short cytoplasmic N-terminus, six transmembrane domains (TMDs), and a long-simulated C-terminus. Our model showed good stability and protein compactness evaluated through accelerated-molecular dynamics, where the conformational ensemble exhibited compact folds, particularly in the TMDs. Our results suggest that specific domains or motifs, predominantly in the C-terminal domain of LMP1, show promise as potential drug targets. As a whole, our work provides insights into key structural features of LMP1 that will allow the development of novel LMP1 therapies.
Collapse
Affiliation(s)
- Dayang-Sharyati
D. A. Salam
- Faculty
of Engineering, Computing and Science, Swinburne
University of Technology Sarawak, Kuching 93350, Malaysia
| | | | - Siaw San Hwang
- Faculty
of Engineering, Computing and Science, Swinburne
University of Technology Sarawak, Kuching 93350, Malaysia
| | - Irine Runnie Henry Ginjom
- Faculty
of Engineering, Computing and Science, Swinburne
University of Technology Sarawak, Kuching 93350, Malaysia
| | - Xavier Chee Wezen
- Faculty
of Engineering, Computing and Science, Swinburne
University of Technology Sarawak, Kuching 93350, Malaysia
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, U.K.
| |
Collapse
|
45
|
T Magalhães B, S Coimbra JT, M Silva R, Ferreira M, S Santos R, Gameiro P, Azevedo NF, Fernandes PA. Crosstalk of Nucleic Acid Mimics with Lipid Membranes: A Multifaceted Computational and Experimental Study. Biochemistry 2024; 63:3381-3394. [PMID: 39571107 DOI: 10.1021/acs.biochem.4c00349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Nucleic acid mimics (NAMs) have demonstrated high potential as antibacterial drugs. However, very few studies have assessed their possible diffusion across the bacterial envelope. In this work, we studied NAMs' diffusion in lipid bilayer systems that mimic the bacterial outer membrane using molecular dynamics (MD) simulations. Additionally, we examined the interactions of a NAM sequence with lipid membranes and ascertained the partition constants (Kp) through MD and spectroscopic investigations. The NAM sequences were composed of locked nucleic acid (LNA) and 2'-O-methyl (2'-OMe) residues, whereas the membrane models were composed of 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC) or 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (POPG) phospholipids. The parametrization protocol followed was validated against literature data and demonstrated the reliability of our approach for simulating NAM sequences. Investigation into the interaction of the sequences with zwitterionic and anionic membranes revealed a preference for hydrogen bond formation with the anionic model over the zwitterionic one. Additionally, potential of mean force (PMF) calculations unveiled a lower free energy barrier for translocation across the zwitterionic bilayer model. Contrarily, the partition constants derived suggested a slightly higher partitioning within the anionic membrane, emphasizing a nuanced interplay of factors. Finally, spectroscopic partition measurements with liposomes presented challenges in quantifying the partition of NAMs due to minimal signal variations. However, a tendency for quenching in anionic vesicles suggested a potential, albeit small, partitioning effect that warrants further investigation. In summary, our study revealed that NAMs will not, in principle, be able to cross an intact bacterial outer membrane by passive diffusion.
Collapse
Affiliation(s)
- Beatriz T Magalhães
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - João T S Coimbra
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Raquel M Silva
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Mariana Ferreira
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Rita S Santos
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Paula Gameiro
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Nuno F Azevedo
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Pedro A Fernandes
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências Universidade do Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| |
Collapse
|
46
|
Somboon K, Melling O, Lejeune M, Pinheiro GMS, Paquelin A, Bardiaux B, Nilges M, Delepelaire P, Khalid S, Izadi-Pruneyre N. Dynamic interplay between a TonB-dependent heme transporter and a TonB protein in a membrane environment. mBio 2024; 15:e0178124. [PMID: 39475239 PMCID: PMC11633176 DOI: 10.1128/mbio.01781-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/03/2024] [Indexed: 12/12/2024] Open
Abstract
The envelope of Gram-negative bacteria is composed of two membranes separated by the periplasmic space. This organization imposes geometrical and distance constraints that are key for the mechanism of action of multicomponent systems spanning the envelope. However, consideration of all three compartments by experimental approaches is still elusive. Here, we have used the state-of-the-art molecular dynamics simulation in an Escherichia coli envelope model to obtain a dynamic view of molecular interactions between the outer membrane heme transporter HasR and the inner membrane TonB-like protein HasB. Their interaction allows the transfer of the inner membrane proton-motive force derived energy to the transporter for heme internalization. The simulations that incorporate both membranes show the key role of periplasmic domains of both proteins and their dynamics in complex formation and stability. They revealed a previously unidentified network of HasR-HasB protein-protein interactions in the periplasm. Experimental validation (mutations, in vivo phenotypic and biophysical assays) provides support for the simulation-predicted interactions. Based on structural and sequence conservation, the network of interaction revealed in this study is expected to occur in other nutrient import systems. IMPORTANCE Gram-negative bacteria import scarce nutrients such as metals and vitamins by an energized mechanism involving a multicomponent protein system that spans the cell envelope. It consists of an outer membrane TonB-dependent transporter (TBDT) and a TonB complex in the inner membrane that provides the proton motive force energy for the nutrient entry. Despite the intense research efforts focused on this system (a) from structural and fundamental microbiology perspectives and (b) for the interest in the development of new antibacterial strategies, the molecular mechanism of the system is not at all well understood. The lack of understanding comes from incomplete structural data and the experimental difficulties of studying an inherently flexible multicomponent complex that resides within the heterogeneous environment of the double membrane bacterial cell envelope. To address these challenges and obtain a comprehensive view of the molecular interactions at atomic level, here, we have used the combined power of advanced molecular simulations and complementary microbiology and biochemical experiments. Our results represent a significant step forward in understanding the structural and molecular bases of this vital mechanism.
Collapse
Affiliation(s)
- Kamolrat Somboon
- School of Chemistry, University of Southampton, Southampton, United Kingdom
| | - Oliver Melling
- School of Chemistry, University of Southampton, Southampton, United Kingdom
| | - Maylis Lejeune
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Bacterial Transmembrane Systems Unit, Paris, France
| | - Glaucia M. S. Pinheiro
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Bacterial Transmembrane Systems Unit, Paris, France
| | - Annick Paquelin
- Institut de Biologie Physico-Chimique, UMR 7099, CNRS Université de Paris, Paris, France
| | - Benjamin Bardiaux
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Bacterial Transmembrane Systems Unit, Paris, France
| | - Michael Nilges
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, Paris, France
| | - Phillippe Delepelaire
- Institut de Biologie Physico-Chimique, UMR 7099, CNRS Université de Paris, Paris, France
| | - Syma Khalid
- School of Chemistry, University of Southampton, Southampton, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Nadia Izadi-Pruneyre
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Structural Bioinformatics Unit, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR3528, Bacterial Transmembrane Systems Unit, Paris, France
| |
Collapse
|
47
|
Paudel Y, Valiente PA, Kim J, Kim PM. A High-Throughput Method for Screening Peptide Activators of G-Protein-Coupled Receptors. ACS OMEGA 2024; 9:48471-48479. [PMID: 39676964 PMCID: PMC11635519 DOI: 10.1021/acsomega.4c07071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/16/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Abstract
Here, we describe an innovative and efficient method for screening peptide activators of G-protein-coupled receptors (GPCRs) utilizing a protein-protein interaction (PPI) approach. We designed a library of 92,918 peptides fused with transmembrane domains of glycosylphosphatidylinositol-anchored proteins (GPI-APs). We employed a pooled lentiviral system to promote the expression of these proteins at the cellular membrane and evaluate their ability to activate GPCRs. We then used fluorescence-activated cell sorting (FACS) to screen the GPI-AP-peptide library and identify novel peptide activators of the glucagon-like peptide-1 receptor (GLP-1R). We discovered one peptide PepA3 derived from the Frizzled-like (FZ) domain of human Carboxypeptidase Z (CPZ), a regulated secreted metallocarboxypeptidase. Notably, PepA3 and its two related variants, PepA and PepA2, activated the GLP-1R receptor with less potency but comparable efficacy to that of GLP-1. We then hypothesized that all of these peptides will bind differently to the GLP-1R than the normal ligand. Our technology could identify novel GPCR-activating peptides for structure-function or drug discovery research.
Collapse
Affiliation(s)
- Yagya
Prasad Paudel
- Donnelly
Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Pedro A. Valiente
- Donnelly
Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Jisun Kim
- Donnelly
Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Philip M. Kim
- Donnelly
Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
- Department
of Computer Science, University of Toronto, Toronto, ON M5S 3E1, Canada
- Department
of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| |
Collapse
|
48
|
Doktorova M, Symons JL, Zhang X, Wang HY, Schlegel J, Lorent JH, Heberle FA, Sezgin E, Lyman E, Levental KR, Levental I. Cell membranes sustain phospholipid imbalance via cholesterol asymmetry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.30.551157. [PMID: 39713443 PMCID: PMC11661119 DOI: 10.1101/2023.07.30.551157] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Membranes are molecular interfaces that compartmentalize cells to control the flow of nutrients and information. These functions are facilitated by diverse collections of lipids, nearly all of which are distributed asymmetrically between the two bilayer leaflets. Most models of biomembrane structure and function often include the implicit assumption that these leaflets have similar abundances of phospholipids. Here, we show that this assumption is generally invalid and investigate the consequences of lipid abundance imbalances in mammalian plasma membranes (PM). Using quantitative lipidomics, we discovered that cytoplasmic leaflets of human erythrocyte membranes have >50% overabundance of phospholipids compared to exoplasmic leaflets. This imbalance is enabled by an asymmetric interleaflet distribution of cholesterol, which regulates cellular cholesterol homeostasis. These features produce unique functional characteristics, including low PM permeability and resting tension in the cytoplasmic leaflet that regulates protein localization. These largely overlooked aspects of membrane asymmetry represent an evolution of classic paradigms of biomembrane structure and physiology.
Collapse
Affiliation(s)
- Milka Doktorova
- Department of Biochemistry and Biophysics, Stockholm University; Science for Life Laboratory, Solna 171 65, Sweden
| | - Jessica L. Symons
- Department of Integrative Biology & Pharmacology, University of Texas Health Science Center at Houston; Houston, TX 77030, USA
| | - Xiaoxuan Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia; Charlottesville, VA 22903, USA
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia; Charlottesville, VA 22903, USA
| | - Jan Schlegel
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet; 17165 Solna, Sweden
| | - Joseph H. Lorent
- Department of Cellular and Molecular Pharmacology, TFAR, LDRI, UCLouvain; Avenue Mounier 73, B1.73.05, B-1200 Brussels
| | - Frederick A. Heberle
- Department of Chemistry, University of Tennessee Knoxville; Knoxville, TN 37916, USA
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet; 17165 Solna, Sweden
| | - Edward Lyman
- Department of Physics and Astronomy; Department of Chemistry and Biochemistry, University of Delaware; Newark, DE 19716, USA
| | - Kandice R. Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia; Charlottesville, VA 22903, USA
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia; Charlottesville, VA 22903, USA
| |
Collapse
|
49
|
Gao Q, Ge L, Wang Y, Zhu Y, Liu Y, Zhang H, Huang J, Qin Z. An explainable few-shot learning model for the directed evolution of antimicrobial peptides. Int J Biol Macromol 2024; 285:138272. [PMID: 39631577 DOI: 10.1016/j.ijbiomac.2024.138272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/20/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Due to the persistent threat of antibiotic resistance posed by Gram-negative pathogens, the discovery of new antimicrobial agents is of critical importance. In this study, we employed deep learning-guided directed evolution to explore the chemical space of antimicrobial peptides (AMPs), which present promising alternatives to traditional small-molecule antibiotics. Utilizing a fine-tuned protein language model tailored for small dataset learning, we achieved structural modifications of the lipopolysaccharide-binding domain (LBD) derived from Marsupenaeus japonicus, a prawn species of considerable value in aquaculture and commercial fisheries. The engineered LBDs demonstrated exceptional activity against a range of Gram-negative pathogens. Drawing inspiration from evolutionary principles, we elucidated the bactericidal mechanism through molecular dynamics simulations and mapped the directed evolution pathways using a ladderpath framework. This work highlights the efficacy of explainable few-shot learning in the rational design of AMPs through directed evolution.
Collapse
Affiliation(s)
- Qiandi Gao
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Liangjun Ge
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yihan Wang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yanran Zhu
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Yu Liu
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| | - Heqian Zhang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China.
| | - Jiaquan Huang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China.
| | - Zhiwei Qin
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
50
|
Morinelli L, Corradi B, Arnaldi P, Cortese K, Muià M, Zara F, Maragliano L, Sterlini B, Corradi A. Unraveling the Membrane Topology of TMEM151A: A Step Towards Understanding its Cellular Role. J Mol Biol 2024; 436:168834. [PMID: 39454747 DOI: 10.1016/j.jmb.2024.168834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/04/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
Transmembrane protein 151A (TMEM151A) has been identified as a causative gene for paroxysmal kinesigenic dyskinesia, though its molecular function remains almost completely unknown. Understanding the membrane topology of transmembrane proteins is crucial for elucidating their functions and possible interacting partners. In this study, we utilized molecular dynamics simulations, immunocytochemistry, and electron microscopy to define the topology of TMEM151A. Our results validate a starting AlphaFold model of TMEM151A and reveal that it comprises a transmembrane domain with two membrane-spanning alpha helices connected by a short extracellular loop and an intramembrane helix-hinge-helix structure. Notably, most of the protein is oriented towards the intracellular side of the membranes with a large cytosolic domain featuring a combination of alpha-helix and beta-sheet structures, as well as the protein N- and C-termini. These insights into TMEM151A's topology and orientation of its domains with respect of the cell membranes provide essential information for future functional studies and represent a first fundamental step for understanding its role in the pathogenesis of paroxysmal kinesigenic dyskinesia.
Collapse
Affiliation(s)
- Lisastella Morinelli
- University of Genova, Department of Experimental Medicine, Genova, Italy; Istituto Italiano di Tecnologia, Center for Synaptic Neuroscience and Technology, Genova, Italy
| | - Beatrice Corradi
- University of Genova, Department of Experimental Medicine, Genova, Italy; Istituto Italiano di Tecnologia, Center for Synaptic Neuroscience and Technology, Genova, Italy
| | - Pietro Arnaldi
- University of Genova, Department of Experimental Medicine, Genova, Italy
| | - Katia Cortese
- University of Genova, Department of Experimental Medicine, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Martina Muià
- University of Genova, Department of Experimental Medicine, Genova, Italy
| | - Federico Zara
- University of Genova, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Genova, Italy; IRCCS Istituto Giannina Gaslini, Unit of Medical Genetics, Genova, Italy
| | - Luca Maragliano
- Istituto Italiano di Tecnologia, Center for Synaptic Neuroscience and Technology, Genova, Italy; Polytechnic University of Marche, Department of Life and Environmental Sciences, Ancona, Italy
| | - Bruno Sterlini
- University of Genova, Department of Experimental Medicine, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Anna Corradi
- University of Genova, Department of Experimental Medicine, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|