1
|
Tormena N, Pilizota T, Voïtchovsky K. A Minimalist Model Lipid System Mimicking the Biophysical Properties of Escherichia coli's Inner Membrane. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:12301-12310. [PMID: 40335890 DOI: 10.1021/acs.langmuir.5c01138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Biological membranes are essential for the development and survival of organisms. They can be highly complex, usually comprising a variety of lipids, proteins, and other biomolecules organized around a lipid bilayer structure. This complexity makes studying specific features of biological membranes difficult, with many research studies relying on simplified models, such as artificial vesicles or supported lipid bilayers. Here, we search for a minimal, lipid-only model system of the Escherichia coli inner membrane. We aim to retain the main lipidomic components in their native ratio while mimicking the membrane's thermal and mechanical properties. Based on previous studies, we identify 18 potential model systems reflecting key aspects of the known lipidomic composition and progressively narrow down our selection based on the systems' phase transition temperature and mechanical properties. We identify three ternary model systems able to form stable bilayers that can be made of the commercially available synthetic lipids 16:0-18:1 phosphatidylethanolamine (POPE), 16:0-18:1 phosphatidylglycerol (POPG), and 16:0-18:1 cardiolipin (CL). We anticipate our results to be of interest for future studies making use of E. coli models, for example, investigating membrane proteins' function or macromolecule-membrane interactions.
Collapse
Affiliation(s)
- Nicolo Tormena
- Physics Department, Durham University, South Road, Durham DH1 3LE, U.K
| | - Teuta Pilizota
- School of Biological Sciences and Centre for Engineering Biology, The University of Edinburgh, Alexander Crum Brown Road, Edinburgh EH9 3FF, U.K
- Department of Physics, University of Cambridge, JJ Thompson Avenue, Cambridge CB3 0HE, U.K
| | | |
Collapse
|
2
|
Ji J, Giraud Q, Diedhiou N, Lipkow E, Spiegelhalter C, Laporte J. BIN1 gene replacement reverses BIN1-related centronuclear myopathy. Mol Ther 2025:S1525-0016(25)00315-6. [PMID: 40308061 DOI: 10.1016/j.ymthe.2025.04.036] [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: 12/06/2024] [Revised: 04/07/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025] Open
Abstract
Centronuclear myopathies (CNMs) are severe genetic disorders characterized by generalized muscle weakness associated with organelle mispositioning in myofibers. Most CNM cases are caused by mutations in proteins involved in membrane remodeling, including amphiphysin 2 (BIN1). There is no treatment, and the pathological mechanisms are not understood. Here, we aimed to cure the Bin1-CNM mouse model (Bin1mck-/-) via an adeno-associated virus (AAV)-based gene replacement strategy. Early systemic exogenous BIN1 expression efficiently prevented disease progression. Moreover, BIN1 expression after disease onset reversed all disease signs 4 weeks after treatment, including motor defects, muscle weakness, muscle and myofiber hypotrophy, kyphosis, nuclei and mitochondria misposition, and altered T-tubules network. We then validated the most efficient construct combining a myotropic AAV serotype with the muscle BIN1 isoform. The rescue correlated with the normalization of autophagy and excitation-contraction coupling markers. Cellular and in vivo investigations revealed that different BIN1 natural isoforms shared similar beneficial effects. Artificial constructs coding for separated protein domains rescued different CNM hallmarks. Only the muscle-specific BIN1 isoform combined the different cellular functions of BIN1 on membrane tubulation and dynamin (DNM2) regulation necessary for a full rescue. Overall, this study validates BIN1 gene replacement as a promising strategy to cure BIN1-related CNM.
Collapse
Affiliation(s)
- Jacqueline Ji
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch-Graffenstaden, France
| | - Quentin Giraud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch-Graffenstaden, France
| | - Nadège Diedhiou
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch-Graffenstaden, France
| | - Eva Lipkow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch-Graffenstaden, France
| | - Coralie Spiegelhalter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch-Graffenstaden, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch-Graffenstaden, France.
| |
Collapse
|
3
|
Komikawa T, Okochi M, Tanaka M. Exploration and analytical techniques for membrane curvature-sensing proteins in bacteria. J Bacteriol 2025; 207:e0048224. [PMID: 40135904 PMCID: PMC12004969 DOI: 10.1128/jb.00482-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
The mechanism by which cells regulate protein localization is an important topic in the field of bacterial biology. In certain instances, the morphology of the biological membrane has been demonstrated to function as a spatial cue for the subcellular localization of proteins. These proteins are capable of sensing membrane curvature and are involved in a number of physiological functions such as cytokinesis and the formation of membrane-bound organelles. This review presents recent advances in the in vitro evaluation of curvature-sensing properties using artificially controlled membranes and purified proteins, as well as microscopic live cell assays. However, these evaluation methodologies often require sophisticated experiments, and the number of identified curvature sensors remains limited. Thus, we present a comprehensive exploration of recently reported curvature-sensing proteins. Subsequently, we summarize the known curvature-sensing proteins in bacteria, in conjunction with the analytical methodologies employed in this field. Finally, future prospects and further requirements in the study of curvature-sensing proteins are discussed.
Collapse
Affiliation(s)
- Takumi Komikawa
- School of Materials and Chemical Technology, Institute of Science Tokyo, Yokohama, Kanagawa, Japan
| | - Mina Okochi
- School of Materials and Chemical Technology, Institute of Science Tokyo, Meguro, Tokyo, Japan
| | - Masayoshi Tanaka
- School of Materials and Chemical Technology, Institute of Science Tokyo, Yokohama, Kanagawa, Japan
| |
Collapse
|
4
|
Felsztyna I, Galassi VV, Wilke N. Selectivity of membrane-active peptides: the role of electrostatics and other membrane biophysical properties. Biophys Rev 2025; 17:591-604. [PMID: 40376421 PMCID: PMC12075043 DOI: 10.1007/s12551-025-01309-7] [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: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 05/18/2025] Open
Abstract
Membrane-active peptides (MAPs) are versatile molecules that interact with lipid bilayers, facilitating processes such as antimicrobial defense, anticancer activity, and membrane translocation. Given that most MAPs are cationic, their selectivity for specific cell membranes has traditionally been attributed to variations in membrane surface charge. However, growing evidence suggests that electrostatics alone cannot fully explain MAPs selectivity. Instead, MAPs activity is also strongly influenced by other membrane biophysical properties, such as lipid packing, phase state, curvature, and the spatial distribution of hydrophobic and charged residues within the peptide sequence. In this review, we summarize the current knowledge on the biophysical determinants of MAPs selectivity. We begin by examining membrane and cell surface electrostatics and their influence on MAPs-membrane interactions, including electrostatically driven peptide conformational changes and lipid recruitment. We then broaden the discussion to include non-electrostatic factors, such as membrane curvature and rheology, which are primarily influenced by sterol or hopanoid content, as well as acyl chain unsaturation and branching. Together, these processes highlight that MAPs selectivity is not governed by any single membrane property but instead emerges from a synergistic interplay of electrostatic, hydrophobic, and topological factors. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-025-01309-7.
Collapse
Affiliation(s)
- Iván Felsztyna
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, X5000HUA Córdoba Argentina
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, X5000HUA Córdoba Argentina
| | - Vanesa V. Galassi
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Padre Jorge Contreras 1300, Mendoza, M5502JMA Mendoza Argentina
- Instituto Interdisciplinario en Ciencias Básicas (ICB), CONICET, Padre Jorge Contreras 1300, Mendoza, M5502JMA Mendoza Argentina
| | - Natalia Wilke
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, X5000HUA Córdoba Argentina
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba, X5000HUA Córdoba Argentina
| |
Collapse
|
5
|
Koiri D, Nandi M, Hameem P M A, Aher JB, Kumar A, Behura A, Meher G, Choudhary V, Choubey S, Saleem M. Real-time visualization reveals Mycobacterium tuberculosis ESAT-6 disrupts phagosome-like compartment via fibril-mediated vesiculation. Cell Rep 2025; 44:115328. [PMID: 39982820 PMCID: PMC7617678 DOI: 10.1016/j.celrep.2025.115328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/10/2024] [Accepted: 01/28/2025] [Indexed: 02/23/2025] Open
Abstract
Mycobacterium tuberculosis (Mtb) evades host defense by hijacking and rupturing the phagosome. ESAT-6, a secreted virulence protein of Mtb, is known to be critical for phagosome rupture. However, the mechanism of ESAT-6-mediated disruption of the phagosomal membrane remains unknown. Using in vitro reconstitution, live-cell imaging, and numerical simulations, we discover that ESAT-6 polymerization forces remodeling and vesiculation of the phagosome-like compartment both in vitro and in vivo. Shallow insertion of ESAT-6 leads to tubular and bud-like deformations on the membrane facilitated by a reduction in membrane tension. Growing fibrils generate both radial and tangential forces causing local remodeling and shape transition of the membrane into buds. The ESAT-6-bound tensed membrane undergoes local changes in membrane curvature and lipid phase separation that assist the subsequent fission. Overall, the findings provide mechanistic insights into the long-standing question of phagosome disruption by Mtb for its escape.
Collapse
Affiliation(s)
- Debraj Koiri
- School of Biological Sciences, National Institute of Science Education & Research (NISER), Bhubaneshwar, India; Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Mintu Nandi
- Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, India
| | - Abik Hameem P M
- School of Biological Sciences, National Institute of Science Education & Research (NISER), Bhubaneshwar, India; Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Jayesh Bhausaheb Aher
- School of Biological Sciences, National Institute of Science Education & Research (NISER), Bhubaneshwar, India; Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Akhil Kumar
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Assirbad Behura
- School of Biological Sciences, National Institute of Science Education & Research (NISER), Bhubaneshwar, India; Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Geetanjali Meher
- School of Biological Sciences, National Institute of Science Education & Research (NISER), Bhubaneshwar, India; Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vineet Choudhary
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep Choubey
- Institute of Mathematical Sciences (IMSc), Chennai, India; Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Mohammed Saleem
- School of Biological Sciences, National Institute of Science Education & Research (NISER), Bhubaneshwar, India; Homi Bhabha National Institute (HBNI), Mumbai, India; Center for Interdisciplinary Sciences, National Institute of Science Education & Research (NISER), Bhubaneshwar, India.
| |
Collapse
|
6
|
Bonazzi F, Weikl TR. Membrane-mediated interactions between arc-shaped particles strongly depend on membrane curvature. NANOSCALE 2025; 17:6841-6853. [PMID: 39964755 DOI: 10.1039/d4nr04674a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Besides direct molecular interactions, proteins and nanoparticles embedded in or adsorbed to membranes experience indirect interactions that are mediated by the membranes. Membrane-mediated interactions between curvature-inducing proteins or nanoparticles can lead to assemblies of particles that generate highly curved spherical or tubular membrane shapes, but have mainly been quantified for planar or weakly curved membranes. In this article, we systematically investigate the membrane-mediated interactions of arc-shaped particles adsorbed to a variety of tubular and spherical membrane shapes with coarse-grained modelling and simulations. These arc-shaped particles induce membrane curvature by binding to the membrane with their inner, concave side akin to N-BAR domain proteins. We determine both the pairwise interaction free energy, which includes entropic contributions due to rotational entropy loss at close particle distances, and the pairwise interaction energy without entropic components from particle distributions observed in the simulations. For membrane shapes with small curvature, the membrane-mediated interaction free energies of particle pairs exceed the thermal energy kBT and can lead to particle ordering and aggregation. The interactions strongly decrease with increasing curvature of the membrane shape and are minimal for tubular shapes with membrane curvatures close to the particle curvature.
Collapse
Affiliation(s)
- Francesco Bonazzi
- Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany.
| | - Thomas R Weikl
- Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany.
| |
Collapse
|
7
|
Panda MS, Raghav S, Ghosh SK, Chakraborty H. gp41 Fusion Peptide Alters the Properties of Lipid Monolayer at the Air-Water Interface in a Cholesterol-Dependent Fashion: Implications in Membrane Fusion. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:6103-6112. [PMID: 40025733 DOI: 10.1021/acs.langmuir.4c05071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Enveloped viruses fuse at the cell surface or the endosome after the virus is endocytosed for cellular entry. Membrane fusion is a crucial stage in infection regardless of the pathway. The effect of the fusion peptide, a 20-25 N-terminal residue of the fusion domain, facilitates membrane fusion in multiple ways. In this work, we have assessed the effect of the gp41 fusion peptide from the human immunodeficiency virus (HIV) on the DOPC/DOPE/DOPG monolayers with varying concentrations of cholesterol. The ability of the gp41 fusion peptide to promote fusion among small unilamellar vesicles (SUVs) was examined with the same lipid composition, for which we measured the monolayer properties. Our results show that the peptide is adsorbed on the lipid headgroup in the aqueous subphase in the absence of cholesterol, whereas it gets integrated (orients parallel to the lipid molecule) with the addition of cholesterol in the lipid mixture. To support our findings from monolayer measurements, we have further assessed the peptide's impact on the depth-dependent ordering and polarity of the bilayer membrane using steady-state and time-resolved fluorescence techniques. The peptide-induced change in the elastic compressional modulus also depends on the mole fraction of cholesterol in the lipid mixture. We further compared the ability of the peptide to induce fusion with the change in monolayer properties. Our results demonstrate that the orientation of the peptide in the lipid is crucial for its fusogenic ability as the percentage change in the lipid mixing is correlated to the change in area lift-off parameter in monolayer measurements. Taken together, our results offer a better understanding of the cholesterol-dependent fusogenic ability of the gp41 fusion peptide.
Collapse
Affiliation(s)
| | - Sonam Raghav
- Department of Physics, School of Natural Sciences, Shiv Nadar Institution of Eminence, NH 91, Tehsil Dadri 201314, Uttar Pradesh, India
| | - Sajal K Ghosh
- Department of Physics, School of Natural Sciences, Shiv Nadar Institution of Eminence, NH 91, Tehsil Dadri 201314, Uttar Pradesh, India
| | - Hirak Chakraborty
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla 768 109, Odisha, India
| |
Collapse
|
8
|
Lamari F, Rossignol F, Mitchell GA. Glycerophospholipids: Roles in Cell Trafficking and Associated Inborn Errors. J Inherit Metab Dis 2025; 48:e70019. [PMID: 40101691 PMCID: PMC11919462 DOI: 10.1002/jimd.70019] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/20/2025]
Abstract
Glycerophospholipids (GPLs) are the main lipid components of cellular membranes. They are implicated in membrane structure, vesicle trafficking, neurotransmission, and cell signalling. GPL molecules are amphiphilic, organized around the three carbons of glycerol. Positions sn-1 and sn-2 are each esterified to a fatty acid (FA). At position sn-3, a phosphate group is linked, which in turn can bind a polar head group, the most prevalent classes being phosphatidic acid (PA, phosphate alone as head group), phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS), phosphatidylinositol (PI), and cardiolipin (CL). Pathways of GPL biosynthesis span several cell compartments (endoplasmic reticulum (ER), Golgi mitochondria). Particularly important are mitochondria-associated membranes (MAMs), where the ER and mitochondrial outer membrane are in proximity. After synthesis, GPLs continuously undergo remodelling by FA hydrolysis and re-esterification. Esterification with different FAs alters membrane properties. Many steps in GPL synthesis and remodelling can be mediated by more than one enzyme, suggesting complexity that requires further exploration. The 38 known GPL-related inborn errors are clinically diverse. 23 (61%) have neurologic features, sometimes progressive and severe, particularly developmental delay/encephalopathy in 16 (42%) and spastic paraplegia in 12 (32%). Photoreceptor/neuroretinal disease occurs in 14 (37%). Three present skeletal dysplasias (8%). Most GPL inborn errors have been diagnosed by broad molecular testing. Lipidomics holds promise for diagnostic testing and for the discovery of functionally relevant metabolite profiles for monitoring natural history and treatment response.
Collapse
Affiliation(s)
- Foudil Lamari
- Metabolic Biochemistry, Neurometabolic and Neurodegenerative Unit – DMU BioGeMH Hôpital Pitié‐Salpêtrière, AP‐HP. Sorbonne UniversitéParisFrance
- Brain Institute ‐ Institut du Cerveau ‐ ICM, Inserm U1127, Hôpital Pitié‐SalpêtrièreParisFrance
| | - Francis Rossignol
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of HealthBethesdaMarylandUSA
- Division of Medical Genetics and Genomics, Department of PediatricsCHU Sainte Justine and Université de MontréalMontréalCanada
| | - Grant A. Mitchell
- Division of Medical Genetics and Genomics, Department of PediatricsCHU Sainte Justine and Université de MontréalMontréalCanada
| |
Collapse
|
9
|
Zhang H, Wu C, Zhang M, Li M, Xu H, Zhao W, Zhang T, Lei H. Integrating widely targeted and oxylipin-targeted lipidomics unravels lipid characteristic evolution and oxidation markers in walnuts during deterioration. Food Chem 2025; 466:142197. [PMID: 39612839 DOI: 10.1016/j.foodchem.2024.142197] [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: 07/04/2024] [Revised: 10/28/2024] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
The susceptibility of walnut lipids to deterioration constitutes challenges for industry development, and the oxylipins formed during this process remain to be explored. This study employed lipidomics to reveal the dynamic evolution of lipid characteristics and identify oxidation markers from oxylipins in walnuts during accelerated storage. Glycerophospholipid (GP) content continuously declined in the initial and severe deterioration stages. The accumulation of diglycerides and partial lysophospholipids characterized initial deterioration. Triglycerides were prone to direct oxidation, while GPs tended to be first hydrolyzed. GP metabolism especially phosphatidylethanolamine degradation triggered walnut deterioration. Moreover, ten oxylipins derived from linoleic acids were identified in walnuts. Trans-EKODE-(E)-Ib, 13-HODE, 9-HODE, and 9(S),12(S),13(S)-TriHOME were screened as oxidation markers. The cellular structure exhibited the cell membrane and oil body membrane rupture during deterioration. Potential mechanisms of lipid deterioration were proposed, providing a scientific basis and guidance in optimizing quality control strategies and assessing practical deterioration degrees of walnuts.
Collapse
Affiliation(s)
- Hexin Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Caiyun Wu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Mengmeng Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Mei Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Huaide Xu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Wenge Zhao
- Aksu Zhejiang Fruit Industry Co., Ltd., Aksu 843000, China
| | - Ting Zhang
- Institute of Farm Product Storage and Processing, Xinjiang Academy of Agricultural Science, Urumqi 830091, China.
| | - Hongjie Lei
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
10
|
Lou T, Zhuang X, Chang J, Gao Y, Yuan C, Bai X. Development of a Multifunctional Antimicrobial Peptide for Marine Antifouling by Theoretical Calculations and Experimental Approaches. ACS APPLIED MATERIALS & INTERFACES 2025; 17:12899-12910. [PMID: 39953978 DOI: 10.1021/acsami.4c21224] [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: 02/17/2025]
Abstract
Marine biofouling poses significant challenges to the economy, safety, and reliability of marine infrastructure. While antimicrobial peptides (AMPs) have emerged as green and efficient antifouling agents, their single functionality and the complexity of preparing antifouling surfaces remain key challenges. This study introduces a multifunctional AMP with combined adhesion and antimicrobial properties, derived from 3,4-dihydroxy-l-phenylalanine (DOPA) and IP12 (sequence IRLRWRWKWPWP). The directional recombination of AMP was guided by theoretical calculations. Density functional theory (DFT) simulations identify that the hydroxyl groups of DOPA were the main activating groups that react with aluminum alloy. Coarse-grained molecular dynamics (CG MD) and all-atom molecular dynamics (AA MD) simulations revealed that amino acid residues near the N-terminal of the IP12 could induce cell membrane bending and rupture. The AMP surfaces were fabricated to validate the accurate calibration of the simulations and performance of multifunctional AMP. Atomic force microscopy, Fourier transform infrared, and X-ray photoelectron spectroscopy results confirm the successful construction of AMP surfaces through adhesion function. Antifouling evaluations demonstrated the antifouling properties of AMP surfaces against Escherichia coli (Gram-negative) and Bacillus sp. (Gram-positive), achieving antifouling rates of 85.8 and 82.4%, respectively. This study provides valuable insights into the design of multifunctional AMPs.
Collapse
Affiliation(s)
- Tong Lou
- School of Marine Engineering, Jimei University, Xiamen 361021, China
- Fujian Institute of Innovation for Marine Equipment Detection and Remanufacturing Industrial Technology, Xiamen 361021, China
| | - Xueqiang Zhuang
- School of Marine Engineering, Jimei University, Xiamen 361021, China
- Fujian Institute of Innovation for Marine Equipment Detection and Remanufacturing Industrial Technology, Xiamen 361021, China
| | - Jiangfan Chang
- School of Marine Engineering, Jimei University, Xiamen 361021, China
- Fujian Institute of Innovation for Marine Equipment Detection and Remanufacturing Industrial Technology, Xiamen 361021, China
| | - Yali Gao
- School of Marine Engineering, Jimei University, Xiamen 361021, China
- Fujian Institute of Innovation for Marine Equipment Detection and Remanufacturing Industrial Technology, Xiamen 361021, China
| | - Chengqing Yuan
- State Key Laboratory of Maritime Technology and Safety, Wuhan University of Technology, Wuhan 430063, China
- Reliability Engineering Institute, National Engineering Research Center for Water Transport Safety, Wuhan University of Technology, Wuhan 430063, China
| | - Xiuqin Bai
- State Key Laboratory of Maritime Technology and Safety, Wuhan University of Technology, Wuhan 430063, China
- Reliability Engineering Institute, National Engineering Research Center for Water Transport Safety, Wuhan University of Technology, Wuhan 430063, China
| |
Collapse
|
11
|
Valderas-Gutiérrez J, Davtyan R, Prinz CN, Sparr E, Jönsson P, Linke H, Höök F. Comparative Kinetics of Supported Lipid Bilayer Formation on Silica Coated Vertically Oriented Highly Curved Nanowires and Planar Silica Surfaces. NANO LETTERS 2025; 25:3085-3092. [PMID: 39914804 PMCID: PMC11869362 DOI: 10.1021/acs.nanolett.4c05303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
Supported lipid bilayers (SLBs), formed via lipid vesicle adsorption on highly curved silica surfaces, are widely used in biosensor applications and as models for curved cell membranes. However, SLB formation is often hindered on convex structures with radii comparable to the vesicles. In this study, lightguiding semiconductor nanowires (NWs), engineered for fluorescence signal enhancement, were used to compare the kinetics of SLB formation on vertically oriented NWs and planar silica surfaces. Time resolved fluorescence microscopy with single-molecule sensitivity revealed that while vesicle adsorption rates were similar on both surfaces lateral expansion of the SLB was up to three times faster on NWs than on the planar control. This accelerated expansion is attributed to lower energy penalties when SLBs spread along the cylindrical NWs compared with a planar surface, accompanied by accelerated SLB expansion driven by the merging of the SLB with excess lipids from vesicles accumulated on the NWs.
Collapse
Affiliation(s)
- Julia Valderas-Gutiérrez
- NanoLund, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
- Solid
State Physics, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
| | - Rubina Davtyan
- NanoLund, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
- Solid
State Physics, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
| | - Christelle N. Prinz
- NanoLund, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
- Solid
State Physics, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
| | - Emma Sparr
- Physical
Chemistry, Lund University, P.O. Box 124, SE-22100 Lund, Sweden
| | - Peter Jönsson
- Physical
Chemistry, Lund University, P.O. Box 124, SE-22100 Lund, Sweden
| | - Heiner Linke
- NanoLund, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
- Solid
State Physics, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
| | - Fredrik Höök
- NanoLund, Lund University, P.O. Box 118, SE-22100 Lund, Sweden
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| |
Collapse
|
12
|
Xiao K, Park S, Stachowiak JC, Rangamani P. Biophysical modeling of membrane curvature generation and curvature sensing by the glycocalyx. Proc Natl Acad Sci U S A 2025; 122:e2418357122. [PMID: 39969997 PMCID: PMC11873937 DOI: 10.1073/pnas.2418357122] [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: 09/09/2024] [Accepted: 01/11/2025] [Indexed: 02/21/2025] Open
Abstract
Generation of membrane curvature is fundamental to cellular function. Recent studies have established that the glycocalyx, a sugar-rich polymer layer at the cell surface, can generate membrane curvature. While there have been some theoretical efforts to understand the interplay between the glycocalyx and membrane bending, there remain open questions about how the properties of the glycocalyx affect membrane bending. For example, the relationship between membrane curvature and the density of glycosylated proteins on its surface remains unclear. In this work, we use polymer brush theory to develop a detailed biophysical model of the energetic interactions of the glycocalyx with the membrane. Using this model, we identify the conditions under which the glycocalyx can both generate and sense curvature. Our model predicts that the extent of membrane curvature generated depends on the grafting density of the glycocalyx and the backbone length of the polymers constituting the glycocalyx. Furthermore, when coupled with the intrinsic membrane properties such as spontaneous curvature and a line tension along the membrane, the curvature generation properties of the glycocalyx are enhanced. These predictions were tested experimentally by examining the propensity of glycosylated transmembrane proteins to drive the assembly of highly curved filopodial protrusions at the plasma membrane of adherent mammalian cells. Our model also predicts that the glycocalyx has curvature-sensing capabilities, in agreement with the results of our experiments. Thus, our study develops a quantitative framework for mapping the properties of the glycocalyx to the curvature generation capability of the membrane.
Collapse
Affiliation(s)
- Ke Xiao
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA92093
| | - Sujeong Park
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX78712
| | - Jeanne C. Stachowiak
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX78712
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX78712
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA92093
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
13
|
Straub VM, Barti B, Tandar ST, Stevens AF, van Egmond N, van der Wel T, Zhu N, Rüegger J, van der Horst C, Heitman LH, Li Y, Stella N, van Hasselt JGC, Katona I, van der Stelt M. The endocannabinoid 2-arachidonoylglycerol is released and transported on demand via extracellular microvesicles. Proc Natl Acad Sci U S A 2025; 122:e2421717122. [PMID: 39977325 PMCID: PMC11873938 DOI: 10.1073/pnas.2421717122] [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: 10/23/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
While it is known that endocannabinoids (eCB) modulate multiple neuronal functions, the molecular mechanism governing their release and transport remains elusive. Here, we propose an "on-demand release" model, wherein the formation of microvesicles, a specific group of extracellular vesicles (EVs) containing the eCB, 2-arachidonoylglycerol (2-AG), is an important step. A coculture model system that combines a reporter cell line expressing the fluorescent eCB sensor, G protein-coupled receptor-based (GRAB)eCB2.0, and neuronal cells revealed that neurons release EVs containing 2-AG, but not anandamide, in a stimulus-dependent process regulated by protein kinase C, Diacylglycerol lipase, Adenosinediphosphate (ADP) ribosylation factor 6 (Arf6), and which was sensitive to inhibitors of eCB facilitated diffusion. A vesicle contained approximately 2,000 2-AG molecules. Accordingly, hippocampal eCB-mediated synaptic plasticity was modulated by Arf6 and transport inhibitors. The "on-demand release" model, supported by mathematical analysis, offers a cohesive framework for understanding eCB trafficking at the molecular level and suggests that microvesicles carrying signaling lipids in their membrane regulate neuronal functions in parallel to canonical synaptic vesicles.
Collapse
Affiliation(s)
- Verena M. Straub
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Benjamin Barti
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN47405-2204
| | - Sebastian T. Tandar
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden2333 CC, The Netherlands
| | - A. Floor Stevens
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Noëlle van Egmond
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Tom van der Wel
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Na Zhu
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Joel Rüegger
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Cas van der Horst
- Department of Medicinal Chemistry, Leiden University, Leiden2333 CC, The Netherlands
| | - Laura H. Heitman
- Department of Medicinal Chemistry, Leiden University, Leiden2333 CC, The Netherlands
- Oncode Institute, Leiden2333 CC, The Netherlands
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Beijing100871, China
| | - Nephi Stella
- Department of Pharmacology, School of Medicine, University of Washington, Seattle, WA98195
| | - J. G. Coen van Hasselt
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden2333 CC, The Netherlands
| | - István Katona
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN47405-2204
- Molecular Neurobiology Research Group, Hungarian Research Network, Institute of Experimental Medicine, BudapestH-1083, Hungary
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
- Oncode Institute, Leiden2333 CC, The Netherlands
| |
Collapse
|
14
|
Grage SL, Guschtschin-Schmidt N, Meng B, Kohlmeyer A, Afonin S, Ulrich AS. Interaction of Squalamine with Lipid Membranes. J Phys Chem B 2025; 129:1760-1773. [PMID: 39905636 DOI: 10.1021/acs.jpcb.4c06576] [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: 02/06/2025]
Abstract
Squalamine is an aminosterol from dogfish shark which has drawn attention, besides its antimicrobial activity, as a drug candidate in the treatment of Parkinson's disease due to its ability to prevent binding of α-synuclein to lipid membranes. To get insight into the mode of action of this steroid, we studied the influence of squalamine on lipid bilayers and whether it could inhibit the binding of a model peptide. Solid-state 19F NMR of labeled [KIGAKI]3 indicated that, indeed, this peptide no longer binds as a flexible chain to the bilayer in the presence of squalamine. When the cationic squalamine was added to lipid vesicles containing phosphatidylglycerol lipids, the aminosterol was found in differential scanning calorimetry and solid-state 31P NMR experiments to lower the gel-to-fluid phase transition and cause the phase separation of domains enriched in anionic lipids. Squalamine had only a little influence on 2H NMR relaxation and on the order parameters of the chains. These findings indicate that the aminosterol does not affect the molecular mobility of the hydrophobic core of the bilayer; hence, it does not insert into the membrane, nor causes thinning as found for molecules inserting in the headgroup region. On the other hand, squalamine was found to interact with lipid headgroups through electrostatic interactions, as seen by solid-state 2H NMR on headgroup-labeled lipids. Furthermore, 31P NMR showed that squalamine shifted the lamellar-to-hexagonal phase transition of phosphatidylethanolamine lipids to higher temperatures, indicating a preference for positively curved membranes. Altogether, our experiments indicate a strong interaction of the cationic squalamine with lipid headgroups, in particular with anionic lipids. This affinity for membranes is strong enough to efficiently displace cationic polypeptides, confirming the proposed action mechanism in Parkinson treatment. Notably, supported by 1H-1H NOESY experiments, it was found that squalamine does not insert into the bilayer, but rather acts as facial amphiphile binding to the membrane surface. The binding to membranes may be envisaged in the form of oligomeric or micellar assemblies, which can disrupt the membrane at high concentrations, thereby explaining the antimicrobial and antifungal activities of squalamine.
Collapse
Affiliation(s)
- Stephan L Grage
- Karlsruhe Institute of Technology (KIT), Institute of Biological Interfaces (IBG-2), P.O. Box 3640, 76021 Karlsruhe, Germany
| | - Nadja Guschtschin-Schmidt
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
| | - Beibei Meng
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
| | - Annika Kohlmeyer
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
| | - Sergii Afonin
- Karlsruhe Institute of Technology (KIT), Institute of Biological Interfaces (IBG-2), P.O. Box 3640, 76021 Karlsruhe, Germany
| | - Anne S Ulrich
- Karlsruhe Institute of Technology (KIT), Institute of Biological Interfaces (IBG-2), P.O. Box 3640, 76021 Karlsruhe, Germany
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
| |
Collapse
|
15
|
Yao S, Yang B, Li J, Tang S, Tang S, Kim SC, Wang X. Phosphatidic acid signaling in modulating plant reproduction and architecture. PLANT COMMUNICATIONS 2025; 6:101234. [PMID: 39722455 PMCID: PMC11897466 DOI: 10.1016/j.xplc.2024.101234] [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: 09/30/2024] [Revised: 11/27/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Phosphatidic acid (PA) is an important class of signaling lipids involved in various biological processes in plants. Functional characterization of mutants of PA-metabolizing enzymes, combined with lipidomics and protein-lipid interaction analyses, has revealed the key role of PA signaling in plant responses to biotic and abiotic stresses. Moreover, PA and its metabolizing enzymes influence several reproductive processes, including gametogenesis, pollen tube growth, self-incompatibility, haploid embryo formation, embryogenesis, and seed development. They also play a significant role in shaping plant reproductive and root architecture. Recent studies have shed light on the diverse mechanisms of PA's action, though much remains to be elucidated. Here, we summarize recent advances in the study of PA and its metabolizing enzymes, emphasizing their roles in plant sexual reproduction and architecture. We also explore potential mechanisms underlying PA's functions and discuss future research directions.
Collapse
Affiliation(s)
- Shuaibing Yao
- Department of Biology, University of Missouri-St. Louis, St. Louis, MO 63121, USA; Donald Danforth Plant Science Center, St. Louis, MO 63132, USA
| | - Bao Yang
- Department of Biology, University of Missouri-St. Louis, St. Louis, MO 63121, USA; Donald Danforth Plant Science Center, St. Louis, MO 63132, USA
| | - Jianwu Li
- Department of Biology, University of Missouri-St. Louis, St. Louis, MO 63121, USA; Donald Danforth Plant Science Center, St. Louis, MO 63132, USA
| | - Shan Tang
- Department of Biology, University of Missouri-St. Louis, St. Louis, MO 63121, USA; Donald Danforth Plant Science Center, St. Louis, MO 63132, USA
| | - Shaohua Tang
- Department of Biology, University of Missouri-St. Louis, St. Louis, MO 63121, USA; Donald Danforth Plant Science Center, St. Louis, MO 63132, USA
| | - Sang-Chul Kim
- Department of Biology, University of Missouri-St. Louis, St. Louis, MO 63121, USA; Donald Danforth Plant Science Center, St. Louis, MO 63132, USA
| | - Xuemin Wang
- Department of Biology, University of Missouri-St. Louis, St. Louis, MO 63121, USA; Donald Danforth Plant Science Center, St. Louis, MO 63132, USA.
| |
Collapse
|
16
|
Di Cicco A, Manzi J, Maufront J, Cheng X, Dezi M, Lévy D. Galactocerebroside Lipid Nanotubes, a Model Membrane System for Studying Membrane-Associated Proteins on a Molecular Scale. Methods Mol Biol 2025; 2888:237-248. [PMID: 39699735 DOI: 10.1007/978-1-0716-4318-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Galactocerebroside lipid nanotubes are membrane-mimicking systems for studying the function and structure of proteins involved in membrane shape remodeling, such as in intracellular trafficking, cell division, and migration or involved in the formation of membrane contact sites. They exhibit a constant and small diameter of 30 nm and a length of up to 2 μm. They can be functionalized with lipid ligands, providing a large binding surface for protein without membrane shape remodeling. These features make it possible to study protein assemblies on membranes different from those accessible with vesicular systems. This chapter describes the process of galactocerebroside nanotube formation, the incorporation of different lipid ligands, factors influencing protein binding, and the experimental conditions for their use in flotation assay and imaging by transmission electron and cryo-electron microscopy.
Collapse
Affiliation(s)
- Aurélie Di Cicco
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris, France.
| | - John Manzi
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris, France
| | - Julien Maufront
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris, France
| | - Xingyi Cheng
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris, France
| | - Manuela Dezi
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris, France
| | - Daniel Lévy
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physique des Cellules et Cancer, Paris, France.
| |
Collapse
|
17
|
Beck C, Kunze A. Parallelized Mechanical Stimulation of Neuronal Calcium Through Cell-Internal Nanomagnetic Forces Provokes Lasting Shifts in the Network Activity State. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406678. [PMID: 39460486 PMCID: PMC11812431 DOI: 10.1002/smll.202406678] [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: 08/14/2024] [Revised: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Neurons differentiate mechanical stimuli force and rate to elicit unique functional responses, driving the need for further tools to generate various mechanical stimuli. Here, cell-internal nanomagnetic forces (iNMF) are introduced by manipulating internalized magnetic nanoparticles with an external magnetic field across cortical neuron networks in vitro. Under iNMF, cortical neurons exhibit calcium (Ca2+) influx, leading to modulation of activity observed through Ca2+ event rates. Inhibiting particle uptake or altering nanoparticle exposure time reduced the neuronal response to nanomagnetic forces, exposing the requirement of nanoparticle uptake to induce the Ca2+ response. In highly active cortical networks, iNMF robustly modulates synchronous network activity, which is lasting and repeatable. Using pharmacological blockers, it is shown that iNMF activates mechanosensitive ion channels to induce the Ca2+ influx. Then, in contrast to transient mechanically evoked neuronal activity, iNMF activates Ca2+-activated potassium (KCa) channels to stabilize the neuronal membrane potential and induce network activity shifts. The findings reveal the potential of magnetic nanoparticle-mediated mechanical stimulation to modulate neuronal circuit dynamics, providing insights into the biophysics of neuronal computation.
Collapse
Affiliation(s)
- Connor Beck
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, USA
| | - Anja Kunze
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, USA
- Montana Nanotechnology Facility, Montana State University, Bozeman, Montana 59717, USA
- Optical Technology Center, Montana State University, Bozeman, Montana, 59717, USA
| |
Collapse
|
18
|
Keeley O, Coyne AN. Nuclear and degradative functions of the ESCRT-III pathway: implications for neurodegenerative disease. Nucleus 2024; 15:2349085. [PMID: 38700207 PMCID: PMC11073439 DOI: 10.1080/19491034.2024.2349085] [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: 02/11/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024] Open
Abstract
The ESCRT machinery plays a pivotal role in membrane-remodeling events across multiple cellular processes including nuclear envelope repair and reformation, nuclear pore complex surveillance, endolysosomal trafficking, and neuronal pruning. Alterations in ESCRT-III functionality have been associated with neurodegenerative diseases including Frontotemporal Dementia (FTD), Amyotrophic Lateral Sclerosis (ALS), and Alzheimer's Disease (AD). In addition, mutations in specific ESCRT-III proteins have been identified in FTD/ALS. Thus, understanding how disruptions in the fundamental functions of this pathway and its individual protein components in the human central nervous system (CNS) may offer valuable insights into mechanisms underlying neurodegenerative disease pathogenesis and identification of potential therapeutic targets. In this review, we discuss ESCRT components, dynamics, and functions, with a focus on the ESCRT-III pathway. In addition, we explore the implications of altered ESCRT-III function for neurodegeneration with a primary emphasis on nuclear surveillance and endolysosomal trafficking within the CNS.
Collapse
Affiliation(s)
- Olivia Keeley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alyssa N. Coyne
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Deolal P, Scholz J, Ren K, Bragulat-Teixidor H, Otsuka S. Sculpting nuclear envelope identity from the endoplasmic reticulum during the cell cycle. Nucleus 2024; 15:2299632. [PMID: 38238284 PMCID: PMC10802211 DOI: 10.1080/19491034.2023.2299632] [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: 10/18/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
The nuclear envelope (NE) regulates nuclear functions, including transcription, nucleocytoplasmic transport, and protein quality control. While the outer membrane of the NE is directly continuous with the endoplasmic reticulum (ER), the NE has an overall distinct protein composition from the ER, which is crucial for its functions. During open mitosis in higher eukaryotes, the NE disassembles during mitotic entry and then reforms as a functional territory at the end of mitosis to reestablish nucleocytoplasmic compartmentalization. In this review, we examine the known mechanisms by which the functional NE reconstitutes from the mitotic ER in the continuous ER-NE endomembrane system during open mitosis. Furthermore, based on recent findings indicating that the NE possesses unique lipid metabolism and quality control mechanisms distinct from those of the ER, we explore the maintenance of NE identity and homeostasis during interphase. We also highlight the potential significance of membrane junctions between the ER and NE.
Collapse
Affiliation(s)
- Pallavi Deolal
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| | - Julia Scholz
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Kaike Ren
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Helena Bragulat-Teixidor
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Shotaro Otsuka
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Department of Molecular Biology, Vienna, Austria
| |
Collapse
|
20
|
Peng Y, Chen B. Role of cell membrane homeostasis in the pathogenicity of pathogenic filamentous fungi. Virulence 2024; 15:2299183. [PMID: 38156783 PMCID: PMC10761126 DOI: 10.1080/21505594.2023.2299183] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024] Open
Abstract
The cell membrane forms a fundamental part of all living cells and participates in a variety of physiological processes, such as material exchange, stress response, cell recognition, signal transduction, cellular immunity, apoptosis, and pathogenicity. Here, we review the mechanisms and functions of the membrane structure (lipid components of the membrane and the biosynthesis of unsaturated fatty acids), membrane proteins (transmembrane proteins and proteins contributing to membrane curvature), transcriptional regulation, and cell wall components that influence the virulence and pathogenicity of filamentous fungi.
Collapse
Affiliation(s)
- Yuejin Peng
- Yunnan State Key Laboratory of Conservation and Utilization of Biological Resources, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Bin Chen
- Yunnan State Key Laboratory of Conservation and Utilization of Biological Resources, Yunnan Agricultural University, Kunming, Yunnan, China
| |
Collapse
|
21
|
Soni J, Gupta S, Mandal T. Recalibration of MARTINI-3 Parameters for Improved Interactions between Peripheral Proteins and Lipid Bilayers. J Chem Theory Comput 2024; 20:9673-9686. [PMID: 39491480 DOI: 10.1021/acs.jctc.4c00645] [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
The MARTINI force field is one of the most used coarse-grained models for biomolecular simulations. Many limitations of the model including the protein-protein overaggregation have been improved in its latest version, MARTINI-3. In this study, we investigate the efficacy of the MARTINI-3 parameters for capturing the interactions of peripheral proteins with model plasma membranes. Particularly, we consider two classes of proteins, namely, annexin and epsin, which are known to generate negative and positive membrane curvatures, respectively. We find that current MARTINI-3 parameters are not able to correctly describe the protein-membrane interface and the protein-induced membrane curvatures for any of these proteins. The problem arises due to the lack of proper hydrophobic interactions between the protein residues and lipid tails. Making systematic adjustments, we show that a combination of reduction in the protein-water interactions and enhancement of protein-lipid hydrophobic interactions is essential for accurate prediction of the interfacial structure including the protein-induced membrane curvature. Next, we apply our model to a couple of other peripheral proteins, namely, Snf7, a core component of the ESCRT-III complex, and the PH domain of evectin-2. We find that our model captures the protein-membrane interfacial structure much more accurately than the MARTINI-3 model for all of the peripheral proteins considered in this study. However, the strategy described in this study may not be suitable for oligomeric transmembrane proteins where protein-protein hydrophobic interactions should be increased instead of protein-lipid hydrophobic interactions.
Collapse
Affiliation(s)
- Jatin Soni
- Department of Physics, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Shivam Gupta
- Department of Physics, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Taraknath Mandal
- Department of Physics, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
22
|
Seychell RM, El Saghir A, Vassallo N. Modulation of Biological Membranes Using Small-Molecule Compounds to Counter Toxicity Caused by Amyloidogenic Proteins. MEMBRANES 2024; 14:231. [PMID: 39590617 PMCID: PMC11596372 DOI: 10.3390/membranes14110231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
The transition of peptides or proteins along a misfolding continuum from soluble functional states to pathological aggregates, to ultimately deposit as amyloid fibrils, is a process that underlies an expanding group of human diseases-collectively known as protein-misfolding disorders (PMDs). These include common and debilitating conditions, such as Alzheimer's disease, Parkinson's disease, and type-2 diabetes. Compelling evidence has emerged that the complex interplay between the misfolded proteins and biological membranes is a key determinant of the pathogenic mechanisms by which harmful amyloid entities are formed and exert their cytotoxicity. Most efforts thus far to develop disease-modifying treatments for PMDs have largely focused on anti-aggregation strategies: to neutralise, or prevent the formation of, toxic amyloid species. Herein, we review the critical role of the phospholipid membrane in mediating and enabling amyloid pathogenicity. We consequently propose that the development of small molecules, which have the potential to uniquely modify the physicochemical properties of the membrane and make it more resilient against damage by misfolded proteins, could provide a novel therapeutic approach in PMDs. By way of an example, natural compounds shown to intercalate into lipid bilayers and inhibit amyloid-lipid interactions, such as the aminosterols, squalamine and trodusquamine, cholesterol, ubiquinone, and select polyphenols, are discussed. Such a strategy would provide a novel approach to counter a wide range of toxic biomolecules implicit in numerous human amyloid pathologies.
Collapse
Affiliation(s)
- Raina Marie Seychell
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Adam El Saghir
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, MSD 2080 Msida, Malta
| |
Collapse
|
23
|
Yang Y, Valencia LA, Lu CH, Nakamoto ML, Tsai CT, Liu C, Yang H, Zhang W, Jahed Z, Lee WR, Santoro F, Liou J, Wu JC, Cui B. Plasma membrane curvature regulates the formation of contacts with the endoplasmic reticulum. Nat Cell Biol 2024; 26:1878-1891. [PMID: 39289582 PMCID: PMC11567891 DOI: 10.1038/s41556-024-01511-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: 11/20/2023] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
Contact sites between the endoplasmic reticulum (ER) and plasma membrane (PM) play a crucial role in governing calcium regulation and lipid homeostasis. Despite their significance, the factors regulating their spatial distribution on the PM remain elusive. Inspired by observations in cardiomyocytes, where ER-PM contact sites concentrate on tubular PM invaginations known as transverse tubules, we hypothesize that PM curvature plays a role in ER-PM contact formation. Through precise control of PM invaginations, we show that PM curvatures locally induce the formation of ER-PM contacts in cardiomyocytes. Intriguingly, the junctophilin family of ER-PM tethering proteins, specifically expressed in excitable cells, is the key player in this process, whereas the ubiquitously expressed extended synaptotagmin-2 does not show a preference for PM curvature. At the mechanistic level, we find that the low-complexity region (LCR) and membrane occupation and recognition nexus (MORN) motifs of junctophilins can bind independently to the PM, but both the LCR and MORN motifs are required for targeting PM curvatures. By examining the junctophilin interactome, we identify a family of curvature-sensing proteins-Eps15 homology domain-containing proteins-that interact with the MORN_LCR motifs and facilitate the preferential tethering of junctophilins to curved PM. These findings highlight the pivotal role of PM curvature in the formation of ER-PM contacts in cardiomyocytes and unveil a mechanism for the spatial regulation of ER-PM contacts through PM curvature modulation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Luis A Valencia
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Melissa L Nakamoto
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Departments of Physiology and Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Wei Zhang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Zeinab Jahed
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Chemical and Nano Engineering, University of California, San Diego, San Diego, CA, USA
| | - Wan-Ru Lee
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples, Italy
- Faculty of Electrical Engineering and Information Technology, RWTH Aachen University, Aachen, Germany
- Institute of Biological Information Processing-Bioelectronics (IBI-3), Forschungszentrum, Jülich, Germany
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
24
|
Lou T, Zhuang X, Chang J, Gao Y, Bai X. Effect of Surface-Immobilized States of Antimicrobial Peptides on Their Ability to Disrupt Bacterial Cell Membrane Structure. J Funct Biomater 2024; 15:315. [PMID: 39590519 PMCID: PMC11595214 DOI: 10.3390/jfb15110315] [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: 09/30/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Antimicrobial peptide (AMP) surfaces are widely used to inhibit biofilm formation and bacterial infection. However, endpoint-immobilized AMPs on surfaces are totally different from free-state AMPs due to the constraints of the surface. In this work, the interactions between AMPs and bacterial cell membranes were analyzed through coarse-grained molecular dynamics and all-atom molecular dynamics simulations. This AMP disrupted membrane structure by altering the thickness and curvature of the membrane. Furthermore, the effect of surface-immobilized states of AMPs on their ability to disrupt membrane structure was revealed. The immobilized AMPs in the freeze-N system could bind to the membrane and disrupt the membrane structure through electrostatic forces between positively charged N-terminal amino acid residues and the negatively charged membrane, while the immobilized AMPs in the freeze-C system were repelled. The results will aid in the rational design of new AMP surfaces with enhanced efficacy and stability.
Collapse
Affiliation(s)
- Tong Lou
- School of Marine Engineering, Jimei University, Xiamen 361021, China; (T.L.)
- Fujian Institute of Innovation for Marine Equipment Detection and Remanufacturing Industrial Technology, Xiamen 361021, China
| | - Xueqiang Zhuang
- School of Marine Engineering, Jimei University, Xiamen 361021, China; (T.L.)
- Fujian Institute of Innovation for Marine Equipment Detection and Remanufacturing Industrial Technology, Xiamen 361021, China
| | - Jiangfan Chang
- School of Marine Engineering, Jimei University, Xiamen 361021, China; (T.L.)
- Fujian Institute of Innovation for Marine Equipment Detection and Remanufacturing Industrial Technology, Xiamen 361021, China
| | - Yali Gao
- School of Marine Engineering, Jimei University, Xiamen 361021, China; (T.L.)
- Fujian Institute of Innovation for Marine Equipment Detection and Remanufacturing Industrial Technology, Xiamen 361021, China
| | - Xiuqin Bai
- State Key Laboratory of Maritime Technology and Safety, Wuhan University of Technology, Wuhan 430063, China
- Reliability Engineering Institute, National Engineering Research Center for Water Transport Safety, Wuhan University of Technology, Wuhan 430063, China
| |
Collapse
|
25
|
Venkatraman K, Lee CT, Budin I. Setting the curve: the biophysical properties of lipids in mitochondrial form and function. J Lipid Res 2024; 65:100643. [PMID: 39303982 PMCID: PMC11513603 DOI: 10.1016/j.jlr.2024.100643] [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: 08/26/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024] Open
Abstract
Mitochondrial membranes are defined by their diverse functions, complex geometries, and unique lipidomes. In the inner mitochondrial membrane, highly curved membrane folds known as cristae house the electron transport chain and are the primary sites of cellular energy production. The outer mitochondrial membrane is flat by contrast, but is critical for the initiation and mediation of processes key to mitochondrial physiology: mitophagy, interorganelle contacts, fission and fusion dynamics, and metabolite transport. While the lipid composition of both the inner mitochondrial membrane and outer mitochondrial membrane have been characterized across a variety of cell types, a mechanistic understanding for how individual lipid classes contribute to mitochondrial structure and function remains nebulous. In this review, we address the biophysical properties of mitochondrial lipids and their related functional roles. We highlight the intrinsic curvature of the bulk mitochondrial phospholipid pool, with an emphasis on the nuances surrounding the mitochondrially-synthesized cardiolipin. We also outline emerging questions about other lipid classes - ether lipids, and sterols - with potential roles in mitochondrial physiology. We propose that further investigation is warranted to elucidate the specific properties of these lipids and their influence on mitochondrial architecture and function.
Collapse
Affiliation(s)
- Kailash Venkatraman
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Christopher T Lee
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Itay Budin
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
26
|
Malik VK, Pak OS, Feng J. Curvature-Assisted Vesicle Explosion Under Light-Induced Asymmetric Oxidation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400504. [PMID: 39136143 PMCID: PMC11481189 DOI: 10.1002/advs.202400504] [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: 01/17/2024] [Revised: 07/14/2024] [Indexed: 10/17/2024]
Abstract
Exposure of cell membranes to reactive oxygen species can cause oxidation of membrane lipids. Oxidized lipids undergo drastic conformational changes, compromising the mechanical integrity of the membrane and causing cell death. For giant unilamellar vesicles, a classic cell mimetic system, a range of mechanical responses under oxidative assault has been observed including formation of nanopores, transient micron-sized pores, and total sudden catastrophic collapse (i.e., explosion). However, the physical mechanism regarding how lipid oxidation causes vesicles to explode remains elusive. Here, with light-induced asymmetric oxidation experiments, the role of spontaneous curvature on vesicle instability and its link to the conformational changes of oxidized lipid products is systematically investigated. A comprehensive membrane model is proposed for pore-opening dynamics incorporating spontaneous curvature and membrane curling, which captures the experimental observations well. The kinetics of lipid oxidation are further characterized and how light-induced asymmetric oxidation generates spontaneous curvature in a non-monotonic temporal manner is rationalized. Using the framework, a phase diagram with an analytical criterion to predict transient pore formation or catastrophic vesicle collapse is provided. The work can shed light on understanding biomembrane stability under oxidative assault and strategizing release dynamics of vesicle-based drug delivery systems.
Collapse
Affiliation(s)
- Vinit Kumar Malik
- Department of Mechanical Science and EngineeringUniversity of Illinois Urbana‐ChampaignUrbanaIL61801USA
| | - On Shun Pak
- Department of Mechanical Engineering and Department of Applied MathematicsSanta Clara UniversitySanta ClaraCA95053USA
| | - Jie Feng
- Department of Mechanical Science and EngineeringUniversity of Illinois Urbana‐ChampaignUrbanaIL61801USA
- Materials Research LaboratoryUniversity of Illinois Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
27
|
Lewis AH, Cronin ME, Grandl J. Piezo1 ion channels are capable of conformational signaling. Neuron 2024; 112:3161-3175.e5. [PMID: 39043183 PMCID: PMC11427155 DOI: 10.1016/j.neuron.2024.06.024] [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: 12/01/2023] [Revised: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024]
Abstract
Piezo1 is a mechanically activated ion channel that senses forces with short latency and high sensitivity. Piezos undergo large conformational changes, induce far-reaching deformation onto the membrane, and modulate the function of two-pore potassium (K2P) channels. Taken together, this led us to hypothesize that Piezos may be able to signal their conformational state to other nearby proteins. Here, we use chemical control to acutely restrict Piezo1 conformational flexibility and show that Piezo1 conformational changes, but not ion permeation through them, are required for modulating the K2P channel K2P2.1 (TREK1). Super-resolution imaging and stochastic simulations further reveal that both channels do not co-localize, which implies that modulation is not mediated through direct binding interactions; however, at high Piezo1 densities, most TREK1 channels are within the predicted Piezo1 membrane footprint, suggesting that the footprint may underlie conformational signaling. We speculate that physiological roles originally attributed to Piezo1 ionotropic function could, alternatively, involve conformational signaling.
Collapse
Affiliation(s)
- Amanda H Lewis
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Marie E Cronin
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
28
|
Rinaldin M, Ten Haaf SLD, Vegter EJ, van der Wel C, Fonda P, Giomi L, Kraft DJ. Lipid membranes supported by polydimethylsiloxane substrates with designed geometry. SOFT MATTER 2024; 20:7379-7386. [PMID: 39046306 DOI: 10.1039/d4sm00380b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The membrane curvature of cells and intracellular compartments continuously adapts to enable cells to perform vital functions, from cell division to signal trafficking. Understanding how membrane geometry affects these processes in vivo is challenging because of the biochemical and geometrical complexity as well as the short time and small length scales involved in cellular processes. By contrast, in vitro model membranes with engineered curvature would provide a versatile platform for this investigation and applications to biosensing and biocomputing. Here, we present a strategy that allows fabrication of lipid membranes with designed shape by combining 3D micro-printing and replica-molding lithography with polydimethylsiloxane to create curved micrometer-sized scaffolds with virtually any geometry. The resulting supported lipid membranes are homogeneous and fluid. We demonstrate the versatility of the system by fabricating structures of interesting combinations of mean and Gaussian curvature. We study the lateral phase separation and how local curvature influences the effective diffusion coefficient. Overall, we offer a bio-compatible platform for understanding curvature-dependent cellular processes and developing programmable bio-interfaces for living cells and nanostructures.
Collapse
Affiliation(s)
- Melissa Rinaldin
- Leiden Institute of Physics, University of Leiden, 2300 RA Leiden, The Netherlands.
- Instituut-Lorentz, Universiteit Leiden, Leiden, 2300 RA, The Netherlands
| | | | - Ernst J Vegter
- Leiden Institute of Physics, University of Leiden, 2300 RA Leiden, The Netherlands.
| | - Casper van der Wel
- Leiden Institute of Physics, University of Leiden, 2300 RA Leiden, The Netherlands.
| | - Piermarco Fonda
- Instituut-Lorentz, Universiteit Leiden, Leiden, 2300 RA, The Netherlands
| | - Luca Giomi
- Instituut-Lorentz, Universiteit Leiden, Leiden, 2300 RA, The Netherlands
| | - Daniela J Kraft
- Leiden Institute of Physics, University of Leiden, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
29
|
Raby A, Missiroli S, Sanatine P, Langui D, Pansiot J, Beaude N, Vezzana L, Saleh R, Marinello M, Laforge M, Pinton P, Buj-Bello A, Burgo A. Spastin regulates ER-mitochondrial contact sites and mitochondrial homeostasis. iScience 2024; 27:110683. [PMID: 39252960 PMCID: PMC11382127 DOI: 10.1016/j.isci.2024.110683] [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: 10/10/2023] [Revised: 05/20/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Mitochondria-endoplasmic reticulum (ER) contact sites (MERCs) emerged to play critical roles in numerous cellular processes, and their dysregulation has been associated to neurodegenerative disorders. Mutations in the SPG4 gene coding for spastin are among the main causes of hereditary spastic paraplegia (HSP). Spastin binds and severs microtubules, and the long isoform of this protein, namely M1, spans the outer leaflet of ER membrane where it interacts with other ER-HSP proteins. Here, we showed that overexpressed M1 spastin localizes in ER-mitochondria intersections and that endogenous spastin accumulates in MERCs. We demonstrated in different cellular models that downregulation of spastin enhances the number of MERCs, alters mitochondrial morphology, and impairs ER and mitochondrial calcium homeostasis. These effects are associated with reduced mitochondrial membrane potential, oxygen species levels, and oxidative metabolism. These findings extend our knowledge on the role of spastin in the ER and suggest MERCs deregulation as potential causes of SPG4-HSP disease.
Collapse
Affiliation(s)
- Amelie Raby
- Genethon, 91000 Evry, France
- Université Paris-Saclay, University Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Sonia Missiroli
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, and Technopole of Ferrara, Laboratory for Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | | | - Dominique Langui
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm U1127, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Julien Pansiot
- Université Paris Cité, NeuroDiderot, Inserm, 75019 Paris, France
| | - Nissai Beaude
- Genethon, 91000 Evry, France
- Université Paris-Saclay, University Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Lucie Vezzana
- Genethon, 91000 Evry, France
- Université Paris-Saclay, University Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Rachelle Saleh
- Université Paris Cité, NeuroDiderot, Inserm, 75019 Paris, France
| | - Martina Marinello
- Genethon, 91000 Evry, France
- Université Paris-Saclay, University Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Mireille Laforge
- Université Paris Cité, NeuroDiderot, Inserm, 75019 Paris, France
| | - Paolo Pinton
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, and Technopole of Ferrara, Laboratory for Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Ana Buj-Bello
- Genethon, 91000 Evry, France
- Université Paris-Saclay, University Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Andrea Burgo
- Genethon, 91000 Evry, France
- Université Paris-Saclay, University Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| |
Collapse
|
30
|
Sharma KD, Doktorova M, Waxham MN, Heberle FA. Cryo-EM images of phase-separated lipid bilayer vesicles analyzed with a machine-learning approach. Biophys J 2024; 123:2877-2891. [PMID: 38689500 PMCID: PMC11393711 DOI: 10.1016/j.bpj.2024.04.029] [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: 12/22/2023] [Revised: 03/08/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
Lateral lipid heterogeneity (i.e., raft formation) in biomembranes plays a functional role in living cells. Three-component mixtures of low- and high-melting lipids plus cholesterol offer a simplified experimental model for raft domains in which a liquid-disordered (Ld) phase coexists with a liquid-ordered (Lo) phase. Using such models, we recently showed that cryogenic electron microscopy (cryo-EM) can detect phase separation in lipid vesicles based on differences in bilayer thickness. However, the considerable noise within cryo-EM data poses a significant challenge for accurately determining the membrane phase state at high spatial resolution. To this end, we have developed an image-processing pipeline that utilizes machine learning (ML) to predict the bilayer phase in projection images of lipid vesicles. Importantly, the ML method exploits differences in both the thickness and molecular density of Lo compared to Ld, which leads to improved phase identification. To assess accuracy, we used artificial images of phase-separated lipid vesicles generated from all-atom molecular dynamics simulations of Lo and Ld phases. Synthetic ground-truth data sets mimicking a series of compositions along a tieline of Ld + Lo coexistence were created and then analyzed with various ML models. For all tieline compositions, we find that the ML approach can correctly identify the bilayer phase with >90% accuracy, thus providing a means to isolate the intensity profiles of coexisting Ld and Lo phases, as well as accurately determine domain-size distributions, number of domains, and phase-area fractions. The method described here provides a framework for characterizing nanoscopic lateral heterogeneities in membranes and paves the way for a more detailed understanding of raft properties in biological contexts.
Collapse
Affiliation(s)
- Karan D Sharma
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee
| | - Milka Doktorova
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| | - M Neal Waxham
- Department of Neurobiology and Anatomy, University of Texas Health Science Center, Houston, Texas
| | | |
Collapse
|
31
|
Parvin A, Erabi G, Mohammadpour D, Maleki-Kakelar H, Sadeghpour S, Pashaei MR, Taheri-Anganeh M, Ghasemnejad-Berenji H. Infertility: Focus on the therapeutic potential of extracellular vesicles. Reprod Biol 2024; 24:100925. [PMID: 39018753 DOI: 10.1016/j.repbio.2024.100925] [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: 03/30/2024] [Revised: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024]
Abstract
Infertility is a well-known problem that arises from a variety of reproductive diseases. Until now, researchers have tried various methods to restore fertility, including medication specific to the cause, hormone treatments, surgical removals, and assisted reproductive technologies. While these methods do produce results, they do not consistently lead to fertility restoration in every instance. The use of exosome therapy has significant potential in treating infertility in patients. This is because exosomes, microvesicles, and apoptotic bodies, which are different types of vesicles, play a crucial role in transferring bioactive molecules that aid in cell-to-cell communication. Reproductive fluids can transport a variety of molecular cargos, such as miRNAs, mRNAs, proteins, lipids, and DNA molecules. The percentage of these cargos in the fluids can be linked to their physiological and pathological status. EVs are involved in several physiological and pathological processes and offer interesting non-cellular therapeutic possibilities to treat infertility. EVs (extracellular vesicles) transplantation has been shown in many studies to be a key part of regenerating different parts of the reproductive system, including the production of oocytes and the start of sperm production. Nevertheless, the existing evidence necessitates testifying to the effectiveness of injecting EVs in resolving reproductive problems among humans. This review focuses on the current literature about infertility issues in both females and males, specifically examining the potential treatments involving extracellular vesicles (EVs).
Collapse
Affiliation(s)
- Ali Parvin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Donna Mohammadpour
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Sonia Sadeghpour
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Obstetrics & Gynecology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Reza Pashaei
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
32
|
Mendes LFS, Oliveira CG, Simões KF, Kava E, Costa-Filho AJ. Exploring liquid-liquid phase separation in the organisation of Golgi matrix proteins. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1872:141029. [PMID: 38917877 DOI: 10.1016/j.bbapap.2024.141029] [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: 04/08/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024]
Abstract
The Golgi apparatus is a critical organelle in protein sorting and lipid metabolism. Characterized by its stacked, flattened cisternal structure, the Golgi exhibits distinct polarity with its cis- and trans-faces orchestrating various protein maturation and transport processes. At the heart of its structural integrity and organisation are the Golgi Matrix Proteins (GMPs), predominantly comprising Golgins and GRASPs. These proteins contribute to this organelle's unique stacked and polarized structure and ensure the precise localization of Golgi-resident enzymes, which is crucial for accurate protein processing. Despite over a century of research since its discovery, the Golgi architecture's intricate mechanisms still need to be fully understood. Here, we discuss that GMPs across different Eukaryotic lineages present a significant tendency to form biomolecular condensates. Moreover, we validated experimentally that members of the GRASP family also exhibit a strong tendency. Our findings offer a new perspective on the possible roles of protein disorder and condensation of GMPs in the Golgi organisation.
Collapse
Affiliation(s)
- Luis Felipe S Mendes
- Group of Biophysics and Structural Biology "Sergio Mascarenhas". São Carlos Institute of Physics, University of São Paulo, São Carlos, SP, Brazil.
| | - Carolina G Oliveira
- Molecular Biophysics Laboratory, Department of Physics, Faculty of Philosophy, Sciences, and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kevin F Simões
- Group of Biophysics and Structural Biology "Sergio Mascarenhas". São Carlos Institute of Physics, University of São Paulo, São Carlos, SP, Brazil
| | - Emanuel Kava
- Molecular Biophysics Laboratory, Department of Physics, Faculty of Philosophy, Sciences, and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Antonio J Costa-Filho
- Molecular Biophysics Laboratory, Department of Physics, Faculty of Philosophy, Sciences, and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
33
|
Parihar K, Ko SHB, Bradley RP, Taylor P, Ramakrishnan N, Baumgart T, Guo W, Weaver VM, Janmey PA, Radhakrishnan R. Asymmetric crowders and membrane morphology at the nexus of intracellular trafficking and oncology. MECHANOBIOLOGY IN MEDICINE 2024; 2:100071. [PMID: 38899029 PMCID: PMC11185830 DOI: 10.1016/j.mbm.2024.100071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
A definitive understanding of the interplay between protein binding/migration and membrane curvature evolution is emerging but needs further study. The mechanisms defining such phenomena are critical to intracellular transport and trafficking of proteins. Among trafficking modalities, exosomes have drawn attention in cancer research as these nano-sized naturally occurring vehicles are implicated in intercellular communication in the tumor microenvironment, suppressing anti-tumor immunity and preparing the metastatic niche for progression. A significant question in the field is how the release and composition of tumor exosomes are regulated. In this perspective article, we explore how physical factors such as geometry and tissue mechanics regulate cell cortical tension to influence exosome production by co-opting the biophysics as well as the signaling dynamics of intracellular trafficking pathways and how these exosomes contribute to the suppression of anti-tumor immunity and promote metastasis. We describe a multiscale modeling approach whose impact goes beyond the fundamental investigation of specific cellular processes toward actual clinical translation. Exosomal mechanisms are critical to developing and approving liquid biopsy technologies, poised to transform future non-invasive, longitudinal profiling of evolving tumors and resistance to cancer therapies to bring us one step closer to the promise of personalized medicine.
Collapse
Affiliation(s)
- Kshitiz Parihar
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Seung-Hyun B. Ko
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan P. Bradley
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Phillip Taylor
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - N. Ramakrishnan
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Tobias Baumgart
- Department of Chemistry, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie M. Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Paul A. Janmey
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Lincoff J, Helsell CVM, Marcoline FV, Natale AM, Grabe M. Membrane curvature sensing and symmetry breaking of the M2 proton channel from Influenza A. eLife 2024; 13:e81571. [PMID: 39150863 PMCID: PMC11383528 DOI: 10.7554/elife.81571] [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: 07/03/2022] [Accepted: 08/07/2024] [Indexed: 08/18/2024] Open
Abstract
The M2 proton channel aids in the exit of mature influenza viral particles from the host plasma membrane through its ability to stabilize regions of high negative Gaussian curvature (NGC) that occur at the neck of budding virions. The channels are homo-tetramers that contain a cytoplasm-facing amphipathic helix (AH) that is necessary and sufficient for NGC generation; however, constructs containing the transmembrane spanning helix, which facilitates tetramerization, exhibit enhanced curvature generation. Here, we used all-atom molecular dynamics (MD) simulations to explore the conformational dynamics of M2 channels in lipid bilayers revealing that the AH is dynamic, quickly breaking the fourfold symmetry observed in most structures. Next, we carried out MD simulations with the protein restrained in four- and twofold symmetric conformations to determine the impact on the membrane shape. While each pattern was distinct, all configurations induced pronounced curvature in the outer leaflet, while conversely, the inner leaflets showed minimal curvature and significant lipid tilt around the AHs. The MD-generated profiles at the protein-membrane interface were then extracted and used as boundary conditions in a continuum elastic membrane model to calculate the membrane-bending energy of each conformation embedded in different membrane surfaces characteristic of a budding virus. The calculations show that all three M2 conformations are stabilized in inward-budding, concave spherical caps and destabilized in outward-budding, convex spherical caps, the latter reminiscent of a budding virus. One of the C2-broken symmetry conformations is stabilized by 4 kT in NGC surfaces with the minimum energy conformation occurring at a curvature corresponding to 33 nm radii. In total, our work provides atomistic insight into the curvature sensing capabilities of M2 channels and how enrichment in the nascent viral particle depends on protein shape and membrane geometry.
Collapse
Affiliation(s)
- James Lincoff
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Cole V M Helsell
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
- Graduate Group in Biophysics, University of California, San Francisco, San Francisco, United States
| | - Frank V Marcoline
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Andrew M Natale
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
- Graduate Group in Biophysics, University of California, San Francisco, San Francisco, United States
| | - Michael Grabe
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
35
|
Dehinwal R, Gopinath T, Smith RD, Ernst RK, Schifferli DM, Waldor MK, Marassi FM. A pH-sensitive motif in an outer membrane protein activates bacterial membrane vesicle production. Nat Commun 2024; 15:6958. [PMID: 39138228 PMCID: PMC11322160 DOI: 10.1038/s41467-024-51364-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: 11/16/2023] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
Outer membrane vesicles (OMVs) produced by Gram-negative bacteria have key roles in cell envelope homeostasis, secretion, interbacterial communication, and pathogenesis. The facultative intracellular pathogen Salmonella Typhimurium increases OMV production inside the acidic vacuoles of host cells by changing expression of its outer membrane proteins and modifying the composition of lipid A. However, the molecular mechanisms that translate pH changes into OMV production are not completely understood. Here, we show that the outer membrane protein PagC promotes OMV production through pH-dependent interactions between its extracellular loops and surrounding lipopolysaccharide (LPS). Structural comparisons and mutational studies indicate that a pH-responsive amino acid motif in PagC extracellular loops, containing PagC-specific histidine residues, is crucial for OMV formation. Molecular dynamics simulations suggest that protonation of histidine residues leads to changes in the structure and flexibility of PagC extracellular loops and their interactions with the surrounding LPS, altering membrane curvature. Consistent with that hypothesis, mimicking acidic pH by mutating those histidine residues to lysine increases OMV production. Thus, our findings reveal a mechanism for sensing and responding to environmental pH and for control of membrane dynamics by outer membrane proteins.
Collapse
Affiliation(s)
- Ruchika Dehinwal
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, USA
- Department of Microbiology, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA
| | - Tata Gopinath
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Richard D Smith
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Robert K Ernst
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Dieter M Schifferli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, USA.
- Department of Microbiology, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA.
| | | |
Collapse
|
36
|
Mazahir F, Yadav AK. Recent progress in engineered extracellular vesicles and their biomedical applications. Life Sci 2024; 350:122747. [PMID: 38797364 DOI: 10.1016/j.lfs.2024.122747] [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: 02/14/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
AIMS To present the recent update on the isolation, engineering techniques for extracellular vesicles, limitations associated with different isolation techniques, different biomedical applications, and challenges of engineered extracellular vesicles for the benefit of researchers from academic, industry, etc. MATERIALS AND METHODS: Peer-reviewed articles from most recognized journals were collected, and presented information was analyzed to discuss collection, chemical, electroporation, cellular, and membrane surface engineering to design extracellular vesicles for various therapeutic applications. In addition, we present the applications and limitations of techniques for the collection of extracellular vesicles. KEY FINDINGS There is a need for isolation techniques with the gold standard. However, advanced extracellular vesicle isolation techniques showed improved recovery, and purity of extracellular vesicles. Tumor therapy is a major part of the therapy section that illustrates the role of engineered extracellular vesicles in synergetic therapy such as phototherapy, theragnostic, and delivery of genetic materials. In addition, extracellular vesicles have shown their potential in the treatment of retinal disorders, neurodegenerative disease, tuberculosis, osteoporosis, inflammatory bowel disease, vaccine production, and wound healing. SIGNIFICANCE Engineered extracellular vesicles can deliver cargo to the specific cells, elicit an immune response and could be used for the development of the vaccines in the future. However, the progress is at the initial stage. Overall, this review will provide a comprehensive understanding and could serve as a reference for researchers in the clinical translation of engineered extracellular vesicles in different biomedical fields.
Collapse
Affiliation(s)
- Farhan Mazahir
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Raebareli, A Transit Campus, Bijnor-Sisendi Road, Bijnor, Lucknow-226002, India
| | - Awesh K Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Raebareli, A Transit Campus, Bijnor-Sisendi Road, Bijnor, Lucknow-226002, India.
| |
Collapse
|
37
|
Zheng W, Chai P, Zhu J, Zhang K. High-resolution in situ structures of mammalian respiratory supercomplexes. Nature 2024; 631:232-239. [PMID: 38811722 PMCID: PMC11222160 DOI: 10.1038/s41586-024-07488-9] [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: 10/26/2023] [Accepted: 04/30/2024] [Indexed: 05/31/2024]
Abstract
Mitochondria play a pivotal part in ATP energy production through oxidative phosphorylation, which occurs within the inner membrane through a series of respiratory complexes1-4. Despite extensive in vitro structural studies, determining the atomic details of their molecular mechanisms in physiological states remains a major challenge, primarily because of loss of the native environment during purification. Here we directly image porcine mitochondria using an in situ cryo-electron microscopy approach. This enables us to determine the structures of various high-order assemblies of respiratory supercomplexes in their native states. We identify four main supercomplex organizations: I1III2IV1, I1III2IV2, I2III2IV2 and I2III4IV2, which potentially expand into higher-order arrays on the inner membranes. These diverse supercomplexes are largely formed by 'protein-lipids-protein' interactions, which in turn have a substantial impact on the local geometry of the surrounding membranes. Our in situ structures also capture numerous reactive intermediates within these respiratory supercomplexes, shedding light on the dynamic processes of the ubiquinone/ubiquinol exchange mechanism in complex I and the Q-cycle in complex III. Structural comparison of supercomplexes from mitochondria treated under different conditions indicates a possible correlation between conformational states of complexes I and III, probably in response to environmental changes. By preserving the native membrane environment, our approach enables structural studies of mitochondrial respiratory supercomplexes in reaction at high resolution across multiple scales, from atomic-level details to the broader subcellular context.
Collapse
Affiliation(s)
- Wan Zheng
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Pengxin Chai
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Jiapeng Zhu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Kai Zhang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA.
| |
Collapse
|
38
|
Kaur M, Fusco S, Van den Broek B, Aseervatham J, Rostami A, Iacovitti L, Grassi C, Lukomska B, Srivastava AK. Most recent advances and applications of extracellular vesicles in tackling neurological challenges. Med Res Rev 2024; 44:1923-1966. [PMID: 38500405 DOI: 10.1002/med.22035] [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: 01/02/2024] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
Over the past few decades, there has been a notable increase in the global burden of central nervous system (CNS) diseases. Despite advances in technology and therapeutic options, neurological and neurodegenerative disorders persist as significant challenges in treatment and cure. Recently, there has been a remarkable surge of interest in extracellular vesicles (EVs) as pivotal mediators of intercellular communication. As carriers of molecular cargo, EVs demonstrate the ability to traverse the blood-brain barrier, enabling bidirectional communication. As a result, they have garnered attention as potential biomarkers and therapeutic agents, whether in their natural form or after being engineered for use in the CNS. This review article aims to provide a comprehensive introduction to EVs, encompassing various aspects such as their diverse isolation methods, characterization, handling, storage, and different routes for EV administration. Additionally, it underscores the recent advances in their potential applications in neurodegenerative disorder therapeutics. By exploring their unique capabilities, this study sheds light on the promising future of EVs in clinical research. It considers the inherent challenges and limitations of these emerging applications while incorporating the most recent updates in the field.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Bram Van den Broek
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jaya Aseervatham
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lorraine Iacovitti
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Jefferson Stem Cell and Regenerative Neuroscience Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Amit K Srivastava
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Yang Y, Valencia LA, Lu CH, Nakamoto ML, Tsai CT, Liu C, Yang H, Zhang W, Jahed Z, Lee WR, Santoro F, Liou J, Wu JC, Cui B. Membrane Curvature Promotes ER-PM Contact Formation via Junctophilin-EHD Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.29.601287. [PMID: 38979311 PMCID: PMC11230412 DOI: 10.1101/2024.06.29.601287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Contact sites between the endoplasmic reticulum (ER) and the plasma membrane (PM) play a crucial role in governing calcium regulation and lipid homeostasis. Despite their significance, the factors regulating their spatial distribution on the PM remain elusive. Inspired by observations in cardiomyocytes, where ER-PM contact sites concentrate on tubular PM invaginations known as transverse tubules (T-tubules), we hypothesize that the PM curvature plays a role in ER-PM contact formation. Through precise control of PM invaginations, we show that PM curvatures locally induce the formation of ER-PM contacts in cardiomyocytes. Intriguingly, the junctophilin family of ER-PM tethering proteins, specifically expressed in excitable cells, is the key player in this process, while the ubiquitously expressed extended synaptotagmin 2 does not show a preference for PM curvature. At the mechanistic level, we find that the low complexity region (LCR) and the MORN motifs of junctophilins can independently bind to the PM, but both the LCR and MORN motifs are required for targeting PM curvatures. By examining the junctophilin interactome, we identify a family of curvature-sensing proteins, Eps15-homology domain containing proteins (EHDs), that interact with the MORN_LCR motifs and facilitate junctophilins' preferential tethering to curved PM. These findings highlight the pivotal role of PM curvature in the formation of ER-PM contacts in cardiomyocytes and unveil a novel mechanism for the spatial regulation of ER-PM contacts through PM curvature modulation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Luis A. Valencia
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Melissa L. Nakamoto
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Present address: Department of Physiology and Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Present address: Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Wei Zhang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Zeinab Jahed
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Present address: Department of Nanoengineering, Jacobs School of Engineering, University of California, San Diego, CA, USA
| | - Wan-Ru Lee
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Faculty of Electrical Engineering and IT, RWTH, Aachen 52074, Germany
- Institute of Biological Information Processing—Bioelectronics, IBI-3, Forschungszentrum, Juelich 52428, Germany
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Medicine (Division of Cardiology), Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| |
Collapse
|
40
|
Prever L, Squillero G, Hirsch E, Gulluni F. Linking phosphoinositide function to mitosis. Cell Rep 2024; 43:114273. [PMID: 38843397 DOI: 10.1016/j.celrep.2024.114273] [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: 03/18/2024] [Revised: 04/12/2024] [Accepted: 05/09/2024] [Indexed: 07/02/2024] Open
Abstract
Phosphoinositides (PtdIns) are a family of differentially phosphorylated lipid second messengers localized to the cytoplasmic leaflet of both plasma and intracellular membranes. Kinases and phosphatases can selectively modify the PtdIns composition of different cellular compartments, leading to the recruitment of specific binding proteins, which control cellular homeostasis and proliferation. Thus, while PtdIns affect cell growth and survival during interphase, they are also emerging as key drivers in multiple temporally defined membrane remodeling events of mitosis, like cell rounding, spindle orientation, cytokinesis, and abscission. In this review, we summarize and discuss what is known about PtdIns function during mitosis and how alterations in the production and removal of PtdIns can interfere with proper cell division.
Collapse
Affiliation(s)
- Lorenzo Prever
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", Via Nizza 52, 10126 Turin, Italy
| | - Gabriele Squillero
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", Via Nizza 52, 10126 Turin, Italy
| | - Emilio Hirsch
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", Via Nizza 52, 10126 Turin, Italy.
| | - Federico Gulluni
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", Via Nizza 52, 10126 Turin, Italy.
| |
Collapse
|
41
|
Díaz M. Multifactor Analyses of Frontal Cortex Lipids in the APP/PS1 Model of Familial Alzheimer's Disease Reveal Anomalies in Responses to Dietary n-3 PUFA and Estrogenic Treatments. Genes (Basel) 2024; 15:810. [PMID: 38927745 PMCID: PMC11202691 DOI: 10.3390/genes15060810] [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: 04/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Brain lipid homeostasis is an absolute requirement for proper functionality of nerve cells and neurological performance. Current evidence demonstrates that lipid alterations are linked to neurodegenerative diseases, especially Alzheimer's disease (AD). The complexity of the brain lipidome and its metabolic regulation has hampered the identification of critical processes associated with the onset and progression of AD. While most experimental studies have focused on the effects of known factors on the development of pathological hallmarks in AD, e.g., amyloid deposition, tau protein and neurofibrillary tangles, neuroinflammation, etc., studies addressing the causative effects of lipid alterations remain largely unexplored. In the present study, we have used a multifactor approach combining diets containing different amounts of polyunsaturated fatty acids (PUFAs), estrogen availabilities, and genetic backgrounds, i.e., wild type (WT) and APP/PS1 (FAD), to analyze the lipid phenotype of the frontal cortex in middle-aged female mice. First, we observed that severe n-3 PUFA deficiency impacts the brain n-3 long-chain PUFA (LCPUFA) composition, yet it was notably mitigated by hepatic de novo synthesis. n-6 LCPUFAs, ether-linked fatty acids, and saturates were also changed by the dietary condition, but the extent of changes was dependent on the genetic background and hormonal condition. Likewise, brain cortex phospholipids were mostly modified by the genotype (FAD>WT) with nuanced effects from dietary treatment. Cholesterol (but not sterol esters) was modified by the genotype (WT>FAD) and dietary condition (higher in DHA-free conditions, especially in WT mice). However, the effects of estrogen treatment were mostly observed in relation to phospholipid remodeling in a genotype-dependent manner. Analyses of lipid-derived variables indicate that nerve cell membrane biophysics were significantly affected by the three factors, with lower membrane microviscosity (higher fluidity) values obtained for FAD animals. In conclusion, our multifactor analyses revealed that the genotype, diet, and estrogen status modulate the lipid phenotype of the frontal cortex, both as independent factors and through their interactions. Altogether, the outcomes point to potential strategies based on dietary and hormonal interventions aimed at stabilizing the brain cortex lipid composition in Alzheimer's disease neuropathology.
Collapse
Affiliation(s)
- Mario Díaz
- Membrane Physiology and Biophysics, Department of Physics, School of Sciences, University of La Laguna, 38206 Tenerife, Spain; or
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38206 Tenerife, Spain
| |
Collapse
|
42
|
Dharan R, Vaknin A, Sorkin R. Extracellular domain 2 of TSPAN4 governs its functions. BIOPHYSICAL REPORTS 2024; 4:100149. [PMID: 38562622 PMCID: PMC10982557 DOI: 10.1016/j.bpr.2024.100149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024]
Abstract
Tetraspanin 4, a protein with four transmembrane helices and three connecting loops, senses membrane curvature and localizes to membrane tubes. This enrichment in tubular membranes enhances its diverse interactions. While the transmembrane part of the protein likely contributes to curvature sensitivity, the possible roles of the ectodomains in curvature sensitivity of tetraspanin 4 are still unknown. Here, using micropipette aspiration combined with confocal microscopy and optical tweezers, we show that the extracellular loop 2 contributes to the curvature sensitivity and curvature-induced interactions of tetraspanin 4. To this end, we created truncated tetraspanin 4 mutants by deleting each of the connecting loops. Subsequently, we pulled membrane tubes from giant plasma membrane vesicles containing tetraspanin 4-GFP or its mutants while maintaining controllable membrane tension and curvature. Among the mutations tested, the removal of the extracellular loop 2 had the most significant impact on both the curvature sensitivity and interactions of tetraspanin 4. Based on the results, we suggest that the extracellular loop 2 regulates the affinity of tetraspanin 4 towards curved membranes and affects its lateral interactions.
Collapse
Affiliation(s)
- Raviv Dharan
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Alisa Vaknin
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Raya Sorkin
- School of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
43
|
Kushwaha S, Mallik B, Bisht A, Mushtaq Z, Pippadpally S, Chandra N, Das S, Ratnaparkhi G, Kumar V. dAsap regulates cellular protrusions via an Arf6-dependent actin regulatory pathway in S2R+ cells. FEBS Lett 2024; 598:1491-1505. [PMID: 38862211 DOI: 10.1002/1873-3468.14954] [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: 05/05/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024]
Abstract
Membrane protrusions are fundamental to cellular functions like migration, adhesion, and communication and depend upon dynamic reorganization of the cytoskeleton. GAP-dependent GTP hydrolysis of Arf proteins regulates actin-dependent membrane remodeling. Here, we show that dAsap regulates membrane protrusions in S2R+ cells by a mechanism that critically relies on its ArfGAP domain and relocalization of actin regulators, SCAR, and Ena. While our data reinforce the preference of dAsap for Arf1 GTP hydrolysis in vitro, we demonstrate that induction of membrane protrusions in S2R+ cells depends on Arf6 inactivation. This study furthers our understanding of how dAsap-dependent GTP hydrolysis maintains a balance between active and inactive states of Arf6 to regulate cell shape.
Collapse
Affiliation(s)
- Shikha Kushwaha
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, India
| | - Bhagaban Mallik
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, India
| | - Anjali Bisht
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, India
| | - Zeeshan Mushtaq
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, India
| | - Srikanth Pippadpally
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, India
| | - Nitika Chandra
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, India
| | - Subhradip Das
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Pune, India
| | - Girish Ratnaparkhi
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Pune, India
| | - Vimlesh Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, India
| |
Collapse
|
44
|
Huster D, Maiti S, Herrmann A. Phospholipid Membranes as Chemically and Functionally Tunable Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312898. [PMID: 38456771 DOI: 10.1002/adma.202312898] [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: 11/29/2023] [Revised: 02/12/2024] [Indexed: 03/09/2024]
Abstract
The sheet-like lipid bilayer is the fundamental structural component of all cell membranes. Its building blocks are phospholipids and cholesterol. Their amphiphilic structure spontaneously leads to the formation of a bilayer in aqueous environment. Lipids are not just structural elements. Individual lipid species, the lipid membrane structure, and lipid dynamics influence and regulate membrane protein function. An exciting field is emerging where the membrane-associated material properties of different bilayer systems are used in designing innovative solutions for widespread applications across various fields, such as the food industry, cosmetics, nano- and biomedicine, drug storage and delivery, biotechnology, nano- and biosensors, and computing. Here, the authors summarize what is known about how lipids determine the properties and functions of biological membranes and how this has been or can be translated into innovative applications. Based on recent progress in the understanding of membrane structure, dynamics, and physical properties, a perspective is provided on how membrane-controlled regulation of protein functions can extend current applications and even offer new applications.
Collapse
Affiliation(s)
- Daniel Huster
- Institute of Medical Physics and Biophysics, University of Leipzig, Härtelstr. 16/18, D-04107, Leipzig, Germany
| | - Sudipta Maiti
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, 400 005, India
| | - Andreas Herrmann
- Freie Universität Berlin, Department Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, D-14195, Berlin, Germany
| |
Collapse
|
45
|
Mak KM, Shekhar AC. Soybean polyenylphosphatidylcholine (PPC) is beneficial in liver and extrahepatic tissue injury: An update in experimental research. Anat Rec (Hoboken) 2024; 307:2162-2186. [PMID: 37814787 DOI: 10.1002/ar.25333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/11/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023]
Abstract
Polyenylphosphatidylcholine (PPC) is a purified polyunsaturated phosphatidylcholine extract of soybeans. This article updates PPC's beneficial effects on various forms of liver cell injury and other tissues in experimental research. PPC downregulates hepatocyte CYP2E1 expression and associated hepatotoxicity, as well as attenuates oxidative stress, apoptosis, lipoprotein oxidation and steatosis in alcoholic and nonalcoholic liver injury. PPC inhibits pro-inflammatory cytokine production, while stimulating anti-inflammatory cytokine secretion in ethanol or lipopolysaccharide-stimulated Kupffer cells/macrophages. It promotes M2-type macrophage polarization and metabolic reprogramming of glucose and lipid metabolism. PPC mitigates steatosis in NAFLD through inhibiting polarization of pro-inflammatory M1-type Kupffer cells, alleviating metabolic inflammation, remodeling hepatic lipid metabolism, correcting imbalances between lipogenesis and lipolysis and enhancing lipoprotein secretion from hepatocytes. PPC is antifibrotic by preventing progression of alcoholic hepatic fibrosis in baboons and also prevents CCl4-induced fibrosis in rats. PPC supplementation replenishes the phosphatidylcholine content of damaged cell membranes, resulting in increased membrane fluidity and functioning. Phosphatidylcholine repletion prevents increased membrane curvature of the endoplasmic reticulum and Golgi and decreases sterol regulatory element binding protein-1-mediated lipogenesis, reducing steatosis. PPC remodels gut microbiota and affects hepatic lipid metabolism via the gut-hepatic-axis and also alleviates brain inflammatory responses and cognitive impairment via the gut-brain-axis. Additionally, PPC protects extrahepatic tissues from injury caused by various toxic compounds by reducing oxidative stress, inflammation, and membrane damage. It also stimulates liver regeneration, enhances sensitivity of cancer cells to radiotherapy/chemotherapy, and inhibits experimental hepatocarcinogenesis. PPC's beneficial effects justify it as a supportive treatment of liver disease.
Collapse
Affiliation(s)
- Ki M Mak
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aditya C Shekhar
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
46
|
Lewis AH, Cronin ME, Grandl J. Piezo1 ion channels are capable of conformational signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596257. [PMID: 38854150 PMCID: PMC11160644 DOI: 10.1101/2024.05.28.596257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Piezo1 is a mechanically activated ion channel that senses forces with short latency and high sensitivity. Piezos undergo large conformational changes, induce far-reaching deformation onto the membrane, and modulate the function of two-pore potassium (K2P) channels. Taken together, this led us to hypothesize that Piezos may be able to signal their conformational state to other nearby proteins. Here, we use chemical control to acutely restrict Piezo1 conformational flexibility and show that Piezo1 conformational changes, but not ion permeation through it, are required for modulating the K2P channel TREK1. Super-resolution imaging and stochastic simulations further reveal that both channels do not co-localize, which implies that modulation is not mediated through direct binding interactions; however, at high Piezo1 densities, most TREK1 channels are within the predicted Piezo1 membrane footprint, suggesting the footprint may underlie conformational signaling. We speculate that physiological roles originally attributed to Piezo1 ionotropic function could, alternatively, involve conformational signaling.
Collapse
Affiliation(s)
- Amanda H. Lewis
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Marie E. Cronin
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jörg Grandl
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
47
|
Gao J, Hou R, Hu W, Weikl TR, Hu J. Which Coverages of Arc-Shaped Proteins Are Required for Membrane Tubulation? J Phys Chem B 2024; 128:4735-4740. [PMID: 38706129 DOI: 10.1021/acs.jpcb.4c01019] [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: 05/07/2024]
Abstract
Arc-shaped BIN/Amphiphysin/Rvs (BAR) domain proteins generate curvature by binding to membranes and induce membrane tubulation at sufficiently large protein coverages. For the amphiphysin N-BAR domain, Le Roux et al., Nat. Commun. 2021, 12, 6550, measured a threshold coverage of 0.44 ± 0.097 for nanotubules emerging from the supported lipid bilayer. In this article, we systematically investigate membrane tubulation induced by arc-shaped protein-like particles with coarse-grained modeling and simulations and determine the threshold coverages at different particle-particle interaction strengths and membrane spontaneous curvatures. In our simulations, the binding of arc-shaped particles induces a membrane shape transition from spherical vesicles to tubules at a particle threshold coverage of about 0.5, which is rather robust to variations of the direct attractive particle interactions or spontaneous membrane curvature in the coarse-grained model. Our study suggests that threshold coverages of around or slightly below 0.5 are a general requirement for membrane tubulation by arc-shaped BAR domain proteins.
Collapse
Affiliation(s)
- Jie Gao
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ruihan Hou
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wenbing Hu
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Thomas R Weikl
- Department of Bio-Molecular Systems, Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14424 Potsdam, Germany
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
| |
Collapse
|
48
|
Johnson DH, Kou OH, Bouzos N, Zeno WF. Protein-membrane interactions: sensing and generating curvature. Trends Biochem Sci 2024; 49:401-416. [PMID: 38508884 PMCID: PMC11069444 DOI: 10.1016/j.tibs.2024.02.005] [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: 09/23/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/22/2024]
Abstract
Biological membranes are integral cellular structures that can be curved into various geometries. These curved structures are abundant in cells as they are essential for various physiological processes. However, curved membranes are inherently unstable, especially on nanometer length scales. To stabilize curved membranes, cells can utilize proteins that sense and generate membrane curvature. In this review, we summarize recent research that has advanced our understanding of interactions between proteins and curved membrane surfaces, as well as work that has expanded our ability to study curvature sensing and generation. Additionally, we look at specific examples of cellular processes that require membrane curvature, such as neurotransmission, clathrin-mediated endocytosis (CME), and organelle biogenesis.
Collapse
Affiliation(s)
- David H Johnson
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Orianna H Kou
- Department of Physics and Astronomy, University of Southern California, Los Angeles, CA 90089, USA
| | - Nicoletta Bouzos
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Wade F Zeno
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
49
|
Umegawa Y, Kato S, Seo S, Shinoda W, Kawatake S, Matsuoka S, Murata M. Protein-lipid acyl chain interactions: Depth-dependent changes of segmental mobility of phospholipid in contact with bacteriorhodopsin. Biophys Chem 2024; 308:107204. [PMID: 38412762 DOI: 10.1016/j.bpc.2024.107204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 02/29/2024]
Abstract
Boundary lipids surrounding membrane proteins play an essential role in protein function and structure. These protein-lipid interactions are mainly divided into electrostatic interactions between the polar amino acids of proteins and polar heads of phospholipids, and hydrophobic interactions between protein transmembrane sites and phospholipid acyl chains. Our previous report (Kawatake et al., Biochim. Biophys. Acta 1858 [2016] 2106-2115) covered a method for selectively analyzing boundary lipid interactions and showed differences in membrane protein-peripheral lipid interactions due to differences in their head group. Interactions in the hydrophobic acyl chains of phospholipids are relatively consistent among proteins, but the details of these interactions have not been elucidated. In this study, we reconstituted bacteriorhodopsin as a model protein into phospholipid membranes labeled with 2H and 13C for solid-state NMR measurement to investigate the depth-dependent effect of the head group structure on the lipid bilayer. The results showed that the position of the phospholipid near the carbonyl carbon was affected by the head group in terms of selectivity for protein surfaces, whereas in the deep interior of the bilayer near the leaflet interface, there was little difference between the head groups, indicating that the dependence of their interactions on the head group was much reduced.
Collapse
Affiliation(s)
- Yuichi Umegawa
- JST-ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
| | - Sho Kato
- JST-ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Sangjae Seo
- Department of Materials Chemistry, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Division of Supercomputing, Korea Institute of Science and Technology Information, 245 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Wataru Shinoda
- Department of Materials Chemistry, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Research Institute for Interdisciplinary Science, Okayama University, 3-1-1, Tsushima-naka, Okayama 700-8530, Japan
| | - Satoshi Kawatake
- JST-ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Shigeru Matsuoka
- JST-ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Michio Murata
- JST-ERATO, Lipid Active Structure Project, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
50
|
Lin SZ, Prost J, Rupprecht JF. Curvature-induced clustering of cell adhesion proteins. Phys Rev E 2024; 109:054406. [PMID: 38907394 DOI: 10.1103/physreve.109.054406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/02/2024] [Indexed: 06/24/2024]
Abstract
Cell adhesion proteins typically form stable clusters that anchor the cell membrane to its environment. Several works have suggested that cell membrane protein clusters can emerge from a local feedback between the membrane curvature and the density of proteins. Here, we investigate the effect of such a curvature-sensing mechanism in the context of cell adhesion proteins. We show how clustering emerges in an intermediate range of adhesion and curvature-sensing strengths. We identify key differences with the tilt-induced gradient sensing mechanism we previously proposed (Lin et al., arXiv:2307.03670).
Collapse
Affiliation(s)
- Shao-Zhen Lin
- Aix Marseille Univ, Université de Toulon, CNRS, CPT (UMR 7332), Turing Centre for Living systems, Marseille, France
| | - Jacques Prost
- Laboratoire Physico-Chimie Curie, UMR 168, Institut Curie, PSL Research University, CNRS, Sorbonne Université, 75005 Paris, France
- Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Jean-François Rupprecht
- Aix Marseille Univ, Université de Toulon, CNRS, CPT (UMR 7332), Turing Centre for Living systems, Marseille, France
| |
Collapse
|