1
|
Luo Y, Sun L, Peng Y. The structural basis of the G protein-coupled receptor and ion channel axis. Curr Res Struct Biol 2025; 9:100165. [PMID: 40083915 PMCID: PMC11904507 DOI: 10.1016/j.crstbi.2025.100165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Sensory neurons play an essential role in recognizing and responding to detrimental, irritating, and inflammatory stimuli from our surroundings, such as pain, itch, cough, and neurogenic inflammation. The transduction of these physiological signals is chiefly mediated by G protein-coupled receptors (GPCRs) and ion channels. The binding of ligands to GPCRs triggers a signaling cascade, recruiting G proteins or β-arrestins, which subsequently interact with ion channels (e.g., GIRK and TRP channels). This interaction leads to the sensitization and activation of these channels, initiating the neuron's protective mechanisms. This review delves into the complex interplay between GPCRs and ion channels that underpin these physiological processes, with a particular focus on the role of structural biology in enhancing our comprehension. Through unraveling the intricacies of the GPCR-ion channel axis, we aim to shed light on the sophisticated intermolecular dynamics within these pivotal membrane protein families, ultimately guiding the development of precise therapeutic interventions.
Collapse
Affiliation(s)
- Yulin Luo
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, L Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Liping Sun
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yao Peng
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| |
Collapse
|
2
|
Saffarian Delkhosh A, Hadadianpour E, Islam MM, Georgieva ER. Highly versatile small virus-encoded proteins in cellular membranes: A structural perspective on how proteins' inherent conformational plasticity couples with host membranes' properties to control cellular processes. J Struct Biol X 2025; 11:100117. [PMID: 39802090 PMCID: PMC11714672 DOI: 10.1016/j.yjsbx.2024.100117] [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: 08/08/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
We investigated several small viral proteins that reside and function in cellular membranes. These proteins belong to the viroporin family because they assemble into ion-conducting oligomers. However, despite forming similar oligomeric structures with analogous functions, these proteins have diverse amino acid sequences. In particular, the amino acid compositions of the proposed channel-forming transmembrane (TM) helices are vastly different-some contain residues (e.g., His, Trp, Asp, Ser) that could facilitate cation transport. Still, other viroporins' TM helices encompass exclusively hydrophobic residues; therefore, it is difficult to explain their channels' activity, unless other mechanisms (e.g., involving a negative lipid headgroups and/or membrane destabilization) take place. For this study, we selected the M2, Vpu, E, p13II, p7, and 2B proteins from the influenza A, HIV-1, human T-cell leukemia, hepatitis C, and picorna viruses, respectively. We provide a brief overview of the current knowledge about these proteins' structures as well as remaining questions about more comprehensive understanding of their structures, conformational dynamics, and function. Finally, we outline strategies to utilize a multi-prong structural and computational approach to overcome current deficiencies in the knowledge about these proteins.
Collapse
Affiliation(s)
| | | | - Md Majharul Islam
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
| | - Elka R. Georgieva
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
3
|
Banerjee C, Wey-Hung Liauw B, Vafabakhsh R. Direct effect of membrane environment on the activation of mGluR2 revealed by single-molecule FRET. Structure 2025; 33:718-727.e4. [PMID: 39909029 PMCID: PMC11972159 DOI: 10.1016/j.str.2025.01.011] [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: 09/10/2024] [Revised: 11/27/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025]
Abstract
The microenvironment of membrane receptors controls their mobility, structure, interactions, and dynamics, but a systematic understanding of how it modulates receptor function is often lacking. Using single-molecule Förster resonance energy transfer (smFRET), we characterized how detergents and cholesterol modulate the conformational dynamics of metabotropic glutamate receptor 2 (mGluR2), a class C G protein-coupled receptor (GPCR). We found that, within the resolution of our measurements, all tested detergents stabilize the same overall active and inactive structure of different domains of mGluR2. However, the degree of stabilization and the equilibrium between active and inactive conformations depended on the detergent. Detergents with a single hydrophobic tail increased the active state occupancy compared to those with long, branched tails. Adding cholesterol to micelles with branched hydrophobic tails shifted the equilibrium toward the inactive state. Mutagenesis identified residues potentially involved in cholesterol interaction with mGluR2. Targeting the cholesterol-binding site with synthetic molecules could be a viable therapeutic approach.
Collapse
Affiliation(s)
- Chiranjib Banerjee
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | | | - Reza Vafabakhsh
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
4
|
Doktorova M, Daum S, Reagle TR, Cannon HI, Ebenhan J, Neudorf S, Han B, Sharma S, Kasson P, Levental K, Bacia K, Kenworthy AK, Levental I. Caveolin assemblies displace one bilayer leaflet to organize and bend membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.28.610209. [PMID: 39257813 PMCID: PMC11383982 DOI: 10.1101/2024.08.28.610209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Caveolin is a monotopic integral membrane protein, widely expressed in metazoa and responsible for constructing enigmatic membrane invaginations known as caveolae. Recently, the high-resolution structure of a purified human caveolin assembly, the CAV1-8S complex, revealed a unique organization of 11 protomers arranged in a tightly packed, radially symmetric spiral disc. One face and the outer rim of this disc are hydrophobic, suggesting that the complex incorporates into membranes by displacing hundreds of lipids from one leaflet. The feasibility of this unique molecular architecture and its biophysical and functional consequences are currently unknown. Using Langmuir film balance measurements, we find that CAV1-8S is highly surface active, intercalating into lipid monolayers of various compositions. CAV1-8S can also incorporate into pre-formed bilayers, but only upon removal of phospholipids from the outer-facing leaflet. Atomistic and coarse-grained simulations of biomimetic bilayers support this 'leaflet replacement' model and also reveal that CAV1-8S accumulates 40-70 cholesterol molecules into a disordered monolayer between the complex and its distal lipid leaflet. We find that CAV1-8S preferentially associates with positively curved membrane surfaces due to its influence on the conformations of distal leaflet lipids, and that these effects laterally sort lipids. Large-scale simulations of multiple caveolin assemblies confirmed their association with large, positively curved membrane morphologies consistent with the shape of caveolae. Further, association with curved membranes regulates the exposure of caveolin residues implicated in protein-protein interactions. Altogether, the unique structure of CAV1-8S imparts unusual modes of membrane interaction with implications for membrane organization, morphology, and physiology.
Collapse
|
5
|
Conflitti P, Lyman E, Sansom MSP, Hildebrand PW, Gutiérrez-de-Terán H, Carloni P, Ansell TB, Yuan S, Barth P, Robinson AS, Tate CG, Gloriam D, Grzesiek S, Eddy MT, Prosser S, Limongelli V. Functional dynamics of G protein-coupled receptors reveal new routes for drug discovery. Nat Rev Drug Discov 2025; 24:251-275. [PMID: 39747671 PMCID: PMC11968245 DOI: 10.1038/s41573-024-01083-3] [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] [Accepted: 10/25/2024] [Indexed: 01/04/2025]
Abstract
G protein-coupled receptors (GPCRs) are the largest human membrane protein family that transduce extracellular signals into cellular responses. They are major pharmacological targets, with approximately 26% of marketed drugs targeting GPCRs, primarily at their orthosteric binding site. Despite their prominence, predicting the pharmacological effects of novel GPCR-targeting drugs remains challenging due to the complex functional dynamics of these receptors. Recent advances in X-ray crystallography, cryo-electron microscopy, spectroscopic techniques and molecular simulations have enhanced our understanding of receptor conformational dynamics and ligand interactions with GPCRs. These developments have revealed novel ligand-binding modes, mechanisms of action and druggable pockets. In this Review, we highlight such aspects for recently discovered small-molecule drugs and drug candidates targeting GPCRs, focusing on three categories: allosteric modulators, biased ligands, and bivalent and bitopic compounds. Although studies so far have largely been retrospective, integrating structural data on ligand-induced receptor functional dynamics into the drug discovery pipeline has the potential to guide the identification of drug candidates with specific abilities to modulate GPCR interactions with intracellular effector proteins such as G proteins and β-arrestins, enabling more tailored selectivity and efficacy profiles.
Collapse
Affiliation(s)
- Paolo Conflitti
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Edward Lyman
- Department of Physics and Astronomy, University of Delaware, Newark, DE, USA
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Peter W Hildebrand
- Institute of Medical Physics and Biophysics, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Hugo Gutiérrez-de-Terán
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Paolo Carloni
- INM-9/IAS-5 Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | - T Bertie Ansell
- Department of Biochemistry, University of Oxford, Oxford, UK
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Shuguang Yuan
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Patrick Barth
- Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Anne S Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - David Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen, Denmark
| | - Stephan Grzesiek
- Focal Area Structural Biology and Biophysics, Biozentrum, University of Basel, Basel, Switzerland
| | - Matthew T Eddy
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Scott Prosser
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - Vittorio Limongelli
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland.
| |
Collapse
|
6
|
Idikuda V, Roy Chowdhury S, Chang Y, Ren Q, Bao H, Goldsmith R, Chanda B. Lipid bilayers determine the allostery but not intrinsic affinity of cAMP binding to pacemaker channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.23.630133. [PMID: 39763952 PMCID: PMC11703277 DOI: 10.1101/2024.12.23.630133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2025]
Abstract
Cyclic adenosine monophosphate (cAMP), a second messenger, binds to hyperpolarization and cyclic nucleotide-gated (HCN) ion channels and regulates the automaticity of pacemaking activity. While cellular studies suggest that cAMP binding to HCN channels exhibits unusual cooperativity, recent findings using purified detergent-solubilized channels indicate independent binding to each subunit. This discrepancy raises the question of whether the lipid environment or endogenous cellular cofactors influence cAMP-dependent gating. To address this, we reconstituted purified human HCN channels in nanodiscs and resolved cAMP binding energetics at single-molecule resolution using nanophotonic waveguides. Our measurements reveal that, in contrast to detergent-solubilized channels, cAMP binds cooperatively to HCN channels reconstituted in a variety of lipid nanodiscs. Remarkably, the presence of lipid bilayer promotes ligand-binding allostery but not intrinsic binding affinity. To explore the molecular basis of bilayer-induced allostery, we determine the cryo-EM structure of HCN1 in soy polar lipid nanodiscs at a nominal resolution of 3.77 angstrom resolution. Although the overall architecture is conserved, the average interfacial distance between the transmembrane domain and C-terminal domain of neighboring subunits are shorter in lipid nanodiscs. These findings indicate that the lipid bilayer regulates the function of pacemaker ion channels by enhancing inter-subunit interactions and underscore the fundamental role of membranes in amplifying the gating sensitivity of ion channels by promoting long-range cooperative interactions.
Collapse
|
7
|
Liu J, Zhang X, Zhao B, Ling H, Li Y, Sun K, Chen S, Zhang Y, Zhai T, Zhang Y, Li F, Liu Q. In Situ Monitoring of Membrane Protein Dynamics Using High-Throughput Red-Light-Activated Single-Molecule Tracking. ACS NANO 2025. [PMID: 40153256 DOI: 10.1021/acsnano.5c03182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2025]
Abstract
Single-molecule tracking offers nanometer resolution for studying individual molecule dynamics but is often limited by sparse labeling to avoid signal overlap. We present Red-Light-Activated Single-molecule Tracking (RE-LAST) strategy to address this challenge utilizing a photoactivatable probe, SiR670. SiR670 combines traditional silicon rhodamine with a photocage called SO, quenching fluorescence via photoinduced electron transfer (PET). Red light triggers SiR670 excitation, generating singlet oxygen that oxidizes the SO cage, halting PET and restoring fluorescence. RE-LAST used red light for both activation and imaging, eliminating harmful UV exposure. This method enables high-throughput single-molecule tracking, achieving approximately 9 times more tracks than conventional methods and allowing detailed classification of CD56 membrane protein motion. Furthermore, in situ imaging of single live cells revealed the effects of triplet quencher and oxygen scavenging system (OSS) on membrane protein dynamics. While triplet quenchers like Trolox had minimal impact on protein movement patterns, OSS significantly accelerated protein movement and increased the proportion of mobile proteins. This approach provides a comprehensive method for investigating membrane protein dynamics in living cells, contributing to further developments in cellular and molecular biology.
Collapse
Affiliation(s)
- Jinyang Liu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Xuebo Zhang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Bingjie Zhao
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Huan Ling
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Yanzhong Li
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Kuangshi Sun
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Song Chen
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Yanxin Zhang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Tianli Zhai
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Yunxiang Zhang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| | - Fuyou Li
- Institute of Translational Medicine, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai200240, P.R. China
| | - Qian Liu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials Institution, Fudan University, Shanghai 200438, China
| |
Collapse
|
8
|
Gorman BL, Shafer CC, Ragi N, Sharma K, Neumann EK, Anderton CR. Imaging and spatially resolved mass spectrometry applications in nephrology. Nat Rev Nephrol 2025:10.1038/s41581-025-00946-1. [PMID: 40148534 DOI: 10.1038/s41581-025-00946-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
The application of spatially resolved mass spectrometry (MS) and MS imaging approaches for studying biomolecular processes in the kidney is rapidly growing. These powerful methods, which enable label-free and multiplexed detection of many molecular classes across omics domains (including metabolites, drugs, proteins and protein post-translational modifications), are beginning to reveal new molecular insights related to kidney health and disease. The complexity of the kidney often necessitates multiple scales of analysis for interrogating biofluids, whole organs, functional tissue units, single cells and subcellular compartments. Various MS methods can generate omics data across these spatial domains and facilitate both basic science and pathological assessment of the kidney. Optimal processes related to sample preparation and handling for different MS applications are rapidly evolving. Emerging technology and methods, improvement of spatial resolution, broader molecular characterization, multimodal and multiomics approaches and the use of machine learning and artificial intelligence approaches promise to make these applications even more valuable in the field of nephology. Overall, spatially resolved MS and MS imaging methods have the potential to fill much of the omics gap in systems biology analysis of the kidney and provide functional outputs that cannot be obtained using genomics and transcriptomic methods.
Collapse
Affiliation(s)
- Brittney L Gorman
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Catelynn C Shafer
- Department of Chemistry, University of California, Davis, Davis, CA, 95695, USA
| | - Nagarjunachary Ragi
- Center for Precision Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Kumar Sharma
- Center for Precision Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
- Division of Nephrology, Department of Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Elizabeth K Neumann
- Department of Chemistry, University of California, Davis, Davis, CA, 95695, USA
| | - Christopher R Anderton
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
9
|
Šolinc G, Srnko M, Merzel F, Crnković A, Kozorog M, Podobnik M, Anderluh G. Cryo-EM structures of a protein pore reveal a cluster of cholesterol molecules and diverse roles of membrane lipids. Nat Commun 2025; 16:2972. [PMID: 40140423 PMCID: PMC11947440 DOI: 10.1038/s41467-025-58334-z] [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: 07/04/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The structure and function of membrane proteins depend on their interactions with lipids that constitute membranes. Actinoporins are α-pore-forming proteins that bind preferentially to sphingomyelin-containing membranes, where they oligomerize and form transmembrane pores. Through a comprehensive cryo-electron microscopic analysis of a pore formed by an actinoporin Fav from the coral Orbicella faveolata, we show that the octameric pore interacts with 112 lipids in the upper leaflet of the membrane, reveal the roles of lipids, and demonstrate that the actinoporin surface is suited for binding multiple receptor sphingomyelin molecules. When cholesterol is present in the membrane, it forms a cluster of four molecules associated with each protomer. Atomistic simulations support the structural data and reveal additional effects of the pore on the lipid membrane. These data reveal a complex network of protein-lipid and lipid-lipid interactions and an underrated role of lipids in the structure and function of transmembrane protein complexes.
Collapse
Affiliation(s)
- Gašper Šolinc
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marija Srnko
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Franci Merzel
- Theory Department, National Institute of Chemistry, Ljubljana, Slovenia
| | - Ana Crnković
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Mirijam Kozorog
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| |
Collapse
|
10
|
Kontiza A, von Gerichten J, Spick M, Fraser E, Costa C, Saunders KDG, Whetton AD, Newman CF, Bailey MJ. Single-cell lipidomics: protocol development for reliable cellular profiling using capillary sampling. Analyst 2025; 150:1261-1270. [PMID: 40052368 PMCID: PMC11886952 DOI: 10.1039/d5an00037h] [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: 01/10/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
Single-cell lipidomics enables detailed analysis of the lipidomes of cells, but is challenged by small sample volumes, the risk of background interference and a lack of validation data. In this study, we explore the effect of different sampling variables on the lipid profiles of single pancreatic cancer cells, detected using liquid chromatography-mass spectrometry (LC-MS). We use automated and manual capillary sampling methods to isolate living single cells and evaluate different sampling media, capillary tips, aspiration volume, and temperature and humidity control. We demonstrate that automated and manual capillary sampling yield comparable lipid profiles when key parameters are controlled. Our findings highlight that appropriate blank correction, capillary tip type, and the control of aspiration volumes are all critical to preserving detection sensitivity. Conversely, choice of sampling medium does not affect lipidomics results. We also set out suggested best practices for these methodological variables, laying a foundation for robust, adaptable workflows in single-cell lipidomics for applications such as biomarker discovery and metabolic research.
Collapse
Affiliation(s)
- Anastasia Kontiza
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Surrey, GU2 7XH, Guildford, UK.
| | - Johanna von Gerichten
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Surrey, GU2 7XH, Guildford, UK.
| | - Matt Spick
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, GU2 7XH, Guildford, UK
| | - Emily Fraser
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Surrey, GU2 7XH, Guildford, UK.
| | - Catia Costa
- School of Computer Science and Electronic Engineering, Faculty of Engineering and Physical Sciences, University of Surrey, GU2 7XH, Guildford, UK
| | - Kyle D G Saunders
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Surrey, GU2 7XH, Guildford, UK.
| | - Anthony D Whetton
- vHive, School of Veterinary Medicine, School of Biosciences and Medicine, University of Surrey, Guildford, GU2 7XH, UK
| | - Carla F Newman
- GlaxoSmithKline, Cellular Imaging and Dynamics, Stevenage, SG1 2NY, UK
| | - Melanie J Bailey
- School of Chemistry and Chemical Engineering, Faculty of Engineering and Physical Sciences, University of Surrey, GU2 7XH, Guildford, UK.
| |
Collapse
|
11
|
Barkdull AP, Holcomb M, Forli S. A quantitative analysis of ligand binding at the protein-lipid bilayer interface. Commun Chem 2025; 8:89. [PMID: 40121339 PMCID: PMC11929912 DOI: 10.1038/s42004-025-01472-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
The majority of drugs target membrane proteins, and many of these proteins contain ligand binding sites embedded within the lipid bilayer. However, targeting these therapeutically relevant sites is hindered by limited characterization of both the sites and the molecules that bind to them. Here, we introduce the Lipid-Interacting LigAnd Complexes Database (LILAC-DB), a curated dataset of 413 structures of ligands bound at the protein-bilayer interface. Analysis of these structures reveals that ligands binding to lipid-exposed sites exhibit distinct chemical properties, such as higher calculated partition coefficient (clogP), molecular weight, and a greater number of halogen atoms, compared to ligands that bind to soluble proteins. Additionally, we demonstrate that the atomic properties of these ligands vary significantly depending on their depth within and exposure to the lipid bilayer. We also find that ligand binding sites exposed to the bilayer have distinct amino acid compositions compared to other protein regions, which may aid in the identification of lipid-exposed binding sites. This analysis provides valuable guidelines for researchers pursuing structure-based drug discovery targeting underexploited ligand binding sites at the protein-lipid bilayer interface.
Collapse
Affiliation(s)
- Allison Pearl Barkdull
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Matthew Holcomb
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
12
|
Yu F, Fei J, Jia Y, Wang T, Martin WF, Li J. Chemiosmotic ATP synthesis by minimal protocells. CELL REPORTS. PHYSICAL SCIENCE 2025; 6:102461. [PMID: 40123866 PMCID: PMC11922820 DOI: 10.1016/j.xcrp.2025.102461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 03/25/2025]
Abstract
Energy conservation is crucial to life's origin and evolution. The common ancestor of all cells used ATP synthase to convert proton gradients into ATP. However, pumps generating proton gradients and lipids maintaining proton gradients are not universally conserved across all lineages. A solution to this paradox is that ancestral ATP synthase could harness naturally formed geochemical ion gradients with simpler environmentally provided precursors preceding both proton pumps and biogenic membranes. This runs counter to traditional views that phospholipid bilayers are required to maintain proton gradients. Here, we show that fatty acid membranes can maintain sufficient proton gradients to synthesize ATP by ATP synthase under the steep pH and temperature gradients observed in hydrothermal vent systems. These findings shed substantial light on early membrane bioenergetics, uncovering a functional intermediate in the evolution of chemiosmotic ATP synthesis during protocellular stages postdating the ATP synthase's origin but preceding the advent of enzymatically synthesized cell membranes.
Collapse
Affiliation(s)
- Fanchen Yu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinbo Fei
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Jia
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
| | - Tonghui Wang
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - William F. Martin
- Institute of Molecular Evolution, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Bang S, Park B, Park JC, Jin H, Shim JS, Koo J, Lee KH, Shim MK, Kim H. Exosome-Inspired Lipid Nanoparticles for Enhanced Tissue Penetration. ACS NANO 2025; 19:8882-8894. [PMID: 40017353 DOI: 10.1021/acsnano.4c16629] [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: 03/01/2025]
Abstract
The extracellular matrix (ECM) is a complex network of biomolecules with varying pore sizes, posing a challenge for the effective penetration of lipid nanoparticles. In contrast, cell-derived lipid nanoparticles, such as exosomes, have demonstrated the ability to travel to distant organs, indicating their capacity to penetrate the ECM. Here, we designed exosome-like vesicles (ELVs) inspired by exosomes' distinct transport phenomena. Specifically, we integrated three exosomal components (anionic lipid, cholesterol, and aquaporin-1) associated with transport into our ELVs to mimic the superior diffusion behavior of exosomes over synthetic lipid nanoparticles. Surprisingly, both bulk- and single-particle-diffusion studies revealed a more than 33 times increase in the effective diffusion coefficient within model ECM compared to conventional lipid nanoparticles. Furthermore, ELVs show an 80% increase in the effective diffusion coefficient within biological tissues. The excellent transport behavior of ELVs was further validated in vivo, where intratumoral injection showcased their superior transport. These findings provide insights into lipid nanoparticle design for improved tissue penetration.
Collapse
Affiliation(s)
- Seunghwan Bang
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Byeongmin Park
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jae Chul Park
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Harin Jin
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Ji Sung Shim
- Department of Urology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Jahyun Koo
- School of Biomedical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Kwan Hyi Lee
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Man Kyu Shim
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hojun Kim
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
14
|
Karedla N, Schneider F, Enderlein J, Chen T. Leaflet-Specific Structure and Dynamics of Solid and Polymer Supported Lipid Bilayers. Angew Chem Int Ed Engl 2025:e202423784. [PMID: 40059717 DOI: 10.1002/anie.202423784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 04/08/2025]
Abstract
Polymer-supported or tethered lipid bilayers serve as versatile platforms for mimicking plasma membrane structure and dynamics, yet the impact of polymer supports on lipid bilayers remains largely unresolved. In this study, we introduce a novel methodology that combines graphene-induced energy transfer (GIET) with line-scan fluorescence lifetime correlation spectroscopy (lsFLCS) to examine the structural and dynamic properties of lipid bilayers. Our findings reveal that polymer supports markedly influence both the structural parameters, such as the membrane height from the substrate, its thickness, as well as dynamic properties, including leaflet-specific diffusion coefficients and interleaflet coupling. These findings highlight the complex interplay between a polymer support and the lipid bilayers. By resolving leaflet-specific diffusion and heights of the two leaflets from the substrate, this study emphasizes the potential of GIET-lsFLCS for probing membrane dynamics and structure. These insights significantly advance the understanding and application of polymer-supported membranes across diverse research contexts.
Collapse
Affiliation(s)
- Narain Karedla
- Third Institute of Physics - Biophysics, Georg August University, Friedrich-Hund-Platz 1, Göttingen, 37077, Germany
- The Rosalind Franklin Institute, Harwell Campus, Didcot, OX11 0FA, UK
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Falk Schneider
- Translational Imaging Center, University of Southern California, Los Angeles, CA, 90089, USA
- Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Jörg Enderlein
- Third Institute of Physics - Biophysics, Georg August University, Friedrich-Hund-Platz 1, Göttingen, 37077, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), Universitätsmedizin Göttingen, Robert-Koch-Str. 40, Göttingen, 37075, Germany
| | - Tao Chen
- Third Institute of Physics - Biophysics, Georg August University, Friedrich-Hund-Platz 1, Göttingen, 37077, Germany
| |
Collapse
|
15
|
Akhtar S, Zuhair F. Advancing Nanomedicine Through Electron Microscopy: Insights Into Nanoparticle Cellular Interactions and Biomedical Applications. Int J Nanomedicine 2025; 20:2847-2878. [PMID: 40078651 PMCID: PMC11899938 DOI: 10.2147/ijn.s500978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Nanomedicine has revolutionized cancer treatment by the development of nanoparticles (NPs) that offer targeted therapeutic delivery and reduced side effects. NPs research in nanomedicine significantly focuses on understanding their cellular interactions and intracellular mechanisms. A precise understanding of nanoparticle interactions at the subcellular level is crucial for their effective application in cancer therapy. Electron microscopy has proven essential, offering high-resolution insights into nanoparticle behavior within biological systems. This article reviews the role of electron microscopy in elucidating the cellular uptake and intracellular interactions of NPs. Transmission electron microscopy (TEM) provides imaging capabilities, such as cryo three-dimensional tomography, which offer in-depth insights into nanoparticle localization, endocytosis pathways, and subcellular interactions, while high resolution-TEM is primarily used for studying the atomic structure of isolated NPs rather than nanoparticles within cells or tissues. On the other hand, scanning electron microscopy (SEM) is ideal for examining larger surface areas and provides a broader perspective on the morphology and topography of the samples. The review highlights the advantages of electron microscopy in visualizing nanoparticle interactions with cellular structures and tracking their mechanisms of action. It also addresses the challenges associated with electron microscopy characterization, such as tedious sample preparation, static imaging limitations, and a restricted field of view. By examining various nanoparticle uptake pathways, and cellular destination of NPs with examples, the article emphasizes the importance of these pathways to optimize nanoparticle design and enhance therapeutic efficacy. This review underscores the need for continued advancement in electron microscopy techniques to improve the effectiveness of nanomedicine and address existing challenges. In summary, electron microscopy is a key tool for advancing our understanding of nanoparticle behavior in biological contexts, aiding in the design and optimization of nanomedicines by providing insights into nanoparticle cellular dynamics, uptake mechanisms, and therapeutic applications.
Collapse
Affiliation(s)
- Sultan Akhtar
- Department of Biophysics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Fatimah Zuhair
- Department of Infection Control, Alzahra General Hospital, Qatif, 31911, Saudi Arabia
| |
Collapse
|
16
|
Ramirez RX, Bosch AM, Pérez R, Guzman HV, Monje V. 2Danalysis: A toolbox for analysis of lipid membranes and biopolymers in two-dimensional space. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640563. [PMID: 40060461 PMCID: PMC11888481 DOI: 10.1101/2025.02.27.640563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Molecular simulations expand our ability to learn about the interplay of biomolecules. Biological membranes, composed of diverse lipids with varying physicochemical properties, are highly dynamic environments involved in cellular functions. Proteins, nucleic acids, glycans and bio-compatible polymers are the machinery of cellular processes both in the cytosol and at the lipid membrane interface. Lipid species directly modulate membrane properties, and affect the interaction and function of other biomolecules. Natural molecular diffusion results in changes of local lipid distribution, affecting the membrane properties. Projecting biophysical and structural membrane and biopolymer properties to a two-dimensional plane can be beneficial to quantify molecular signatures in a reduced dimensional space to identify relevant interactions at the interface of interest, i.e. the membrane surface or biopolymer-surface interface. Here, we present a toolbox designed to project membrane and biopolymer properties to a two-dimensional plane to characterize patterns of interaction and spatial correlations between lipid-lipid and lipid-biopolymer interfaces. The toolbox contains two hubs implemented using MDAKits architecture, one for membranes and one for biopolymers, that can be used independently or together. Three case studies demonstrate the versatility of the toolbox with detailed tutorials in GitHub. The toolbox and tutorials will be periodically updated with other functionalities and resolutions to expand our understanding of the structure-function relationship of biomolecules in two-dimensions.
Collapse
Affiliation(s)
- Ricardo X. Ramirez
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, 308 Furnas Hall, Buffalo, NY 14260, USA
| | - Antonio M. Bosch
- Departamento de Física Teórica de la Materia Condensada, Universidad Autónoma de Madrid, E-28049 Madrid, Spain
| | - Rubén Pérez
- Departamento de Física Teórica de la Materia Condensada, Universidad Autónoma de Madrid, E-28049 Madrid, Spain
- Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, E-28049 Madrid, Spain
| | - Horacio V. Guzman
- Departamento de Física Teórica de la Materia Condensada, Universidad Autónoma de Madrid, E-28049 Madrid, Spain
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), E-08193 Barcelona, Spain
| | - Viviana Monje
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, 308 Furnas Hall, Buffalo, NY 14260, USA
| |
Collapse
|
17
|
Yadav R, Gupta DP, Singh C. Lock and key: Quest to find the most compatible membrane mimetic for studying membrane proteins in native environment. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184414. [PMID: 39914557 DOI: 10.1016/j.bbamem.2025.184414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/16/2025] [Accepted: 02/01/2025] [Indexed: 02/12/2025]
Abstract
Membrane proteins play crucial roles in cellular signal transduction, molecule transport, host-pathogen interactions, and metabolic processes. However, mutations, changes in membrane properties, and environmental factors can lead to loss of protein function. This results in impaired ligand binding and misfolded structures that prevent proteins from adopting their native conformation. Many membrane proteins are also therapeutic targets in various diseases, where drugs can either restore or inhibit their specific functions. Understanding membrane protein structure and function is vital for advancing cell biology and physiology. Experimental studies often involve extracting proteins from their native environments and reconstituting them in membrane mimetics like detergents, bicelles, amphipols, nanodiscs, and liposomes. These mimetics replicate aspects of native membranes, aiding in the study of protein behavior outside living cells. Scientists continuously explore new, more native-like membrane mimetics to improve experimental accuracy. This dynamic field involves evaluating the advantages and disadvantages of different mimetics and optimizing the reconstitution process to better mimic natural conditions.
Collapse
Affiliation(s)
- Rahul Yadav
- Department of Biochemistry, Institute of Science, Banaras Hindu university (BHU), Varanasi Uttar Pradesh 221005, India
| | - Debarghya Pratim Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu university (BHU), Varanasi Uttar Pradesh 221005, India; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Chandan Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu university (BHU), Varanasi Uttar Pradesh 221005, India.
| |
Collapse
|
18
|
Romanova N, Sule K, Issler T, Hebrok D, Persicke M, Thévenod F, Prenner EJ, Lee WK. Cadmium-cardiolipin disruption of respirasome assembly and redox balance through mitochondrial membrane rigidification. J Lipid Res 2025; 66:100750. [PMID: 39880166 PMCID: PMC11905837 DOI: 10.1016/j.jlr.2025.100750] [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: 08/29/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
The environmental pollutant cadmium (Cd) poses a threat to human health through the consumption of contaminated foodstuffs culminating in chronic nephrotoxicity. Mitochondrial dysfunction and excessive reactive oxygen species (ROS) are key to Cd cellular toxicity. Cd-lipid interactions have been less considered. We hypothesized Cd binding to the inner mitochondrial membrane (IMM) phospholipid cardiolipin (CL) and membrane rigidification underlies defective electron transfer by disrupted respiratory supercomplexes (SCs). In Cd-treated rat kidney cortex (rKC) mitoplasts, laurdan (lipid-water interface), and diphenylhexatriene (hydrophobic core) revealed increased and decreased membrane fluidity, respectively. Laurdan-loaded pure CL or IMM biomimetic (40 mol % POPC, 35 mol % DOPE, 20 mol % TOCL, 5 mol % SAPI) nanoliposomes were rigidified by 25 μM Cd, which was confirmed in live-cell imaging of laurdan or di-4-ANEPPDHQ loaded human proximal convoluted tubule (HPCT) cells. Blue native gel electrophoresis evidenced ∼30% loss of I+III2+IVn SC formation after 5 μM Cd for 6 h in HPCTs, which was reversed by CL-binding drug MTP-131/SS-31/elamipretide (0.1 μM), yet α-tocopherol-insensitive. Moreover, MTP-131 attenuated Cd-induced H2O2 (∼30%) and cytochrome c release (∼25%), but not osmotic swelling, in rKC mitochondria as well as Cd-induced ROS (∼25%) in HPCTs. MTP-131 binding to IMM biomimetic nanoliposomes decreased zeta potential, prevented Cd-induced liposome size increase, and membrane rigidification reported by laurdan. Heterologous CRLS1 expression reversed Cd (5 μM, 24 h) cytotoxicity (∼25%) by MTT assay, Cd (5 μM, 3 h)-induced ROS and mitochondrial membrane rigidification by Cd (1 μM, 1 h) in HPCT cells. In summary, we report a novel mechanism for Cd toxicity in which Cd-CL interactions cause IMM rigidification, thereby disrupting correct SC assembly and increasing ROS.
Collapse
Affiliation(s)
- Nadiya Romanova
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Kevin Sule
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Travis Issler
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Daniel Hebrok
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Marcus Persicke
- Proteomics and Metabolomics Core Facility, Medical School OWL, Bielefeld University, Bielefeld, Germany; Microbial Genomic and Biotechnology, Center for Biotechnology, Bielefeld University, Bielefeld, Germany
| | - Frank Thévenod
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Bielefeld, Germany; Institute of Physiology and Pathophysiology, ZBAF, Witten/Herdecke University, Witten, Germany
| | - Elmar J Prenner
- Department of Biological Sciences, University of Calgary, Calgary, Canada.
| | - Wing-Kee Lee
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Bielefeld, Germany.
| |
Collapse
|
19
|
Hu L, Xiao S, Sun J, Wang F, Yin G, Xu W, Cheng J, Du G, Chen J, Kang Z. Regulating cellular metabolism and morphology to achieve high-yield synthesis of hyaluronan with controllable molecular weights. Nat Commun 2025; 16:2076. [PMID: 40021631 PMCID: PMC11871322 DOI: 10.1038/s41467-025-56950-3] [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: 08/15/2024] [Accepted: 02/06/2025] [Indexed: 03/03/2025] Open
Abstract
High-yield biosynthesis of hyaluronan (HA) with controllable molecular weights (MWs) remains challenging due to the poorly understood function of Class I HA synthase (HAS) and the metabolic imbalance between HA biosynthesis and cellular growth. Here, we systematically characterize HAS to identify crucial regions involved in HA polymerization, secretion, and MW control. We construct HAS mutants that achieve complete HA secretion and expand the MW range from 300 to 1400 kDa. By dynamically regulating UDP-glucose 6-dehydrogenase activity and applying an adaptive evolution approach, we recover cell normal growth with increased metabolic capacities. Final titers and productivities for high MW HA (500 kDa) and low MW HA (10 kDa) reach 45 g L-1 and 105 g L-1, 0.94 g L-1 h-1 and 1.46 g L-1 h-1, respectively. Our findings advance our understanding of HAS function and the interplay between cell metabolism and morphology, and provide a shape-guided engineering strategy to optimize microbial cell factories.
Collapse
Affiliation(s)
- Litao Hu
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- The Science Center for Future Foods, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China
- Institute of Future Food Technology, JITRI, Yixing, China
- College of Environment and Energy, South China University of Technology, Guangzhou, China
| | - Sen Xiao
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- The Science Center for Future Foods, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China
| | - Jieyu Sun
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- The Science Center for Future Foods, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China
| | - Faying Wang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- The Science Center for Future Foods, Jiangnan University, Wuxi, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China
| | - Guobin Yin
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- The Science Center for Future Foods, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China
| | - Wenjie Xu
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- The Science Center for Future Foods, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China
| | - Jianhua Cheng
- Institute of Future Food Technology, JITRI, Yixing, China
- College of Environment and Energy, South China University of Technology, Guangzhou, China
| | - Guocheng Du
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- The Science Center for Future Foods, Jiangnan University, Wuxi, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China
| | - Jian Chen
- The Science Center for Future Foods, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China
- Institute of Future Food Technology, JITRI, Yixing, China
| | - Zhen Kang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.
- The Science Center for Future Foods, Jiangnan University, Wuxi, China.
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China.
- Jiangsu Province Basic Research Center for Synthetic Biology, Jiangnan University, Wuxi, China.
- Institute of Future Food Technology, JITRI, Yixing, China.
| |
Collapse
|
20
|
Yang Q, Cai Y, Wang Z, Guo S, Qiu S, Zhang A. Understanding the physiological mechanisms and therapeutic targets of diseases: Lipidomics strategies. Life Sci 2025; 363:123411. [PMID: 39848598 DOI: 10.1016/j.lfs.2025.123411] [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: 11/25/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
As a pivotal branch of metabolomics, lipidomics studies global changes in lipid metabolism under different physiological and pathological conditions or drug interventions, discovers key lipid markers, and elaborates the associated lipid metabolism network. There are a considerable number of lipids in the host, which act on various functional networks such as metabolism and immune regulation. As an indispensable research method, lipidomics plays a key character in the analysis of lipid composition in organisms, the elaboration of the physiological mechanism of lipids, and the decoding of their character in the occurrence and development of diseases by exploring the character of lipids in the host environmental network. As an essential means of driving lipidomics research, High-throughput and High-resolution mass spectrometry is helpful in exploring disease phenotypic characteristics, diagnosing disease biomarkers, regulating related metabolic pathways, and discovering related active components. In this paper, we discuss the specific role of lipidomics in the analysis of disease diagnosis, prognosis and treatment, which is conducive to the realization of accurate and personalized medicine.
Collapse
Affiliation(s)
- Qiang Yang
- GAP Center, Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Cai
- GAP Center, Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China; International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China
| | - Zhibo Wang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China
| | - Sifan Guo
- GAP Center, Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shi Qiu
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China.
| | - Aihua Zhang
- GAP Center, Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China; International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China.
| |
Collapse
|
21
|
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
|
22
|
Sigg D, Carnevale V. Markov models and long-term memory in ion channels: A contradiction in terms? Biophys J 2025:S0006-3495(25)00063-3. [PMID: 39949059 DOI: 10.1016/j.bpj.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/07/2024] [Accepted: 02/06/2025] [Indexed: 02/28/2025] Open
Abstract
The opening kinetics of ion channels are typically modeled using Markov schemes, which assume a finite number of states linked by time-independent rate constants. Although aggregate closed or open states may, under the right conditions, experience short-term (exponential) memory of previous gating events, there is experimental evidence for stretched-exponential or power-law memory decay that does not conform to Markov theory. Here, using Monte Carlo simulations of a lattice system, we investigate long-term memory in channels coupled to a heterogeneous membrane near the critical temperature. We observed that increasing the strength of the channel-lipid coupling parameter from zero to nearly 1 kT per lipid binding site leads to a progression in the autocorrelation of successive open dwell times. This evolution changes from 1) multiexponential decay to 2) power-law decay, and finally to 3) stretched exponential decay, mirroring changes in channel distribution from: 1) complete independence, 2) partitioning in the interphase between lipid domains, and 3) partitioning inside the domain favorable to the activation state of the channel. The intermediate power-law regime demonstrates characteristics of long-term memory, such as trend-reinforcing values of the Hurst exponent. Still, this regime passes a previously proposed Markovianity test utilizing conditional dwell time histograms. We conclude that low-energy state-dependent interactions between ion channels and a dynamic membrane soften the Markov assumption by maintaining a fluctuating microenvironment and storing configurational memory, thus supporting the existence of long memory tails without necessarily diminishing the usefulness of Markov modeling.
Collapse
Affiliation(s)
- Daniel Sigg
- dPET, Spokane, Washington; Institute for Computational Molecular Science, Temple University, Philadelphia, Pennsylvania
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, Temple University, Philadelphia, Pennsylvania.
| |
Collapse
|
23
|
Lung SC, Chye ML. Mechanistic effects of lipid binding pockets within soluble signaling proteins: lessons from acyl-CoA-binding and START-domain-containing proteins. PLANT PHYSIOLOGY 2025; 197:kiae565. [PMID: 39431550 DOI: 10.1093/plphys/kiae565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
While lipids serve as important energy reserves, metabolites, and cellular constituents in all forms of life, these macromolecules also function as unique carriers of information in plant communication given their diverse chemical structures. The signal transduction process involves a sophisticated interplay between messengers, receptors, signal transducers, and downstream effectors. Over the years, an array of plant signaling proteins have been identified for their crucial roles in perceiving lipid signals. However, the mechanistic effects of lipid binding on protein functions remain largely elusive. Recent literature has presented numerous fascinating models that illustrate the significance of protein-lipid interactions in mediating signaling responses. This review focuses on the category of lipophilic signaling proteins that encompass a hydrophobic binding pocket located outside of cellular membranes and provides an update on the lessons learned from two of these structures, namely the acyl-CoA-binding and steroidogenic acute regulatory protein-related lipid transfer domains. It begins with a brief overview of the latest advances in understanding the functions of the two protein families in plant communication. The second part highlights five functional mechanisms of lipid ligands in concert with their target signaling proteins.
Collapse
Affiliation(s)
- Shiu-Cheung Lung
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Mee-Len Chye
- Department of Agricultural and Food Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, Kampar 31900, Malaysia
| |
Collapse
|
24
|
Brown C, Ghosh S, McAllister R, Kumar M, Walker G, Sun E, Aman T, Panda A, Kumar S, Li W, Coleman J, Liu Y, Rothman JE, Bhattacharyya M, Gupta K. A proteome-wide quantitative platform for nanoscale spatially resolved extraction of membrane proteins into native nanodiscs. Nat Methods 2025; 22:412-421. [PMID: 39609567 PMCID: PMC11810782 DOI: 10.1038/s41592-024-02517-x] [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: 03/01/2024] [Accepted: 10/16/2024] [Indexed: 11/30/2024]
Abstract
The native membrane environment profoundly influences every aspect of membrane protein (MP) biology. Despite this, the most prevalent method of studying MPs uses detergents to disrupt and remove this vital membrane context, impeding our ability to decipher the local molecular context and its effect. Here we develop a membrane proteome-wide platform that enables rapid spatially resolved extraction of target MPs directly from cellular membranes into native nanodiscs that maintain the local membrane context, using a library of membrane-active polymers. We accompany this with an open-access database that quantifies the polymer-specific extraction efficiency for 2,065 unique mammalian MPs and provides the most optimized extraction condition for each. To validate, we demonstrate how this resource can enable rapid extraction and purification of target MPs from different organellar membranes with high efficiency and purity. Further, we show how the database can be extended to capture overexpressed multiprotein complexes by taking two synaptic vesicle MPs. We expect these publicly available resources to empower researchers across disciplines to efficiently capture membrane 'nano-scoops' containing a target MP and interface with structural, functional and bioanalytical approaches.
Collapse
Affiliation(s)
- Caroline Brown
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD, USA
| | - Snehasish Ghosh
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD, USA
| | - Rachel McAllister
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Mukesh Kumar
- F.M. Kirby Neurobiology Center, Department of Neurobiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Cell Signaling Technology, Danvers, MA, USA
| | - Gerard Walker
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Eric Sun
- Nanobiology Institute, Yale University, West Haven, CT, USA
| | - Talat Aman
- Nanobiology Institute, Yale University, West Haven, CT, USA
| | - Aniruddha Panda
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Shailesh Kumar
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Wenxue Li
- Department of Pharmacology, Yale University, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Jeff Coleman
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Yansheng Liu
- Department of Pharmacology, Yale University, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - James E Rothman
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Kallol Gupta
- Nanobiology Institute, Yale University, West Haven, CT, USA.
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.
- Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
25
|
Sosa Ponce ML, Cobb JA, Zaremberg V. Lipids and chromatin: a tale of intriguing connections shaping genomic landscapes. Trends Cell Biol 2025; 35:141-152. [PMID: 39060139 DOI: 10.1016/j.tcb.2024.06.004] [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: 03/01/2024] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024]
Abstract
Recent studies in yeast reveal an intricate interplay between nuclear envelope (NE) architecture and lipid metabolism, and between lipid signaling and both epigenome and genome integrity. In this review, we highlight the reciprocal connection between lipids and histone modifications, which enable metabolic reprogramming in response to nutrients. The endoplasmic reticulum (ER)-NE regulates the compartmentalization and temporal availability of epigenetic metabolites and its lipid composition also impacts nuclear processes, such as transcriptional silencing and the DNA damage response (DDR). We also discuss recent work providing mechanistic insight into lipid droplet (LD) formation and sterols in the nucleus, and the collective data showing Opi1 as a central factor in both membrane sensing and transcriptional regulation of lipid-chromatin interrelated processes.
Collapse
Affiliation(s)
- Maria Laura Sosa Ponce
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Jennifer A Cobb
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Vanina Zaremberg
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
26
|
Bysack A, Jash C, Raghuraman H. Structural Dynamics of the Slide Helix of Inactive/Closed Conformation of KirBac1.1 in Micelles and Membranes: A Fluorescence Approach. J Membr Biol 2025; 258:97-112. [PMID: 39789244 PMCID: PMC11779782 DOI: 10.1007/s00232-024-00335-y] [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: 10/25/2024] [Accepted: 12/25/2024] [Indexed: 01/12/2025]
Abstract
Inward rectifying potassium (Kir) channels play a critical role in maintaining the resting membrane potential and cellular homeostasis. The high-resolution crystal structure of homotetrameric KirBac1.1 in detergent micelles provides a snapshot of the closed state. Similar to micelles, KirBac1.1 is reported to be in the inactive/closed conformation in POPC membranes. The slide helix of KirBac1.1 is an important structural motif that regulates channel gating. Despite the importance of slide helix in lipid-dependent gating, conflicting models have emerged for the location of slide helix and its structural dynamics in membrane mimetics is poorly understood. Here, we monitored the structural dynamics of the slide helix (residues 46-57) of KirBac1.1 in both DM micelles and POPC membranes utilizing various site-directed fluorescence approaches. We show, using ACMA-based liposome-flux assay, the cysteine mutants of the slide helix are not functional, ensuring the inactive/closed conformation in POPC membranes similar to wild-type channel. Time-resolved fluorescence and water accessibility measurements of NBD-labeled single-cysteine mutants of slide-helix residues suggest that the location of the slide helix at the interfacial region might be shallower in membranes compared to micelles. Interestingly, the slide helix of KirBac1.1 is more dynamic in the physiologically relevant membrane environment, which is accompanied by a differential hydration dynamics throughout the slide helix. Further, REES and lifetime distribution analyses suggest significant changes in conformational heterogeneity of the slide helix in membrane mimetics. Overall, our results give an insight into how membrane mimetics affect the organization and dynamics of slide helix of the closed state of KirBac1.1, and highlight the importance of lipid-protein interactions in membranes.
Collapse
Affiliation(s)
- Arpan Bysack
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Chandrima Jash
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - H Raghuraman
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India.
- Homi Bhabha National Institute, Training School Complex, Mumbai, India.
| |
Collapse
|
27
|
Wen P, Huang H, Zhang R, Zheng H, Liang T, Zhuang C, Wu Q, Wang J, Liu F, Zhang K, Wu W, He K, Liu F, Li H, Gu Z. Coacervate vesicles assembled by liquid-liquid phase separation improve delivery of biopharmaceuticals. Nat Chem 2025; 17:279-288. [PMID: 39806140 DOI: 10.1038/s41557-024-01705-8] [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: 03/18/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025]
Abstract
Vesicles play critical roles in cellular materials storage and signal transportation, even in the formation of organelles and cells. Natural vesicles are composed of a lipid layer that forms a membrane for the enclosure of substances inside. Here we report a coacervate vesicle formed by the liquid-liquid phase separation of cholesterol-modified DNA and histones. Unlike a phospholipid-based membrane-bounded vesicle, a coacervate vesicle lacks a membrane structure on the surface and is organized with a high-density liquid layer and a water-filled cavity. Through a straightforward coacervation process, we demonstrate that various biological agents, including virus particles, mRNA, cytokines and peptides, can be innocuously and directly enriched in the liquid phase. In contrast to the droplet-like coacervates that are prone to aggregation challenges, coacervate vesicles display superior kinetic stability, positioning them as a versatile delivery vehicle for biopharmaceuticals. We validate that incorporating oncolytic viruses into these coacervate vesicles endows them with potent oncolytic efficacy and elicits robust anti-tumour immune responses in mouse models.
Collapse
Affiliation(s)
- Ping Wen
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hanwei Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, China Medical University, Ministry of Education, Shenyang, China
- Phase I Clinical Trials Center, The First Hospital, China Medical University, Shenyang, China
| | - Ruizhe Zhang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, China Medical University, Ministry of Education, Shenyang, China
- Phase I Clinical Trials Center, The First Hospital, China Medical University, Shenyang, China
| | - Hanqi Zheng
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Tingxizi Liang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chuyue Zhuang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qing Wu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Junxia Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Feng Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ke Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wei Wu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kaixin He
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, China Medical University, Ministry of Education, Shenyang, China.
- Phase I Clinical Trials Center, The First Hospital, China Medical University, Shenyang, China.
| | - Hongjun Li
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Liangzhu Laboratory, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
28
|
Moore WM, Brea RJ, Knittel C, Wrightsman E, Hui B, Lou J, Ancajas CF, Best MD, Devaraj NK, Budin I. Leaflet specific phospholipid imaging using genetically encoded proximity sensors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.01.592120. [PMID: 38746395 PMCID: PMC11092643 DOI: 10.1101/2024.05.01.592120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The lipid composition of cells varies widely across organelles and between individual membrane leaflets. Transport proteins acting within and between membranes are thought to generate this heterogeneity, but measuring their functions has been hampered by limited tools for imaging lipid composition at relevant spatial resolutions. Here we present fluorogen-activating coincidence sensing (FACES), a chemogenetic tool capable of quantitatively imaging subcellular lipid pools and reporting their transbilayer orientation in living cells. FACES combines bioorthogonal chemistry with genetically encoded fluorogen-activating proteins (FAPs) for reversible proximity sensing of conjugated molecules. We first apply this approach to identify roles for lipid transfer proteins (LTP) that traffic phosphatidylcholine pools between the ER and mitochondria. We then show that transmembrane domain-containing FAPs can be used to reveal the membrane asymmetry of multiple lipid classes that is generated at the trans-Golgi network (TGN). Using FACES, we show how the asymmetry of a phosphatidylserine analogue at the TGN is supported by the activity of both cytosolic LTPs and transmembrane flippases. Lastly, we demonstrate that FACES is a generalizable tool for subcellular detection of other molecule classes by measuring changes in mitochondrial N -acetylhexosamine levels. These results introduce the application of fluorogenic tags for spatially-defined molecular imaging.
Collapse
|
29
|
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
|
30
|
Szabo M, Cs. Szabo B, Kurtan K, Varga Z, Panyi G, Nagy P, Zakany F, Kovacs T. Look Beyond Plasma Membrane Biophysics: Revealing Considerable Variability of the Dipole Potential Between Plasma and Organelle Membranes of Living Cells. Int J Mol Sci 2025; 26:889. [PMID: 39940660 PMCID: PMC11816637 DOI: 10.3390/ijms26030889] [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: 12/14/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Due to the lack of measurement techniques suitable for examining compartments of intact, living cells, membrane biophysics is almost exclusively investigated in the plasma membrane despite the fact that its alterations in intracellular organelles may also contribute to disease pathogenesis. Here, we employ a novel, easy-to-use, confocal microscopy-based approach utilizing F66, an environment-sensitive fluorophore in combination with fluorescent organelle markers and quantitative image analysis to determine the magnitude of the molecular order-related dipole potential in the plasma membrane and intracellular organelles of various tumor and neural cell lines. Our comparative analysis demonstrates considerable intracellular variations of the dipole potential that may be large enough to modulate protein functions, with an inward decreasing gradient on the route of the secretory/endocytic pathway (plasma membrane >> lysosome > Golgi > endoplasmic reticulum), whereas mitochondrial membranes are characterized by a dipole potential slightly larger than that of lysosomes. Our approach is suitable and sensitive enough to quantify membrane biophysical properties selectively in intracellular compartments and their comparative analysis in intact, living cells, and, therefore, to identify the affected organelles and potential therapeutic targets in diseases associated with alterations in membrane lipid composition and thus biophysics such as tumors, metabolic, neurodegenerative, or lysosomal storage disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Florina Zakany
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.S.); (B.C.S.); (K.K.); (Z.V.); (G.P.); (P.N.)
| | - Tamas Kovacs
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.S.); (B.C.S.); (K.K.); (Z.V.); (G.P.); (P.N.)
| |
Collapse
|
31
|
Peleg S, Meron S, Shenberger Y, Hofmann L, Gevorkyan-Airapetov L, Ruthstein S. Exploring the Gating Mechanism of the Human Copper Transporter, hCtr1, Using EPR Spectroscopy. Biomolecules 2025; 15:127. [PMID: 39858521 PMCID: PMC11763644 DOI: 10.3390/biom15010127] [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: 11/12/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Ctr1 is a membrane-spanning homotrimer that facilitates copper uptake in eukaryotic cells with high affinity. While structural details of the transmembrane domain of human Ctr1 have been elucidated using X-ray crystallography and cryo-EM, the transfer mechanisms of copper and the conformational changes that control the gating mechanism remain poorly understood. The role of the extracellular N-terminal domains is particularly unclear due to the absence of a high-resolution structure of the full-length hCtr1 protein and limited biochemical and biophysical characterization of the transporter in solution and in cell. In this study, we employed distance electron paramagnetic resonance to investigate the conformational changes of the extracellular N-terminal domain of full-length hCtr1, both in vitro and in cells, as a function of Cu(I) binding. Our results demonstrate that at specific Cu(I) concentrations, the extracellular chains move closer to the lumen to facilitate copper transfer. Additionally, while at these concentrations the intracellular part is penetrating the lumen, suggesting a ball-and-chain gating mechanism. Moreover, this phenomenon was observed for both reconstituted protein in micelles and in native cell membranes. However, the measured distance values were slightly different, suggesting that the membrane's characteristics and therefore its lipid composition also impact and even regulate the gating mechanism of hCtr1.
Collapse
Affiliation(s)
| | | | | | | | | | - Sharon Ruthstein
- Department of Chemistry and Institute of Nanotechnology and Advanced Materials, Faculty of Exact Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel; (S.P.); (S.M.); (Y.S.); (L.H.); (L.G.-A.)
| |
Collapse
|
32
|
Kuyler G, Barnard E, Sridhar P, Murray RJ, Pollock NL, Wheatley M, Dafforn TR, Klumperman B. Tunable Terpolymer Series for the Systematic Investigation of Membrane Proteins. Biomacromolecules 2025; 26:415-427. [PMID: 39725644 PMCID: PMC11733950 DOI: 10.1021/acs.biomac.4c01219] [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: 09/05/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Membrane proteins (MPs) are critical to cellular processes and serve as essential therapeutic targets. However, their isolation and characterization are often impeded by traditional detergent-based methods, which can compromise their native states, and retention of their native lipid environment. Amphiphilic polymers have emerged as effective alternatives, enabling the formation of nanoscale discs that preserve MPs' structural and functional integrity. We introduce a novel series of poly(styrene-co-maleic acid-co-(N-benzyl)maleimide) (BzAM) terpolymers with tunable amphiphilicity, synthesized through controlled polymerization. Designed to mimic and improve upon industry-standard poly(styrene-co-maleic acid), these well-defined terpolymers offer enhanced control over molecular weight and distribution, allowing for systematic evaluation of polymer properties and their effect on membrane solubilization. The BzAM series effectively solubilized membranes and demonstrated a direct correlation between polymer hydrophobicity and solubilization efficiency of bacterial ABC transporter, Sav1866. This research highlights the importance of rational polymer design in MP research and provides a foundation for future developments.
Collapse
Affiliation(s)
- Gestél
C. Kuyler
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Private Bag X1, Matieland 7602, South Africa
- Centre for
Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
| | - Elaine Barnard
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Private Bag X1, Matieland 7602, South Africa
| | - Pooja Sridhar
- School of
Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Rebecca J. Murray
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Private Bag X1, Matieland 7602, South Africa
- Centre for
Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
| | - Naomi L. Pollock
- School of
Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Mark Wheatley
- Centre for
Health and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
- Centre of
Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands B15 2TT, United Kingdom
| | - Timothy R. Dafforn
- School of
Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Bert Klumperman
- Department
of Chemistry and Polymer Science, Stellenbosch
University, Private Bag X1, Matieland 7602, South Africa
| |
Collapse
|
33
|
Batool I, Ayyaz A, Qin T, Wu X, Chen W, Hannan F, Zafar ZU, Naeem MS, Farooq MA, Zhou W. Morphological, Physiological, and Molecular Responses to Heat Stress in Brassicaceae. PLANTS (BASEL, SWITZERLAND) 2025; 14:152. [PMID: 39861509 PMCID: PMC11768255 DOI: 10.3390/plants14020152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/24/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025]
Abstract
Food security is threatened by global warming, which also affects agricultural output. Various components of cells perceive elevated temperatures. Different signaling pathways in plants distinguish between the two types of temperature increases, mild warm temperatures and extremely hot temperatures. Given the rising global temperatures, heat stress has become a major abiotic challenge, affecting the growth and development of various crops and significantly reducing productivity. Brassica napus, the second-largest source of vegetable oil worldwide, faces drastic reductions in seed yield and quality under heat stress. This review summarizes recent research on the genetic and physiological impact of heat stress in the Brassicaceae family, as well as in model plants Arabidopsis and rice. Several studies show that extreme temperature fluctuations during crucial growth stages negatively affect plants, leading to impaired growth and reduced seed production. The review discusses the mechanisms of heat stress adaptation and the key regulatory genes involved. It also explores the emerging understanding of epigenetic modifications during heat stress. While such studies are limited in B. napus, contrasting trends in gene expression have been observed across different species and cultivars, suggesting these genes play a complex role in heat stress tolerance. Key knowledge gaps are identified regarding the impact of heat stress during the growth stages of B. napus. In-depth studies of these stages are still needed. The profound understanding of heat stress response mechanisms in tissue-specific models are crucial in advancing our knowledge of thermo-tolerance regulation in B. napus and supporting future breeding efforts for heat-tolerant crops.
Collapse
Affiliation(s)
- Iram Batool
- Institute of Crop Science, Ministry of Agriculture and Rural Affairs Key Laboratory of Spectroscopy Sensing, Zhejiang University, Hangzhou 310058, China; (I.B.); (A.A.); (T.Q.); (X.W.); (W.C.); (F.H.)
| | - Ahsan Ayyaz
- Institute of Crop Science, Ministry of Agriculture and Rural Affairs Key Laboratory of Spectroscopy Sensing, Zhejiang University, Hangzhou 310058, China; (I.B.); (A.A.); (T.Q.); (X.W.); (W.C.); (F.H.)
| | - Tongjun Qin
- Institute of Crop Science, Ministry of Agriculture and Rural Affairs Key Laboratory of Spectroscopy Sensing, Zhejiang University, Hangzhou 310058, China; (I.B.); (A.A.); (T.Q.); (X.W.); (W.C.); (F.H.)
| | - Xiaofen Wu
- Institute of Crop Science, Ministry of Agriculture and Rural Affairs Key Laboratory of Spectroscopy Sensing, Zhejiang University, Hangzhou 310058, China; (I.B.); (A.A.); (T.Q.); (X.W.); (W.C.); (F.H.)
| | - Weiqi Chen
- Institute of Crop Science, Ministry of Agriculture and Rural Affairs Key Laboratory of Spectroscopy Sensing, Zhejiang University, Hangzhou 310058, China; (I.B.); (A.A.); (T.Q.); (X.W.); (W.C.); (F.H.)
| | - Fakhir Hannan
- Institute of Crop Science, Ministry of Agriculture and Rural Affairs Key Laboratory of Spectroscopy Sensing, Zhejiang University, Hangzhou 310058, China; (I.B.); (A.A.); (T.Q.); (X.W.); (W.C.); (F.H.)
| | - Zafar Ullah Zafar
- Institute of Botany, Bahauddin Zakariya University, Multan 40162, Pakistan;
| | - Muhammad Shahbaz Naeem
- Department of Agronomy, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan;
| | - Muhammad Ahsan Farooq
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China;
| | - Weijun Zhou
- Institute of Crop Science, Ministry of Agriculture and Rural Affairs Key Laboratory of Spectroscopy Sensing, Zhejiang University, Hangzhou 310058, China; (I.B.); (A.A.); (T.Q.); (X.W.); (W.C.); (F.H.)
| |
Collapse
|
34
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [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: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
35
|
Chan SJW, Zhu JY, Yip BRP, Shyamasundar S, Bazan GC. Molecular Design of a Diketopyrrolopyrrole Conjugated Oligoelectrolyte Capable of Imaging Intracellular Vesicle Membrane Dynamics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411329. [PMID: 39604323 DOI: 10.1002/adma.202411329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/05/2024] [Indexed: 11/29/2024]
Abstract
Conjugated oligoelectrolytes (COEs) are lipid bilayer spanning optical reporters that hold promise for delineating spatiotemporal changes in subcellular compartments. However, their ability to probe a broader range of biological processes remains restricted due to the lack of environmentally-responsive chemical functionalities. Herein, the study reports a novel COE, namely COE-KP, for monitoring spatiotemporal changes in the endolysosomal vesicles. COE-KP features a central diketopyrrolopyrrole functional group in the optically active conjugated core that confers photophysical properties suitable for bioimaging, in particular responding to the presence of hydrogen bonding functionalities within the hydrophobic domain of the lipid bilayer. COE-KP can thus discriminate cells in different growth states through two-photon fluorescence lifetime imaging microscopy (FLIM) with excitation in the NIR-II range. These changes in lifetime most reasonably reflect the degree of water permeability through the membrane and are not linked to notable differences in membrane tension or solvent polarity. Furthermore, using stimulated emission depletion (STED) nanoscopy, it is possible to directly visualize membrane-bound COEs within cellular vesicles. These results illustrate further opportunities for applying COE-based reporters for super-resolution microscopy of organelle membranes, visualization of subcellular membrane morphologies, and imaging long-term changes in membrane properties.
Collapse
Affiliation(s)
- Samuel J W Chan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Ji-Yu Zhu
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Benjamin Rui Peng Yip
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Sukanya Shyamasundar
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - Guillermo C Bazan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore, 636921, Singapore
| |
Collapse
|
36
|
López-Tubau JM, Laibach N, Burciaga-Monge A, Alseekh S, Deng C, Fernie AR, Altabella T, Ferrer A. Differential impact of impaired steryl ester biosynthesis on the metabolome of tomato fruits and seeds. PHYSIOLOGIA PLANTARUM 2025; 177:e70022. [PMID: 39710490 DOI: 10.1111/ppl.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Steryl esters (SE) are a storage pool of sterols that accumulates in cytoplasmic lipid droplets and helps to maintain plasma membrane sterol homeostasis throughout plant growth and development. Ester formation in plant SE is catalyzed by phospholipid:sterol acyltransferase (PSAT) and acyl-CoA:sterol acyltransferase (ASAT), which transfer long-chain fatty acid groups to free sterols from phospholipids and acyl-CoA, respectively. Comparative mass spectrometry-based metabolomic analysis between ripe fruits and seeds of a tomato (Solanum lycopersicum cv Micro-Tom) mutant lacking functional PSAT and ASAT enzymes (slasat1xslpsat1) shows that disruption of SE biosynthesis has a differential impact on the metabolome of these organs, including changes in the composition of free and glycosylated sterols. Significant perturbations were observed in the fruit lipidome in contrast to the mild effect detected in the lipidome of seeds. A contrasting response was also observed in phenylpropanoid metabolism, which is down-regulated in fruits and appears to be stimulated in seeds. Comparison of global metabolic changes using volcano plot analysis suggests that disruption of SE biosynthesis favours a general state of metabolic activation that is more evident in seeds than fruits. Interestingly, there is an induction of autophagy in both tissues, which may contribute along with other metabolic changes to the phenotypes of early seed germination and enhanced fruit tolerance to Botrytis cinerea displayed by the slasat1xslpsat1 mutant. The results of this study reveal unreported connections between SE metabolism and the metabolic status of plant cells and lay the basis for further studies aimed at elucidating the mechanisms underlying the observed effects.
Collapse
Affiliation(s)
- Joan Manel López-Tubau
- Plant Synthetic Biology and Metabolic Engineering Program, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Cerdanyola, Barcelona, Spain
| | - Natalie Laibach
- Plant Synthetic Biology and Metabolic Engineering Program, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Cerdanyola, Barcelona, Spain
- Hochshule Rhein-Waal. Faculty of Life Sciences, Rhine-Waal University of Applied Sciences, Kleve, Germany
| | - Alma Burciaga-Monge
- Plant Synthetic Biology and Metabolic Engineering Program, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Cerdanyola, Barcelona, Spain
| | - Saleh Alseekh
- Max-Planck-Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| | - Cuiyun Deng
- Plant Synthetic Biology and Metabolic Engineering Program, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Cerdanyola, Barcelona, Spain
| | - Alisdair R Fernie
- Max-Planck-Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Teresa Altabella
- Plant Synthetic Biology and Metabolic Engineering Program, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Cerdanyola, Barcelona, Spain
- Department of Biology, Healthcare and the Environment, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Albert Ferrer
- Plant Synthetic Biology and Metabolic Engineering Program, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Cerdanyola, Barcelona, Spain
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
37
|
Wu D, Tang H, Qiu X, Song S, Chen S, Robinson CV. Native MS-guided lipidomics to define endogenous lipid microenvironments of eukaryotic receptors and transporters. Nat Protoc 2025; 20:1-25. [PMID: 39174660 DOI: 10.1038/s41596-024-01037-4] [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: 03/13/2024] [Accepted: 06/06/2024] [Indexed: 08/24/2024]
Abstract
The mammalian membrane is composed of various eukaryotic lipids interacting with extensively post-translationally modified proteins. Probing interactions between these mammalian membrane proteins and their diverse and heterogeneous lipid cohort remains challenging. Recently, native mass spectrometry (MS) combined with bottom-up 'omics' approaches has provided valuable information to relate structural and functional lipids to membrane protein assemblies in eukaryotic membranes. Here we provide a step-by-step protocol to identify and provide relative quantification for endogenous lipids bound to mammalian membrane proteins and their complexes. Using native MS to guide our lipidomics strategies, we describe the necessary sample preparation steps, followed by native MS data acquisition, tailored lipidomics and data interpretation. We also highlight considerations for the integration of different levels of information from native MS and lipidomics and how to deal with the various challenges that arise during the experiments. This protocol begins with the preparation of membrane proteins from mammalian cells and tissues for native MS. The results enable not only direct assessment of copurified endogenous lipids but also determination of the apparent affinities of specific lipids. Detailed sample preparation for lipidomics analysis is also covered, along with comprehensive settings for liquid chromatography-MS analysis. This protocol is suitable for the identification and quantification of endogenous lipids, including fatty acids, sterols, glycerolipids, phospholipids and glycolipids and can be used to interrogate proteins from recombinant sources to native membranes.
Collapse
Affiliation(s)
- Di Wu
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Haiping Tang
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Xingyu Qiu
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Siyuan Song
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Siyun Chen
- Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Carol V Robinson
- Department of Chemistry, University of Oxford, Oxford, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Xu G, Yu J, Liu S, Cai L, Han XX. In situ surface-enhanced Raman spectroscopy for membrane protein analysis and sensing. Biosens Bioelectron 2025; 267:116819. [PMID: 39362137 DOI: 10.1016/j.bios.2024.116819] [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: 04/28/2024] [Revised: 09/08/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Membrane proteins are involved in a variety of dynamic cellular processes and exploration of the structural basis of membrane proteins is of significance for a better understanding of their functions. In situ analysis of membrane proteins and their dynamics is, however, challenging for conventional techniques. Surface-enhanced Raman spectroscopy (SERS) is powerful in protein structural characterization, allowing for sensitive, in-situ and real-time identification and dynamic monitoring under physiological conditions. In this review, the applications of SERS in probing membrane proteins are outlined, discussed and prospected. It starts with a brief introduction to membrane proteins, SERS theories and SERS-based strategies that commonly-used for membrane proteins. How to assemble phospholipid biolayers on SERS-active materials is highlighted, followed by respectively discussing about direct and indirect strategies for membrane protein sensing. SERS-based monitoring of protein-ligand interactions is finally introduced and its potential in biomedical applications is discussed in detail. The review ends with critical discussion about current challenges and limitations of this research field, and the promising perspectives in both fundamental and applied sciences.
Collapse
Affiliation(s)
- Guangyang Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Jiaheng Yu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Shiyi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Linjun Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, 130012, PR China
| | - Xiao Xia Han
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| |
Collapse
|
39
|
Cheng D, Guo Y, Lyu J, Liu Y, Xu W, Zheng W, Wang Y, Qiao P. Advances and challenges in preparing membrane proteins for native mass spectrometry. Biotechnol Adv 2025; 78:108483. [PMID: 39571766 DOI: 10.1016/j.biotechadv.2024.108483] [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: 05/31/2024] [Revised: 10/07/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Native mass spectrometry (nMS) is becoming a crucial tool for analyzing membrane proteins (MPs), yet challenges remain in solubilizing and stabilizing their native conformations while resolving and characterizing the heterogeneity introduced by post-translational modifications and ligand binding. This review highlights recent advancements and persistent challenges in preparing MPs for nMS. Optimizing detergents and additives can significantly reduce sample heterogeneity and surface charge, enhancing MP signal quality and structural preservation in nMS. A strategic workflow incorporating affinity capture, stabilization agents, and size-exclusion chromatography to remove unfolded species demonstrates success in improving nMS characterization. Continued development of customized detergents and reagents tailored for specific MPs may further minimize heterogeneity and boost signals. Instrumental advances are also needed to elucidate more dynamically complex and labile MPs. Effective sample preparation workflows may provide insights into MP structures, dynamics, and interactions underpinning membrane biology. With ongoing methodological innovation, nMS shows promise to complement biophysical studies and facilitate drug discovery targeting this clinically important yet technically demanding protein class.
Collapse
Affiliation(s)
- Di Cheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yi Guo
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Jixing Lyu
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Yang Liu
- Regenxbox In., Rockville, MD 20850, USA
| | - Wenhao Xu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Weiyi Zheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yuchen Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Pei Qiao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
40
|
Zhu J, Cao Y, Qin X, Liang Q. Budding of Asymmetric Lipid Bilayers: Effects of Cholesterol, Anionic Lipid, and Electric Field. J Phys Chem B 2024; 128:12741-12751. [PMID: 39670354 DOI: 10.1021/acs.jpcb.4c07782] [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
Membrane budding is vital for various cellular processes such as synaptic activity regulation, vesicle transport and release, and endocytosis/exocytosis. Although protein-mediated membrane budding has been extensively investigated, the effects of the lipid asymmetry of the two leaflets and the asymmetrically electrical environments of the cellular membrane on membrane budding remain elusive. In this work, using coarse-grained molecular dynamics simulations, we systematically investigate the impacts of lipid bilayer asymmetry and external electric fields mimicking the asymmetric membrane potential on the membrane budding. The results show that the differential stress induced by the asymmetric distribution of lipids in the two leaflets is a crucial factor for the membrane budding. The unidirectional flip of cholesterol induced by the membrane curvature and the asymmetric ion adsorption induced by the anionic lipids promote the budding process. Furthermore, the external electric field applied perpendicularly to the bilayer plane increases the transmembrane potential and produces an additional differential stress across the leaflets by imposing an asymmetric torque on the lipid headgroups in the two leaflets, facilitating the membrane budding. These findings offer insights into how the structural and the environmental asymmetry in natural cellular membranes influence membrane budding in cellular processes.
Collapse
Affiliation(s)
- Jin Zhu
- Center for Statistical and Theoretical Condensed Matter Physics & Department of Physics, Zhejiang Normal University, Jinhua 321004, China
- Zhejiang Institute of Photoelectronics & Zhejiang Institute for Advanced Light Source, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Yu Cao
- Center for Statistical and Theoretical Condensed Matter Physics & Department of Physics, Zhejiang Normal University, Jinhua 321004, China
- Zhejiang Institute of Photoelectronics & Zhejiang Institute for Advanced Light Source, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Xiaoxue Qin
- Center for Statistical and Theoretical Condensed Matter Physics & Department of Physics, Zhejiang Normal University, Jinhua 321004, China
- Zhejiang Institute of Photoelectronics & Zhejiang Institute for Advanced Light Source, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Qing Liang
- Center for Statistical and Theoretical Condensed Matter Physics & Department of Physics, Zhejiang Normal University, Jinhua 321004, China
- Zhejiang Institute of Photoelectronics & Zhejiang Institute for Advanced Light Source, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| |
Collapse
|
41
|
Nemoto K. Applications of the wheat germ cell-free protein synthesis system in plant biochemical studies. PLANT BIOTECHNOLOGY (TOKYO, JAPAN) 2024; 41:325-334. [PMID: 40083572 PMCID: PMC11897732 DOI: 10.5511/plantbiotechnology.24.0501a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/01/2024] [Indexed: 03/16/2025]
Abstract
The development of cell-free protein synthesis technology has made it possible to easily and quickly synthesize recombinant proteins. Among cell-free protein synthesis systems, wheat germ cell-free protein synthesis using eukaryotic ribosomes is an efficient approach to synthesize proteins with diverse and complex structures and functions. However, to date, cell-free protein synthesis systems, including wheat germ cell-free systems, have not been widely used in plant research, and little is known about their applications. Here, I first introduce a basic overview of the cell-free protein synthesis system of wheat germ. Next, I will focus on our previous research examples on plants and present the applications in which the wheat germ cell-free system is used. We provide protein expression and protein function screening methods at the semi-genomic level and also introduce new approaches to enhance study of chemical biology by adapting the cell-free system of wheat germ. With this review, I would like to highlight the potential of the wheat germ cell-free system and position it as a widely used tool for the previously difficult task of recombinant protein preparation and functional analysis.
Collapse
Affiliation(s)
- Keiichirou Nemoto
- Iwate Biotechnology Research Center, 22-174-4 Narita, Kitakami, Iwate 024-0003, Japan
| |
Collapse
|
42
|
Fontana F, Donato AC, Malik A, Gelain F. Unveiling Interactions between Self-Assembling Peptides and Neuronal Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26811-26823. [PMID: 39653368 DOI: 10.1021/acs.langmuir.4c02050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
The use of self-assembling peptide hydrogels in the treatment of spinal cord and brain injuries, especially when combined with adult neural stem cells, has shown great potential. To advance tissue engineering, it is essential to understand the effect of mechanochemical signaling on cellular differentiation. The elucidation of the molecular interactions at the level of the neuronal membrane still represents a promising area of investigation for many drug delivery and tissue engineering applications. An innovative molecular dynamics framework has been introduced to investigate the effect of SAP fibrils with different charges on neural membrane lipid domain dynamics. Such advance enables the in silico exploration of the biomimetic properties of SAP hydrogels and other polymeric biomaterials for tissue engineering applications.
Collapse
Affiliation(s)
- Federico Fontana
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| | - Alice Cristina Donato
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Histology Unit, Department of Molecular Medicine, University of Padova, Padova 35121, Italy
| | - Ashish Malik
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| | - Fabrizio Gelain
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| |
Collapse
|
43
|
Nguyen A, Ondrus AE. In Silico Tools to Score and Predict Cholesterol-Protein Interactions. J Med Chem 2024; 67:20765-20775. [PMID: 39616623 DOI: 10.1021/acs.jmedchem.4c01885] [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: 12/13/2024]
Abstract
Cholesterol is structurally distinct from other lipids, which confers it with singular roles in membrane organization and protein function. As a signaling molecule, cholesterol engages in discrete interactions with transmembrane, peripheral, and certain soluble proteins to control cellular responses. Accordingly, the cholesterol-protein interface is central to cholesterol-related diseases and is an essential consideration in drug design. However, cholesterol's hydrophobic, un-drug-like nature presents a unique challenge to traditional in silico analyses. In this Perspective, we survey a collection of tools designed to predict and evaluate cholesterol binding sites in proteins, including classical sequence motifs, molecular docking, template-based strategies, molecular dynamics simulations, and recent artificial intelligence approaches. We then comment on contemporary tools to evaluate ligand-protein interactions, their applicability to cholesterol, and the yet-untapped potential of cholesterol-protein interactions in human health and disease.
Collapse
Affiliation(s)
- Anna Nguyen
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60607, United States
| | - Alison E Ondrus
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60607, United States
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
44
|
Veeramachaneni RJ, Donelan CA, Tomcho KA, Aggarwal S, Lapinsky DJ, Cascio M. Structural studies of the human α 1 glycine receptor via site-specific chemical cross-linking coupled with mass spectrometry. BIOPHYSICAL REPORTS 2024; 4:100184. [PMID: 39393591 PMCID: PMC11550363 DOI: 10.1016/j.bpr.2024.100184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
By identifying distance constraints, chemical cross-linking coupled with mass spectrometry (CX-MS) can be a powerful complementary technique to other structural methods by interrogating macromolecular protein complexes under native-like conditions. In this study, we developed a CX-MS approach to identify the sites of chemical cross-linking from a single targeted location within the human α1 glycine receptor (α1 GlyR) in its apo state. The human α1 GlyR belongs to the family of pentameric ligand-gated ion channel receptors that function in fast neurotransmission. A single chemically reactive cysteine was reintroduced into a Cys null α1 GlyR construct at position 41 within the extracellular domain of human α1 homomeric GlyR overexpressed in a baculoviral system. After purification and reconstitution into vesicles, methanethiosulfonate-benzophenone-alkyne, a heterotrifunctional cross-linker, was site specifically attached to Cys41 via disulfide bond formation. The resting receptor was then subjected to UV photocross-linking. Afterward, monomeric and oligomeric α1 GlyR bands from SDS-PAGE gels were trypsinized and analyzed by tandem MS in bottom-up studies. Dozens of intrasubunit and intersubunit sites of α1 GlyR cross-linking were differentiated and identified from single gel bands of purified protein, showing the utility of this experimental approach to identify a diverse array of distance constraints of the α1 GlyR in its resting state. These studies highlight CX-MS as an experimental approach to identify chemical cross-links within full-length integral membrane protein assemblies in a native-like lipid environment.
Collapse
Affiliation(s)
- Rathna J Veeramachaneni
- Department of Chemistry and Biochemistry, Duquesne University Pittsburgh, Pittsburgh, Pennsylvania
| | - Chelsee A Donelan
- Department of Chemistry and Biochemistry, Duquesne University Pittsburgh, Pittsburgh, Pennsylvania
| | - Kayce A Tomcho
- Department of Chemistry and Biochemistry, Duquesne University Pittsburgh, Pittsburgh, Pennsylvania; Department of Chemistry, Ohio Wesleyan University, Delaware, Ohio
| | - Shaili Aggarwal
- Graduate School of Pharmaceutical Sciences, Duquesne University Pittsburgh, Pittsburgh, Pennsylvania
| | - David J Lapinsky
- Graduate School of Pharmaceutical Sciences, Duquesne University Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael Cascio
- Department of Chemistry and Biochemistry, Duquesne University Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
45
|
Hu JJ, Yang J, Liu Y, Lu G, Zhao Z, Xia F, Lou X. Tuning the affinity of probes with transmembrane proteins by constructing peptide-conjugated cis/ trans isomers based on molecular scaffolds. J Mater Chem B 2024; 12:12523-12529. [PMID: 39494739 DOI: 10.1039/d4tb01801j] [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: 11/05/2024]
Abstract
For protein analysis, the current peptide-based probes rely almost on the specific recognition of the protein while neglecting the potential influence of the environment near the protein. Herein, we propose that to achieve high recognition of transmembrane protein integrin αvβ3, the interactions from the membrane substrate could be helpful. Moreover, to guarantee the additive effect of different interactions, the cis and trans isomers of peptide-based probes are distinguished. In detail, we synthesized the peptide-conjugated cis/trans isomers (cis-RTP and trans-RTP) by modifying the Arg-Gly-Asp (RGD)-targeting peptide and palmitic acid-conjugated Arg-Arg-Arg-Arg (Pal-RRRR) peptide to the two ends of the molecular scaffold-tetraphenylethene derivative. Due to the difference in spatial structure, isothermal titration calorimetry and simulation experiments demonstrated that cis-RTP can bind more stably to integrin αvβ3 than trans-RTP. As a result, cis-RTP has shown more excellent properties in inhibiting cell migration and killing cells by regulating actin and extracellular signal-regulated kinase. Unlike the existing probe design for protein, this study provides a concept of microenvironment-helpful recognition and a promising strategy of cis/trans isomers to modulate the interaction between proteins and probes.
Collapse
Affiliation(s)
- Jing-Jing Hu
- State Key Laboratory of Biogeology and Environmental Geology, Faculty Materials Science and Chemistry, China University of Geosciences, Wuhan 430078, China.
| | - Juliang Yang
- State Key Laboratory of Biogeology and Environmental Geology, Faculty Materials Science and Chemistry, China University of Geosciences, Wuhan 430078, China.
| | - Yiheng Liu
- State Key Laboratory of Biogeology and Environmental Geology, Faculty Materials Science and Chemistry, China University of Geosciences, Wuhan 430078, China.
| | - Guangwen Lu
- State Key Laboratory of Biogeology and Environmental Geology, Faculty Materials Science and Chemistry, China University of Geosciences, Wuhan 430078, China.
| | - Zujin Zhao
- Department State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou 510640, China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology, Faculty Materials Science and Chemistry, China University of Geosciences, Wuhan 430078, China.
| | - Xiaoding Lou
- State Key Laboratory of Biogeology and Environmental Geology, Faculty Materials Science and Chemistry, China University of Geosciences, Wuhan 430078, China.
| |
Collapse
|
46
|
Samanta R, Harmalkar A, Prathima P, Gray JJ. Advancing Membrane-Associated Protein Docking with Improved Sampling and Scoring in Rosetta. J Chem Theory Comput 2024; 20:10740-10749. [PMID: 39574325 DOI: 10.1021/acs.jctc.4c00927] [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] [Indexed: 11/27/2024]
Abstract
The oligomerization of protein macromolecules on cell membranes plays a fundamental role in regulating cellular function. From modulating signal transduction to directing immune response, membrane proteins (MPs) play a crucial role in biological processes and are often the target of many pharmaceutical drugs. Despite their biological relevance, the challenges in experimental determination have hampered the structural availability of membrane proteins and their complexes. Computational docking provides a promising alternative to model membrane protein complex structures. Here, we present Rosetta-MPDock, a flexible transmembrane (TM) protein docking protocol that captures binding-induced conformational changes. Rosetta-MPDock samples large conformational ensembles of flexible monomers and docks them within an implicit membrane environment. We benchmarked this method on 29 TM-protein complexes of variable backbone flexibility. These complexes are classified based on the root-mean-square deviation between the unbound and bound states (RMSDUB) as rigid (RMSDUB < 1.2 Å), moderately flexible (RMSDUB ∈ [1.2, 2.2] Å), and flexible targets (RMSDUB > 2.2 Å). In a local docking scenario, i.e. with membrane protein partners starting ≈10 Å apart embedded in the membrane in their unbound conformations, Rosetta-MPDock successfully predicts the correct interface (success defined as achieving 3 near-native structures in the 5 top-ranked models) for 67% moderately flexible targets and 60% of the highly flexible targets, a substantial improvement from the existing membrane protein docking methods. Further, by integrating AlphaFold2-multimer for structure determination and using Rosetta-MPDock for docking and refinement, we demonstrate improved success rates over the benchmark targets from 64% to 73%. Rosetta-MPDock advances the capabilities for membrane protein complex structure prediction and modeling to tackle key biological questions and elucidate functional mechanisms in the membrane environment. The benchmark set and the code is available for public use at github.com/Graylab/MPDock.
Collapse
Affiliation(s)
- Rituparna Samanta
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ameya Harmalkar
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Priyamvada Prathima
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
47
|
Engberg O, Mathath AV, Döbel V, Frie C, Lemberg MK, Chakraborty D, Huster D. Evaluating the impact of the membrane thickness on the function of the intramembrane protease GlpG. Biophys J 2024; 123:4067-4081. [PMID: 39488732 PMCID: PMC11628809 DOI: 10.1016/j.bpj.2024.10.019] [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: 07/12/2024] [Revised: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024] Open
Abstract
Cellular membranes exhibit a huge diversity of lipids and membrane proteins that differ in their properties and chemical structure. Cells organize these molecules into distinct membrane compartments characterized by specific lipid profiles and hydrophobic thicknesses of the respective domains. If a hydrophobic mismatch occurs between a membrane protein and the surrounding lipids, there can be functional consequences such as reduced protein activity. This phenomenon has been extensively studied for single-pass transmembrane proteins, rhodopsin, and small polypeptides such as gramicidin. Here, we investigate the E. coli rhomboid intramembrane protease GlpG as a model to systematically explore the impact of membrane thickness on GlpG activity. We used fully saturated 1,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC) and 1,2-dimyristoyl-sn-glycero-3-phosphocholine(DMPC) model lipids and altered membrane thickness by varying the cholesterol content. Physical membrane parameters were determined by 2H and 31P NMR spectroscopy and correlated with GlpG activity measurements in the respective host membranes. Differences in bulk and annular lipids as well as alterations in protein structure in the respective host membranes were determined using molecular dynamics simulations. Our findings indicate that GlpG can influence the membrane thickness in DLPC/cholesterol membranes but not in DMPC/cholesterol membranes. Moreover, we observe that GlpG protease activity is reduced in DLPC membranes at low cholesterol content, which was not observed for DMPC. While a change in GlpG activity can already be due to smallest differences in the lipid environment, potentially enabling allosteric regulation of intramembrane proteolysis, there is no overall correlation to cholesterol-mediated lipid bilayer organization and phase behavior. Additional factors such as the influence of cholesterol on membrane bending rigidity and curvature energy need to be considered. In conclusion, the functionality of α-helical membrane proteins such as GlpG relies not only on hydrophobic matching but also on other membrane properties, specific lipid interaction, and the composition of the annular layer.
Collapse
Affiliation(s)
- Oskar Engberg
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Anjana V Mathath
- Biophysical and Computational Chemistry Laboratory, Department of Chemistry, National Institute of Technology Karnataka, Mangalore, Karnataka, India
| | - Viola Döbel
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany
| | - Christian Frie
- Center for Biochemistry and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Marius K Lemberg
- Center for Biochemistry and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Debashree Chakraborty
- Biophysical and Computational Chemistry Laboratory, Department of Chemistry, National Institute of Technology Karnataka, Mangalore, Karnataka, India
| | - Daniel Huster
- Institute for Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
48
|
Zimmer SE, Giang W, Levental I, Kowalczyk AP. The transmembrane domain of the desmosomal cadherin desmoglein-1 governs lipid raft association to promote desmosome adhesive strength. Mol Biol Cell 2024; 35:ar152. [PMID: 39504468 PMCID: PMC11656464 DOI: 10.1091/mbc.e24-05-0200] [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: 05/01/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Cholesterol- and sphingolipid-enriched domains called lipid rafts are hypothesized to selectively coordinate protein complex assembly within the plasma membrane to regulate cellular functions. Desmosomes are mechanically resilient adhesive junctions that associate with lipid raft membrane domains, yet the mechanisms directing raft association of the desmosomal proteins, particularly the transmembrane desmosomal cadherins, are poorly understood. We identified the desmoglein-1 (DSG1) transmembrane domain (TMD) as a key determinant of desmoglein lipid raft association and designed a panel of DSG1TMD variants to assess the contribution of TMD physicochemical properties (length, bulkiness, and palmitoylation) to DSG1 lipid raft association. Sucrose gradient fractionations revealed that TMD length and bulkiness, but not palmitoylation, govern DSG1 lipid raft association. Further, DSG1 raft association determines plakoglobin recruitment to raft domains. Super-resolution imaging and functional assays uncovered a strong relationship between the efficiency of DSG1TMD lipid raft association and the formation of morphologically and functionally robust desmosomes. Lipid raft association regulated both desmosome assembly dynamics and DSG1 cell surface stability, indicating that DSG1 lipid raft association is required for both desmosome formation and maintenance. These studies identify the biophysical properties of desmoglein transmembrane domains as key determinants of lipid raft association and desmosome adhesive function.
Collapse
Affiliation(s)
- Stephanie E. Zimmer
- Departments of Dermatology and Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - William Giang
- Departments of Dermatology and Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903
| | - Andrew P. Kowalczyk
- Departments of Dermatology and Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
49
|
Ma Z, Nang SC, Liu Z, Zhu J, Mu K, Xu L, Xiao M, Wang L, Li J, Jiang X. Membrane lipid homeostasis dually regulates conformational transition of phosphoethanolamine transferase EptA. Nat Commun 2024; 15:10166. [PMID: 39580503 PMCID: PMC11585620 DOI: 10.1038/s41467-024-54607-1] [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: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024] Open
Abstract
The phosphoethanolamine transferase EptA utilizes phosphatidylethanolamine (PE) in the bacterial cell membrane to modify the structure of lipopolysaccharide, thereby conferring antimicrobial resistance on Gram-negative pathogens. Previous studies have indicated that excessive consumption of PE can disrupt the cell membrane, leading to cell death. This implies the presence of a regulatory mechanism for EptA catalysis to maintain a balance between antimicrobial resistance and bacterial growth. Through microsecond-scale all-atom molecular dynamics simulations, we demonstrate that membrane lipid homeostasis modulates the conformational transition and catalytic activation of EptA. The conformation of EptA oscillates between closed and open states, ensuring the precise spatiotemporal sequence of substrates binding. Interestingly, the conformation of EptA is significantly influenced by its surrounding lipid microenvironment, particularly the PE proportion in the membrane. PE-rich membrane conditions initiate and stabilize the open conformation of EptA through both orthosteric and allosteric effects. Importantly, the reaction mediated by EptA gradually depletes PE in the membrane, ultimately hindering its conformational transition and catalytic activation. These findings collectively establish a self-promoted model, illustrating the regulatory mechanism of EptA during the development of antibiotic resistance.
Collapse
Affiliation(s)
- Zhenyu Ma
- National Glycoengineering Research Center, Shandong University, Qingdao, China
| | - Sue C Nang
- Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Zhuo Liu
- National Glycoengineering Research Center, Shandong University, Qingdao, China
| | - Jingyi Zhu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Kaijie Mu
- Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Limei Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Min Xiao
- National Glycoengineering Research Center, Shandong University, Qingdao, China
| | - Lushan Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Jian Li
- Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
| | - Xukai Jiang
- National Glycoengineering Research Center, Shandong University, Qingdao, China.
| |
Collapse
|
50
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane lipid nanodomains modulate HCN pacemaker channels in nociceptor DRG neurons. Nat Commun 2024; 15:9898. [PMID: 39548079 PMCID: PMC11568329 DOI: 10.1038/s41467-024-54053-z] [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: 03/13/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we find that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels is likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observe reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
Affiliation(s)
- Lucas J Handlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Natalie L Macchi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Nicolas L A Dumaire
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Lyuba Salih
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Erin N Lessie
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Kyle S McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Aubin Moutal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Gucan Dai
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA.
| |
Collapse
|