1
|
Wu F, Bettiga M, Olsson L. Exploring the interplay between yeast cell membrane lipid adaptation and physiological response to acetic acid stress. Appl Environ Microbiol 2024; 90:e0121224. [PMID: 39535190 DOI: 10.1128/aem.01212-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024] Open
Abstract
Acetic acid is a byproduct of lignocellulose pretreatment and a potent inhibitor of yeast-based fermentation processes. A thicker yeast plasma membrane (PM) is expected to retard the passive diffusion of undissociated acetic acid into the cell. Molecular dynamic simulations suggest that membrane thickness can be increased by elongating glycerophospholipids (GPL) fatty acyl chains. Previously, we successfully engineered Saccharomyces cerevisiae to increase GPL fatty acyl chain length but failed to lower acetic acid net uptake. Here, we tested whether altering the relative abundance of diacylglycerol (DAG) might affect PM permeability to acetic acid in cells with longer GPL acyl chains (DAGEN). To this end, we expressed diacylglycerol kinase α (DGKα) in DAGEN. The resulting DAGEN_Dgkα strain exhibited restored DAG levels, grew in medium containing 13 g/L acetic acid, and accumulated less acetic acid. Acetic acid stress and energy burden were accompanied by increased glucose uptake in DAGEN_Dgkα cells. Compared to DAGEN, the relative abundance of several membrane lipids changed in DAGEN_Dgkα in response to acetic acid stress. We propose that the ability to increase the energy supply and alter membrane lipid composition could compensate for the negative effect of high net acetic acid uptake in DAGEN_Dgkα under stressful conditions. IMPORTANCE In the present study, we successfully engineered a yeast strain that could grow under high acetic acid stress by regulating its diacylglycerol metabolism. We compared how the plasma membrane and total cell membranes responded to acetic acid by adjusting their lipid content. By combining physiological and lipidomics analyses in cells cultivated in the absence or presence of acetic acid, we found that the capacity of the membrane to adapt lipid composition together with sufficient energy supply influenced membrane properties in response to stress. We suggest that potentiating the intracellular energy system or enhancing lipid transport to destination membranes should be taken into account when designing membrane engineering strategies. The findings highlight new directions for future yeast cell factory engineering.
Collapse
Affiliation(s)
- Fei Wu
- Department of Life Sciences, Division of Industrial Biotechnology, Chalmers University of Technology, Gothenburg, Sweden
| | - Maurizio Bettiga
- Department of Life Sciences, Division of Industrial Biotechnology, Chalmers University of Technology, Gothenburg, Sweden
- Italbiotec Srl Benefit Corporation, Innovation Unit, Milan, Italy
| | - Lisbeth Olsson
- Department of Life Sciences, Division of Industrial Biotechnology, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
2
|
Kurakin S, Ivankov O, Dushanov E, Murugova T, Ermakova E, Efimov S, Mukhametzyanov T, Smerdova S, Klochkov V, Kuklin A, Kučerka N. Calcium ions do not influence the Aβ(25-35) triggered morphological changes of lipid membranes. Biophys Chem 2024; 313:107292. [PMID: 39018778 DOI: 10.1016/j.bpc.2024.107292] [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: 03/06/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
We have studied the effect of calcium ions (Ca2+) at various concentrations on the structure of lipid vesicles in the presence of amyloid-beta peptide Aβ(25-35). In particular, we have investigated the influence of calcium ions on the formation of recently documented bicelle-like structures (BLSs) emerged as a result of Aβ(25-35) triggered membrane disintegration. First, we have shown by using small-angle X-ray and neutron scattering that peptide molecules rigidify the lipid bilayer of gel phase DPPC unilamellar vesicles (ULVs), while addition of the calcium ions to the system hinders this effect of Aβ(25-35). Secondly, the Aβ(25-35) demonstrates a critical peptide concentration at which the BLSs reorganize from ULVs due to heating and cooling the samples through the lipid main phase transition temperature (Tm). However, addition of calcium ions does not affect noticeably the Aβ-induced formation of BLSs and their structural parameters, though the changes in peptide's secondary structure, e.g. the increased α-helix fraction, has been registered by circular dichroism spectroscopy. Finally, according to 31P nuclear magnetic resonance (NMR) measurements, calcium ions do not affect the lipid-peptide arrangement in BLSs and their ability to align in the magnetic field of NMR spectrometer. The influences of various concentrations of calcium ions on the lipid-peptide interactions may prove biologically important because their local concentrations vary widely in in-vivo conditions. In the present work, calcium ions were investigated as a possible tool aimed at regulating the lipid-peptide interactions that demonstrated the disruptive effect of Aβ(25-35) on lipid membranes.
Collapse
Affiliation(s)
- Sergei Kurakin
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia.
| | - Oleksandr Ivankov
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Ermuhammad Dushanov
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Department of Biophysics, Dubna State University, Universitetskaya 19, Dubna, Moscow Region 141982, Russia
| | - Tatiana Murugova
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Elena Ermakova
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Sergey Efimov
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Timur Mukhametzyanov
- Butlerov Chemistry Institute, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Svetlana Smerdova
- Kazan National Research Technological University, Karl Marx 68, Kazan 420015, Russia
| | - Vladimir Klochkov
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Alexander Kuklin
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Moscow Institute of Physics and Technology, Instytutskiy Pereulok 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Norbert Kučerka
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, Bratislava 832 32, Slovakia.
| |
Collapse
|
3
|
Papadopoulou P, Arias-Alpizar G, Weeda P, Poppe T, van Klaveren N, Slíva T, Aschmann D, van Os W, Zhang Y, Moradi MA, Sommerdijk N, Campbell F, Kros A. Structure-function relationship of phase-separated liposomes containing diacylglycerol analogues. Biomater Sci 2024; 12:5023-5035. [PMID: 39177657 DOI: 10.1039/d4bm00799a] [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: 08/24/2024]
Abstract
The composition and morphology of lipid-based nanoparticles can influence their overall in vivo behavior. Previously, we demonstrated that phase separation in liposomes composed of DSPC and a diacylglycerol lipid analogue (DOaG) drives the in vivo biodistribution towards a specific subset of endothelial cells in zebrafish embryos. In the absence of traditional targeting functionalities (e.g., antibodies, ligands), this selectivity is mediated solely by the unique liposome morphology and composition, characterized by a DOaG-rich lipid droplet within the DSPC-rich phospholipid bilayer. The phase separation is induced due to the geometry of DOaG lipid and its ability to create non-bilayer phases in lipid membranes. To investigate the underlying principles of phase separation and to optimize the liposome colloidal stability, we performed a structure-function relationship study by synthesizing a library of DOaG analogues with varying molecular properties, such as the number, length and sn-position of the acyl chains, as well as the degree of saturation or carbonyl substituents. We assessed the ability of these lipid analogues to assemble into phase-separated liposomes and studied their morphology, colloidal stability, and in vivo biodistribution in zebrafish embryos. We found that analogues containing unsaturated, medium length (C16-C18) fatty acids were required to obtain colloidally stable, phase-separated liposomes with cell-specific biodistribution patterns. Moreover, we observed that using the pure DOaG isomer, with acyl chains at the sn-1,3 positions, leads to more colloidally stable liposomes than when a mixture of sn-1,2 and sn-1,3 isomers is used. Similarly, we observed that incorporating a DOaG analogue with fatty tails shorter than DSPC, as well as PEGylation, endows liposomes with long term stability while retaining cell-selective biodistribution. Diacylglycerols are known to promote fusion, lipid polymorphism, signaling and protein recruitment on lipid membranes. In this study, we showed that diacylglycerol derivatives can induce phase separation in liposomes, unlocking the potential for cell-specific targeting in vivo. We believe that these findings can be the foundation for future use of diacylglycerols in lipid-based nanomedicines and could lead to the development of novel targeted delivery strategies.
Collapse
Affiliation(s)
- Panagiota Papadopoulou
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Gabriela Arias-Alpizar
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Pim Weeda
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Thijs Poppe
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Niels van Klaveren
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Tomas Slíva
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Dennis Aschmann
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Winant van Os
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Yun Zhang
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Mohammad-Amin Moradi
- Materials and Interface Chemistry, Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Nico Sommerdijk
- Electron Microscopy Center, Radboud Technology Center Microscopy, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Biochemistry, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
4
|
Raab JE, Hamilton DJ, Harju TB, Huynh TN, Russo BC. Pushing boundaries: mechanisms enabling bacterial pathogens to spread between cells. Infect Immun 2024; 92:e0052423. [PMID: 38661369 PMCID: PMC11385730 DOI: 10.1128/iai.00524-23] [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: 04/26/2024] Open
Abstract
For multiple intracellular bacterial pathogens, the ability to spread directly into adjacent epithelial cells is an essential step for disease in humans. For pathogens such as Shigella, Listeria, Rickettsia, and Burkholderia, this intercellular movement frequently requires the pathogens to manipulate the host actin cytoskeleton and deform the plasma membrane into structures known as protrusions, which extend into neighboring cells. The protrusion is then typically resolved into a double-membrane vacuole (DMV) from which the pathogen quickly escapes into the cytosol, where additional rounds of intercellular spread occur. Significant progress over the last few years has begun to define the mechanisms by which intracellular bacterial pathogens spread. This review highlights the interactions of bacterial and host factors that drive mechanisms required for intercellular spread with a focus on how protrusion structures form and resolve.
Collapse
Affiliation(s)
- Julie E. Raab
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Desmond J. Hamilton
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Tucker B. Harju
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Thao N. Huynh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Brian C. Russo
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| |
Collapse
|
5
|
Caselli L, Conti L, De Santis I, Berti D. Small-angle X-ray and neutron scattering applied to lipid-based nanoparticles: Recent advancements across different length scales. Adv Colloid Interface Sci 2024; 327:103156. [PMID: 38643519 DOI: 10.1016/j.cis.2024.103156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/28/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024]
Abstract
Lipid-based nanoparticles (LNPs), ranging from nanovesicles to non-lamellar assemblies, have gained significant attention in recent years, as versatile carriers for delivering drugs, vaccines, and nutrients. Small-angle scattering methods, employing X-rays (SAXS) or neutrons (SANS), represent unique tools to unveil structure, dynamics, and interactions of such particles on different length scales, spanning from the nano to the molecular scale. This review explores the state-of-the-art on scattering methods applied to unveil the structure of lipid-based nanoparticles and their interactions with drugs and bioactive molecules, to inform their rational design and formulation for medical applications. We will focus on complementary information accessible with X-rays or neutrons, ranging from insights on the structure and colloidal processes at a nanoscale level (SAXS) to details on the lipid organization and molecular interactions of LNPs (SANS). In addition, we will review new opportunities offered by Time-resolved (TR)-SAXS and -SANS for the investigation of dynamic processes involving LNPs. These span from real-time monitoring of LNPs structural evolution in response to endogenous or external stimuli (TR-SANS), to the investigation of the kinetics of lipid diffusion and exchange upon interaction with biomolecules (TR-SANS). Finally, we will spotlight novel combinations of SAXS and SANS with complementary on-line techniques, recently enabled at Large Scale Facilities for X-rays and neutrons. This emerging technology enables synchronized multi-method investigation, offering exciting opportunities for the simultaneous characterization of the structure and chemical or mechanical properties of LNPs.
Collapse
Affiliation(s)
- Lucrezia Caselli
- Physical Chemistry 1, University of Lund, S-221 00 Lund, Sweden.
| | - Laura Conti
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Ilaria De Santis
- Department of Chemistry, University of Florence, Via della Lastruccia 3, Sesto Fiorentino, Florence 50019, Italy
| | - Debora Berti
- Department of Chemistry, University of Florence, Via della Lastruccia 3, Sesto Fiorentino, Florence 50019, Italy; Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy.
| |
Collapse
|
6
|
Papadopoulou P, van der Pol R, van Hilten N, van Os WL, Pattipeiluhu R, Arias-Alpizar G, Knol RA, Noteborn W, Moradi MA, Ferraz MJ, Aerts JMFG, Sommerdijk N, Campbell F, Risselada HJ, Sevink GJA, Kros A. Phase-Separated Lipid-Based Nanoparticles: Selective Behavior at the Nano-Bio Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310872. [PMID: 37988682 DOI: 10.1002/adma.202310872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Indexed: 11/23/2023]
Abstract
The membrane-protein interface on lipid-based nanoparticles influences their in vivo behavior. Better understanding may evolve current drug delivery methods toward effective targeted nanomedicine. Previously, the cell-selective accumulation of a liposome formulation in vivo is demonstrated, through the recognition of lipid phase-separation by triglyceride lipases. This exemplified how liposome morphology and composition can determine nanoparticle-protein interactions. Here, the lipase-induced compositional and morphological changes of phase-separated liposomes-which bear a lipid droplet in their bilayer- are investigated, and the mechanism upon which lipases recognize and bind to the particles is unravelled. The selective lipolytic degradation of the phase-separated lipid droplet is observed, while nanoparticle integrity remains intact. Next, the Tryptophan-rich loop of the lipase is identified as the region with which the enzymes bind to the particles. This preferential binding is due to lipid packing defects induced on the liposome surface by phase separation. In parallel, the existing knowledge that phase separation leads to in vivo selectivity, is utilized to generate phase-separated mRNA-LNPs that target cell-subsets in zebrafish embryos, with subsequent mRNA delivery and protein expression. Together, these findings can expand the current knowledge on selective nanoparticle-protein communications and in vivo behavior, aspects that will assist to gain control of lipid-based nanoparticles.
Collapse
Affiliation(s)
- Panagiota Papadopoulou
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Rianne van der Pol
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Niek van Hilten
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Winant L van Os
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Roy Pattipeiluhu
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Gabriela Arias-Alpizar
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Renzo Aron Knol
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Willem Noteborn
- NeCEN, Leiden University, Einsteinweg 55, Leiden, 2333 AL, The Netherlands
| | - Mohammad-Amin Moradi
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P. O. Box 513, Eindhoven, 5600 MB, The Netherlands
| | - Maria Joao Ferraz
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | | | - Nico Sommerdijk
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P. O. Box 513, Eindhoven, 5600 MB, The Netherlands
- Department of Medical BioSciences and Radboud Technology Center - Electron Microscopy, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Herre Jelger Risselada
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
- Department of Physics, Technical University Dortmund, 44221, Dortmund, Germany
| | - Geert Jan Agur Sevink
- Department of Biophysical Organic Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| |
Collapse
|
7
|
Dustin ML. Recent advances in understanding TCR signaling: a synaptic perspective. Fac Rev 2023; 12:25. [PMID: 37900153 PMCID: PMC10608137 DOI: 10.12703/r/12-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023] Open
Abstract
The T cell receptor is a multi-subunit complex that carries out a range of recognition tasks for multiple lymphocyte types and translates recognition into signals that regulate survival, growth, differentiation, and effector functions for innate and adaptive host defense. Recent advances include the cryo-electron microscopy-based structure of the extracellular and transmembrane components of the complex, new information about coupling to intracellular partners, lateral associations in the membrane that all add to our picture of the T cell signaling machinery, and how signal termination relates to effector function. This review endeavors to integrate structural and biochemical information through the lens of the immunological synapse- the critical interface with the antigen-presenting cell.
Collapse
Affiliation(s)
- Michael L Dustin
- Kennedy Institute of Rheumatology, The University of Oxford, Oxford, UK
| |
Collapse
|
8
|
Petrišič N, Adamek M, Kežar A, Hočevar SB, Žagar E, Anderluh G, Podobnik M. Structural basis for the unique molecular properties of broad-range phospholipase C from Listeria monocytogenes. Nat Commun 2023; 14:6474. [PMID: 37838694 PMCID: PMC10576769 DOI: 10.1038/s41467-023-42134-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/28/2023] [Indexed: 10/16/2023] Open
Abstract
Listeriosis is one of the most serious foodborne diseases caused by the intracellular bacterium Listeria monocytogenes. Its two major virulence factors, broad-range phospholipase C (LmPC-PLC) and the pore-forming toxin listeriolysin O (LLO), enable the bacterium to spread in the host by destroying cell membranes. Here, we determine the crystal structure of LmPC-PLC and complement it with the functional analysis of this enzyme. This reveals that LmPC-PLC has evolved several structural features to regulate its activity, including the invariant position of the N-terminal tryptophan (W1), the structurally plastic active site, Zn2+-dependent activity, and the tendency to form oligomers with impaired enzymatic activity. We demonstrate that the enzymatic activity of LmPC-PLC can be specifically inhibited by its propeptide added in trans. Furthermore, we show that the phospholipase activity of LmPC-PLC facilitates the pore-forming activity of LLO and affects the morphology of LLO oligomerization on lipid membranes, revealing the multifaceted synergy of the two virulence factors.
Collapse
Affiliation(s)
- Nejc Petrišič
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
- PhD Program 'Biosciences', Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Maksimiljan Adamek
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Andreja Kežar
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Samo B Hočevar
- Department of Analytical Chemistry, National Institute of Chemistry, Ljubljana, Slovenia
| | - Ema Žagar
- Department of Polymer Chemistry and Technology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| |
Collapse
|
9
|
Arias‐Alpizar G, Papadopoulou P, Rios X, Pulagam KR, Moradi M, Pattipeiluhu R, Bussmann J, Sommerdijk N, Llop J, Kros A, Campbell F. Phase-Separated Liposomes Hijack Endogenous Lipoprotein Transport and Metabolism Pathways to Target Subsets of Endothelial Cells In Vivo. Adv Healthc Mater 2023; 12:e2202709. [PMID: 36565694 PMCID: PMC11469146 DOI: 10.1002/adhm.202202709] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Plasma lipid transport and metabolism are essential to ensure correct cellular function throughout the body. Dynamically regulated in time and space, the well-characterized mechanisms underpinning plasma lipid transport and metabolism offers an enticing, but as yet underexplored, rationale to design synthetic lipid nanoparticles with inherent cell/tissue selectivity. Herein, a systemically administered liposome formulation, composed of just two lipids, that is capable of hijacking a triglyceride lipase-mediated lipid transport pathway resulting in liposome recognition and uptake within specific endothelial cell subsets is described. In the absence of targeting ligands, liposome-lipase interactions are mediated by a unique, phase-separated ("parachute") liposome morphology. Within the embryonic zebrafish, selective liposome accumulation is observed at the developing blood-brain barrier. In mice, extensive liposome accumulation within the liver and spleen - which is reduced, but not eliminated, following small molecule lipase inhibition - supports a role for endothelial lipase but highlights these liposomes are also subject to significant "off-target" by reticuloendothelial system organs. Overall, these compositionally simplistic liposomes offer new insights into the discovery and design of lipid-based nanoparticles that can exploit endogenous lipid transport and metabolism pathways to achieve cell selective targeting in vivo.
Collapse
Affiliation(s)
- Gabriela Arias‐Alpizar
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Division of BioTherapeuticsLeiden Academic Centre for Drug ResearchLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Panagiota Papadopoulou
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Xabier Rios
- CIC biomaGUNEBasque Research and Technology Alliance (BRTA)San Sebastián20014Spain
| | | | - Mohammad‐Amin Moradi
- Materials and Interface ChemistryDepartment of Chemical Engineering and ChemistryEindhoven University of TechnologyP.O. Box 513Eindhoven5600The Netherlands
| | - Roy Pattipeiluhu
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Jeroen Bussmann
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Division of BioTherapeuticsLeiden Academic Centre for Drug ResearchLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Nico Sommerdijk
- Department of BiochemistryRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegen6525The Netherlands
- Electron Microscopy CentreRadboudumc Technology Center MicroscopyRadboud University Medical CenterGeert Grooteplein Zuid 28Nijmegen6525The Netherlands
| | - Jordi Llop
- Materials and Interface ChemistryDepartment of Chemical Engineering and ChemistryEindhoven University of TechnologyP.O. Box 513Eindhoven5600The Netherlands
| | - Alexander Kros
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Frederick Campbell
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Present address:
NanoVation Therapeutics2405 Wesbrook Mall 4th floorVancouverBCV6T 1Z3Canada
| |
Collapse
|
10
|
Lowe LA, Wang A. Preparation of Giant Vesicles with Mixed Single-Tailed and Double-Tailed Lipids. Methods Mol Biol 2023; 2622:71-85. [PMID: 36781751 DOI: 10.1007/978-1-0716-2954-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Giant vesicles are model membrane systems that can be characterized with microscopy. Whereas most giant synthetic vesicles are created with a single phospholipid species, vesicles with mixed membrane compositions, including single-tailed and double-tailed lipids, serve as more accurate models of biological membranes and also have applications in the origins of life and drug delivery fields. Here we describe several approaches that can be used to create giant vesicles with mixed lipid compositions.
Collapse
Affiliation(s)
- Lauren A Lowe
- School of Chemistry, UNSW Sydney, Sydney, NSW, Australia.,Australian Centre for Astrobiology, UNSW Sydney, Sydney, NSW, Australia
| | - Anna Wang
- School of Chemistry, UNSW Sydney, Sydney, NSW, Australia. .,Australian Centre for Astrobiology, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
11
|
Gouveia MG, Wesseler JP, Ramaekers J, Weder C, Scholten PBV, Bruns N. Polymersome-based protein drug delivery - quo vadis? Chem Soc Rev 2023; 52:728-778. [PMID: 36537575 PMCID: PMC9890519 DOI: 10.1039/d2cs00106c] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Indexed: 12/24/2022]
Abstract
Protein-based therapeutics are an attractive alternative to established therapeutic approaches and represent one of the fastest growing families of drugs. While many of these proteins can be delivered using established formulations, the intrinsic sensitivity of proteins to denaturation sometimes calls for a protective carrier to allow administration. Historically, lipid-based self-assembled structures, notably liposomes, have performed this function. After the discovery of polymersome-based targeted drug-delivery systems, which offer manifold advantages over lipid-based structures, the scientific community expected that such systems would take the therapeutic world by storm. However, no polymersome formulations have been commercialised. In this review article, we discuss key obstacles for the sluggish translation of polymersome-based protein nanocarriers into approved pharmaceuticals, which include limitations imparted by the use of non-degradable polymers, the intricacies of polymersome production methods, and the complexity of the in vivo journey of polymersomes across various biological barriers. Considering this complex subject from a polymer chemist's point of view, we highlight key areas that are worthy to explore in order to advance polymersomes to a level at which clinical trials become worthwhile and translation into pharmaceutical and nanomedical applications is realistic.
Collapse
Affiliation(s)
- Micael G Gouveia
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Justus P Wesseler
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Jobbe Ramaekers
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Christoph Weder
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Philip B V Scholten
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Nico Bruns
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
- Department of Chemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany.
| |
Collapse
|
12
|
Recent Advances in Oral Peptide or Protein-Based Drug Liposomes. Pharmaceuticals (Basel) 2022; 15:ph15091072. [PMID: 36145293 PMCID: PMC9501131 DOI: 10.3390/ph15091072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
The high physiology and low toxicity of therapeutic peptides and proteins have made them a hot spot for drug development in recent years. However, their poor oral bioavailability and unstable metabolism make their clinical application difficult. The bilayer membrane of liposomes provides protection for the drug within the compartment, and their high biocompatibility makes the drug more easily absorbed by the body. However, phospholipids—which form the membranes—are subjected to various digestive enzymes and mucosal adhesion in the digestive tract and disintegrate before absorption. Improvements in the composition of liposomes or modifying their surface can enhance the stability of the liposomes in the gastrointestinal tract. This article reviews the basic strategies for liposome preparation and surface modification that promote the oral administration of therapeutic polypeptides.
Collapse
|
13
|
Du R, Li X, Ma YH, Luo Y, Wang C, Ma Q, Lu X. Exploring Interfacial Hydrolysis of Artificial Neutral Lipid Monolayer and Bilayer Catalyzed by Phospholipase C. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:8104-8113. [PMID: 35749224 DOI: 10.1021/acs.langmuir.2c00995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Phospholipase C (PLC) represents an important type of enzymes with the feature of hydrolyzing phospholipids at the position of the glycerophosphate bond, among which PLC extracted from Bacillus cereus (BC-PLC) has been extensively studied owing to its similarity to hitherto poorly characterized mammalian analogues. This study focuses on investigating the interfacial hydrolysis mechanism of phosphatidylcholine (PC) monolayer and bilayer membranes catalyzed by BC-PLC using sum frequency generation vibrational spectroscopy (SFG-VS) and laser scanning confocal microscopy (LSCM). We found that, upon interfacial hydrolysis, BC-PLC was adsorbed onto the lipid interface and catalyzed the lipolysis with no net orientation, as evidenced by the silent amide I band, indicating that ordered PLC alignment was not a prerequisite for the enzyme activity, which is very different from what we have reported for phospholipase A1 (PLA1) and phospholipase A2 (PLA2) [Kai, S. Phys. Chem. Chem. Phys. 2018, 20(1), 63-67; Wang, F. Langmuir 2019, 35(39), 12831-12838; Zhang, F. Langmuir 2020, 36(11), 2946-2953]. For the PC monolayer, one of the two hydrolysates, phosphocholine, desorbed from the interface into the aqueous phase, while the other one, diacylglycerol (DG), stayed well packed with high order at the interface. For the PC bilayer, phosphocholine dispersed into the aqueous phase too, similar to the monolayer case; however, DG, presumably formed clusters with the unreacted lipid substrates and desorbed from the interface. With respect to both the monolayer and bilayer cases, mechanistic schematics were presented to illustrate the different interfacial hydrolysis processes. Therefore, this model experimental study in vitro provides significant molecular-level insights and contributes necessary knowledge to reveal the lipolysis kinetics with respect to PLC and lipid membranes with monolayer and bilayer structures.
Collapse
Affiliation(s)
- Rongrong Du
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Xu Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Yong-Hao Ma
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Yongsheng Luo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Chu Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Qian Ma
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, P. R. China
- Department of General Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Xiaolin Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
14
|
Barriga HM, Pence IJ, Holme MN, Doutch JJ, Penders J, Nele V, Thomas MR, Carroni M, Stevens MM. Coupling Lipid Nanoparticle Structure and Automated Single-Particle Composition Analysis to Design Phospholipase-Responsive Nanocarriers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2200839. [PMID: 35358374 PMCID: PMC7615489 DOI: 10.1002/adma.202200839] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/11/2022] [Indexed: 06/14/2023]
Abstract
Lipid nanoparticles (LNPs) are versatile structures with tunable physicochemical properties that are ideally suited as a platform for vaccine delivery and RNA therapeutics. A key barrier to LNP rational design is the inability to relate composition and structure to intracellular processing and function. Here Single Particle Automated Raman Trapping Analysis (SPARTA) is combined with small-angle X-ray and neutron scattering (SAXS/SANS) techniques to link LNP composition with internal structure and morphology and to monitor dynamic LNP-phospholipase D (PLD) interactions. This analysis demonstrates that PLD, a key intracellular trafficking mediator, can access the entire LNP lipid membrane to generate stable, anionic LNPs. PLD activity on vesicles with matched amounts of enzyme substrate is an order of magnitude lower, indicating that the LNP lipid membrane structure can be used to control enzyme interactions. This represents an opportunity to design enzyme-responsive LNP solutions for stimuli-responsive delivery and diseases where PLD is dysregulated.
Collapse
Affiliation(s)
- Hanna M.G. Barriga
- Department of Medical Biochemistry and Biophysics Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Isaac J. Pence
- Department of Materials, Department of Bioengineering,and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Margaret N. Holme
- Department of Medical Biochemistry and Biophysics Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - James J. Doutch
- ISIS Neutron and Muon Source, STFC, Rutherford Appleton Laboratory Didcot OX11 ODE, UK
| | - Jelle Penders
- Department of Materials, Department of Bioengineering,and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Valeria Nele
- Department of Materials, Department of Bioengineering,and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Michael R. Thomas
- Department of Materials, Department of Bioengineering,and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Marta Carroni
- Department of Biochemistry and Biophysics, Science for Life Laboratory Stockholm University, Stockholm 171 65, Sweden
| | - Molly M. Stevens
- Department of Medical Biochemistry and Biophysics Karolinska Institutet, Stockholm SE-171 77, Sweden
| |
Collapse
|
15
|
Villa Nova M, Gan K, Wacker MG. Biopredictive tools for the development of injectable drug products. Expert Opin Drug Deliv 2022; 19:671-684. [PMID: 35603724 DOI: 10.1080/17425247.2022.2081682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Biopredictive release tests are commonly used in the evaluation of oral medicines. They support decision-making in formulation development and allow predictions of the expected in-vivo performances. So far, there is limited experience in the application of these methodologies to injectable drug products. AREAS COVERED Parenteral drug products cover a variety of dosage forms and administration sites including subcutaneous, intramuscular, and intravenous injections. In this area, developing biopredictive and biorelevant methodologies often confronts us with unique challenges and knowledge gaps. Here, we provide a formulation-centric approach and explain the key considerations and workflow when designing biopredictive assays. Also, we outline the key role of computational methods in achieving clinical relevance and put all considerations into context using liposomal nanomedicines as an example. EXPERT OPINION Biopredictive tools are the need of the hour to exploit the tremendous opportunities of injectable drug products. A growing number of biopharmaceuticals such as peptides, proteins, and nucleic acids require different strategies and a better understanding of the influences on drug absorption. Here, our design strategy must maintain the balance of robustness and complexity required for effective formulation development.
Collapse
Affiliation(s)
- Mônica Villa Nova
- State University of Maringá, Department of Pharmacy, Maringá, Paraná, Brazil
| | - Kennard Gan
- National University of Singapore, Department of Pharmacy, Singapore
| | | |
Collapse
|
16
|
Utterström J, Barriga HMG, Holme MN, Selegård R, Stevens MM, Aili D. Peptide-Folding Triggered Phase Separation and Lipid Membrane Destabilization in Cholesterol-Rich Lipid Vesicles. Bioconjug Chem 2022; 33:736-746. [PMID: 35362952 PMCID: PMC9026255 DOI: 10.1021/acs.bioconjchem.2c00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Liposome-based drug
delivery systems are widely used to improve
drug pharmacokinetics but can suffer from slow and unspecific release
of encapsulated drugs. Membrane-active peptides, based on sequences
derived or inspired from antimicrobial peptides (AMPs), could offer
means to trigger and control the release. Cholesterol is used in most
liposomal drug delivery systems (DDS) to improve the stability of
the formulation, but the activity of AMPs on cholesterol-rich membranes
tends to be very low, complicating peptide-triggered release strategies.
Here, we show a de novo designed AMP-mimetic peptide that efficiently
triggers content release from cholesterol-containing lipid vesicles
when covalently conjugated to headgroup-functionalized lipids. Binding
to vesicles induces peptide folding and triggers a lipid phase separation,
which in the presence of cholesterol results in high local peptide
concentrations at the lipid bilayer surface and rapid content release.
We anticipate that these results will facilitate the development of
peptide-based strategies for controlling and triggering drug release
from liposomal drug delivery systems.
Collapse
Affiliation(s)
- Johanna Utterström
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, SE-581 83 Linköping, Sweden
| | - Hanna M G Barriga
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Margaret N Holme
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, SE-581 83 Linköping, Sweden
| | - Molly M Stevens
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden.,Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, SE-581 83 Linköping, Sweden
| |
Collapse
|
17
|
Nele V, Holme MN, Rashid MH, Barriga HMG, Le TC, Thomas MR, Doutch JJ, Yarovsky I, Stevens MM. Design of Lipid-Based Nanocarriers via Cation Modulation of Ethanol-Interdigitated Lipid Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:11909-11921. [PMID: 34581180 DOI: 10.1021/acs.langmuir.1c02076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Short-chain alcohols (i.e., ethanol) can induce membrane interdigitation in saturated-chain phosphatidylcholines (PCs). In this process, alcohol molecules intercalate between phosphate heads, increasing lateral separation and favoring hydrophobic interactions between opposing acyl chains, which interpenetrate forming an interdigitated phase. Unraveling mechanisms underlying the interactions between ethanol and model lipid membranes has implications for cell biology, biochemistry, and for the formulation of lipid-based nanocarriers. However, investigations of ethanol-lipid membrane systems have been carried out in deionized water, which limits their applicability. Here, using a combination of small- and wide-angle X-ray scattering, small-angle neutron scattering, and all-atom molecular dynamics simulations, we analyzed the effect of varying CaCl2 and NaCl concentrations on ethanol-induced interdigitation. We observed that while ethanol addition leads to the interdigitation of bulk phase 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) bilayers in the presence of CaCl2 and NaCl regardless of the salt concentration, the ethanol-induced interdigitation of vesicular DPPC depends on the choice of cation and its concentration. These findings unravel a key role for cations in the ethanol-induced interdigitation of lipid membranes in either bulk phase or vesicular form.
Collapse
Affiliation(s)
- Valeria Nele
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Margaret N Holme
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - M Harunur Rashid
- School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia
- Department of Mathematics and Physics, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Hanna M G Barriga
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Tu C Le
- School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia
| | - Michael R Thomas
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K
- London Centre for Nanotechnology and Department of Biochemical Engineering, University College London, 17-19 Gordon Street, London WC1H 0AH, U.K
| | - James J Doutch
- ISIS Neutron and Muon Source, STFC, Rutherford Appleton Laboratory, Didcot OX11 ODE, U.K
| | - Irene Yarovsky
- School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
18
|
Tharushi Perera PG, Todorova N, Vilagosh Z, Bazaka O, Nguyen THP, Bazaka K, Crawford RJ, Croft RJ, Yarovsky I, Ivanova EP. Translocation of silica nanospheres through giant unilamellar vesicles (GUVs) induced by a high frequency electromagnetic field. RSC Adv 2021; 11:31408-31420. [PMID: 35496859 PMCID: PMC9041541 DOI: 10.1039/d1ra05459g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/14/2021] [Indexed: 01/20/2023] Open
Abstract
Membrane model systems capable of mimicking live cell membranes were used for the first time in studying the effects arising from electromagnetic fields (EMFs) of 18 GHz where membrane permeability was observed following exposure. A present lack of understanding of the mechanisms that drive such a rapid change in membrane permeabilization as well as any structural or dynamic changes imparted on biomolecules affected by high-frequency electromagnetic irradiation limits the use of 18 GHz EMFs in biomedical applications. A phospholipid, 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) labelled with a fluorescent marker 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(lissamine rhodamine B sulfonyl) (rhodamine-DOPE) was used in constructing the giant unilamellar vesicles (GUVs). After three cycles of exposure, enhanced membrane permeability was observed by the internalisation of hydrophilic silica nanospheres of 23.5 nm and their clusters. All-atom molecular dynamics simulations of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) membranes exposed to high frequency electric fields of different field strengths showed that within the simulation timeframe only extremely high strength fields were able to cause an increase in the interfacial water dynamics characterized by water dipole realignments. However, a lower strength, high frequency EMF induced changes of the water hydrogen bond network, which may contribute to the mechanisms that facilitate membrane permeabilization in a longer timeframe.
Collapse
Affiliation(s)
- Palalle G Tharushi Perera
- School of Science, RMIT University PO Box 2476 Melbourne VIC 3001 Australia
- Faculty Science, Engineering and Technology, Swinburne University of Technology PO Box 218 Hawthorn VIC 3122 Australia
| | - Nevena Todorova
- School of Engineering, RMIT University PO Box 2476 Melbourne VIC 3001 Australia
| | - Zoltan Vilagosh
- Faculty Science, Engineering and Technology, Swinburne University of Technology PO Box 218 Hawthorn VIC 3122 Australia
| | - Olha Bazaka
- School of Science, RMIT University PO Box 2476 Melbourne VIC 3001 Australia
| | | | - Kateryna Bazaka
- School of Engineering, College of Engineering and Computer Science, The Australian National University Canberra ACT 2600 Australia
| | - Russell J Crawford
- School of Science, RMIT University PO Box 2476 Melbourne VIC 3001 Australia
| | - Rodney J Croft
- School of Psychology, Illawarra Health and Medical Research Institute, University of Wollongong Wollongong NSW 2522 Australia
| | - Irene Yarovsky
- School of Engineering, RMIT University PO Box 2476 Melbourne VIC 3001 Australia
| | - Elena P Ivanova
- School of Science, RMIT University PO Box 2476 Melbourne VIC 3001 Australia
| |
Collapse
|
19
|
Jash A, Ubeyitogullari A, Rizvi SSH. Liposomes for oral delivery of protein and peptide-based therapeutics: challenges, formulation strategies, and advances. J Mater Chem B 2021; 9:4773-4792. [PMID: 34027542 DOI: 10.1039/d1tb00126d] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Throughout the past decade, there has been a rapid growth in the development of protein/peptide-based therapeutics. These therapeutics have found widespread applications in the treatment of cancer, infectious diseases, and other metabolic disorders owing to their several desirable attributes, such as reduced toxicity, diverse biological activities, high specificity, and potency. Most protein/peptide-based drugs are still administered parenterally, and there is an unprecedented demand in the pharmaceutical industry to develop oral delivery routes to increase patient acceptability and convenience. Recent advancements in nanomedicine discoveries have led to the development of several nano and micro-particle-based oral delivery platforms for protein/peptide-based therapeutics and among these, liposomes have emerged as a prominent candidate. Liposomes are spherical vesicles composed of one or more phospholipid bilayers enclosing a core aqueous phase. Their unique amphiphilic nature enables encapsulation of a diverse range of bioactives/drugs including both hydrophobic and hydrophilic compounds for delivery. Against this backdrop, this review provides an overview of the current approaches and challenges associated with the routes and methods of oral administration of protein/peptide-based therapeutics by using liposomes as a potential vehicle. First, the conventional and innovative liposome formation approaches have been discussed along with their applications. Next, the challenges associated with current approaches for oral delivery of protein and peptide-derived therapeutics have been thoroughly addressed. Lastly, we have critically reviewed the potential of liposomes utilization as vehicles for oral delivery of proteins emphasizing the current status and future directions in this area.
Collapse
Affiliation(s)
- Apratim Jash
- Department of Food Science, Cornell University, Ithaca, NY 14850, USA
| | | | | |
Collapse
|
20
|
The molecular mechanisms of listeriolysin O-induced lipid membrane damage. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183604. [PMID: 33722646 DOI: 10.1016/j.bbamem.2021.183604] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/22/2022]
Abstract
Listeria monocytogenes is an intracellular food-borne pathogen that causes listeriosis, a severe and potentially life-threatening disease. Listeria uses a number of virulence factors to proliferate and spread to various cells and tissues. In this process, three bacterial virulence factors, the pore-forming protein listeriolysin O and phospholipases PlcA and PlcB, play a crucial role. Listeriolysin O belongs to a family of cholesterol-dependent cytolysins that are mostly expressed by gram-positive bacteria. Its unique structural features in an otherwise conserved three-dimensional fold, such as the acidic triad and proline-glutamate-serine-threonine-like sequence, enable the regulation of its intracellular activity as well as distinct extracellular functions. The stability of listeriolysin O is pH- and temperature-dependent, and this provides another layer of control of its activity in cells. Moreover, many recent studies have demonstrated a unique mechanism of pore formation by listeriolysin O, i.e., the formation of arc-shaped oligomers that can subsequently fuse to form membrane defects of various shapes and sizes. During listerial invasion of host cells, these membrane defects can disrupt phagosome membranes, allowing bacteria to escape into the cytosol and rapidly multiply. The activity of listeriolysin O is profoundly dependent on the amount and accessibility of cholesterol in the lipid membrane, which can be modulated by the phospholipase PlcB. All these prominent features of listeriolysin O play a role during different stages of the L. monocytogenes life cycle by promoting the proliferation of the pathogen while mitigating excessive damage to its replicative niche in the cytosol of the host cell.
Collapse
|
21
|
Fong WK, Sánchez-Ferrer A, Rappolt M, Boyd BJ, Mezzenga R. Structural Transformation in Vesicles upon Hydrolysis of Phosphatidylethanolamine and Phosphatidylcholine with Phospholipase C. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:14949-14958. [PMID: 31642682 DOI: 10.1021/acs.langmuir.9b02288] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This study provides insights into dynamic nanostructural changes in phospholipid systems during hydrolysis with phospholipase C, the fate of the hydrolysis products, and the kinetics of lipolysis. The effect of lipid restructuring of the vesicle was investigated using small-angle X-ray scattering and cryogenic scanning electron microscopy. The rate and extent of phospholipid hydrolysis were quantified using nuclear magnetic resonance. Hydrolysis of two phospholipids, phosphatidylethanolamine (PE) and phosphatidylcholine (PC), results in the cleavage of the molecular headgroup, causing two strikingly different changes in lipid self-assembly. The diacylglycerol product of PC escapes the lipid bilayer, whereas the diacylglycerol product adopts a different configuration within the lipid bilayer of the PE vesicles. These results are then discussed concerning the change of the lipid configuration upon the lipid membrane and its potential implications in vivo, which is of significant importance for the detailed understanding of the fate of lipidic particles and the rational design of enzyme-responsive lipid-based drug delivery systems.
Collapse
Affiliation(s)
- Wye-Khay Fong
- Department of Health Sciences & Technology , ETH Zürich , 8092 Zürich , Switzerland
- Drug Delivery, Disposition and Dynamics, and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville Campus, 381 Royal Parade , Parkville , 3052 Victoria , Australia
- Adolphe Merkle Institute , University of Fribourg , Chemin des Verdiers 4 , 1700 Fribourg , Switzerland
| | | | - Michael Rappolt
- School of Food Science and Nutrition , University of Leeds , LS2 9JT Leeds , Yorkshire , U.K
| | - Ben J Boyd
- Drug Delivery, Disposition and Dynamics, and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences , Monash University , Parkville Campus, 381 Royal Parade , Parkville , 3052 Victoria , Australia
| | - Raffaele Mezzenga
- Department of Health Sciences & Technology , ETH Zürich , 8092 Zürich , Switzerland
| |
Collapse
|
22
|
Local accumulation of diacylglycerol alters membrane properties nonlinearly due to its transbilayer activity. Commun Chem 2019. [DOI: 10.1038/s42004-019-0175-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
23
|
Nele V, Holme MN, Kauscher U, Thomas MR, Doutch JJ, Stevens MM. Effect of Formulation Method, Lipid Composition, and PEGylation on Vesicle Lamellarity: A Small-Angle Neutron Scattering Study. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:6064-6074. [PMID: 30977658 PMCID: PMC6506804 DOI: 10.1021/acs.langmuir.8b04256] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Liposomes are well-established systems for drug delivery and biosensing applications. The design of a liposomal carrier requires careful choice of lipid composition and formulation method. These determine many vesicle properties including lamellarity, which can have a strong effect on both encapsulation efficiency and the efflux rate of encapsulated active compounds. Despite this, a comprehensive study on how the lipid composition and formulation method affect vesicle lamellarity is still lacking. Here, we combine small-angle neutron scattering and cryogenic transmission electron microscopy to study the effect of three different well-established formulation methods followed by extrusion through 100 nm polycarbonate membranes on the resulting vesicle membrane structure. Specifically, we examine vesicles formulated from the commonly used phospholipids 1-palmitoyl-2-oleoyl- sn-glycero-3-phosphocholine (POPC), 1,2-dipalmitoyl- sn-glycero-3-phosphocholine (DPPC) and 1,2-dioleoyl- sn-glycero-3-phosphocholine (DOPC) via film hydration followed by (i) agitation on a shaker or (ii) freeze-thawing, or (iii) the reverse-phase evaporation vesicle method. After extrusion, up to half of the total lipid content is still assembled into multilamellar structures. However, we achieved unilamellar vesicle populations when as little as 0.1 mol % PEG-modified lipid was included in the vesicle formulation. Interestingly, DPPC with 5 mol % PEGylated lipid produces a combination of cylindrical micelles and vesicles. In conclusion, our results provide important insights into the effect of the formulation method and lipid composition on producing liposomes with a defined membrane structure.
Collapse
Affiliation(s)
- Valeria Nele
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Margaret N. Holme
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- E-mail: (M.N.H.)
| | - Ulrike Kauscher
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - Michael R. Thomas
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
| | - James J. Doutch
- ISIS
Neutron and Muon Source, STFC, Rutherford
Appleton Laboratory, Didcot OX11 ODE, U.K.
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- E-mail: (M.M.S.)
| |
Collapse
|
24
|
Xu X, Pan J, Li X, Cui Y, Mao Z, Wu B, Xu H, Zhou W, Liu Y. Inhibition of Methamphetamine Self-Administration and Reinstatement by Central Blockade of Angiotensin II Receptor in Rats. J Pharmacol Exp Ther 2019; 369:244-258. [PMID: 30867225 DOI: 10.1124/jpet.118.255729] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
The molecular mechanism and treatment of methamphetamine (METH) use disorder remain unclear. The current study aimed to investigate the role of central angiotensin II receptor (ATR) in drug taking and seeking behavior associated with METH use disorder. The effect of an ATR type 1 (AT1R) antagonist, candesartan cilexetil, on the reinforcing and motivational effects of METH was first assessed using the animal model of METH self-administration (SA) and reinstatement. The levels of dopamine D2 receptor (D2R) and AT1R were subsequently examined. Furthermore, the present study determined the expression of microRNAs (miRNAs) by comparing METH SA, METH-yoked, and Saline-yoked groups. The target miRNAs were further overexpressed in the nucleus accumbens (NAc) via a lentivirus vector to investigate the effects of target miRNAs on METH SA maintained under a fixed ratio 1, progressive ratio, and cue/drug reinstatement of METH SA. The potential role of the AT1R-PLCβ-CREB signaling pathway was finally investigated. The results suggest that AT1R blockade effectively reduced METH SA and reinstatement, in conjunction with the counter-regulation of D2R and AT1R. A total of 17 miRNAs targeting Ang II in NAc were found to be associated with the voluntary intake of METH. Furthermore, overexpression of specific miR-219a-5p targeting AT1R-regulated METH SA and reinstatement. The AT1R-PLCβ-CREB signaling pathway was found to be associated with the effect of AT1R on the drug-taking and drug-seeking behavior involving METH use disorder.
Collapse
Affiliation(s)
- Xing Xu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Jian Pan
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Xingxing Li
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Yan Cui
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Zijuan Mao
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Boliang Wu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Huachong Xu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Wenhua Zhou
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Yu Liu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| |
Collapse
|