1
|
Winnikoff JR, Milshteyn D, Vargas-Urbano SJ, Pedraza MA, Armando AM, Quehenberger O, Sodt A, Gillilan RE, Dennis EA, Lyman E, Haddock SHD, Budin I. Homeocurvature adaptation of phospholipids to pressure in deep-sea invertebrates. Science 2024; 384:1482-1488. [PMID: 38935710 PMCID: PMC11593575 DOI: 10.1126/science.adm7607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/17/2024] [Indexed: 06/29/2024]
Abstract
Hydrostatic pressure increases with depth in the ocean, but little is known about the molecular bases of biological pressure tolerance. We describe a mode of pressure adaptation in comb jellies (ctenophores) that also constrains these animals' depth range. Structural analysis of deep-sea ctenophore lipids shows that they form a nonbilayer phase at pressures under which the phase is not typically stable. Lipidomics and all-atom simulations identified phospholipids with strong negative spontaneous curvature, including plasmalogens, as a hallmark of deep-adapted membranes that causes this phase behavior. Synthesis of plasmalogens enhanced pressure tolerance in Escherichia coli, whereas low-curvature lipids had the opposite effect. Imaging of ctenophore tissues indicated that the disintegration of deep-sea animals when decompressed could be driven by a phase transition in their phospholipid membranes.
Collapse
Affiliation(s)
- Jacob R. Winnikoff
- Department of Chemistry and Biochemistry, University of California San Diego; 9500 Gilman Dr., La Jolla, CA 92093, USA
- Department of Organismic and Evolutionary Biology, Harvard University; 16 Divinity Ave., Cambridge, MA 02138, USA
- Monterey Bay Aquarium Research Institute; 7700 Sandholdt Rd., Moss Landing, CA 95039, USA
- Department of Ecology and Evolutionary Biology, University of California Santa Cruz; 1156 High St., Santa Cruz, CA 95064, USA
| | - Daniel Milshteyn
- Department of Chemistry and Biochemistry, University of California San Diego; 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Sasiri J. Vargas-Urbano
- Department of Physics and Astronomy, University of Delaware; 210 South College Ave., Newark, DE 19716, USA
| | - Miguel A. Pedraza
- Department of Physics and Astronomy, University of Delaware; 210 South College Ave., Newark, DE 19716, USA
| | - Aaron M. Armando
- Department of Pharmacology, University of California San Diego Health Sciences; 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Oswald Quehenberger
- Department of Pharmacology, University of California San Diego Health Sciences; 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Alexander Sodt
- Unit on Membrane Chemical Physics, National Institute of Child Health and Human Development; 29 Lincoln Drive, Bethesda, MD 20892
| | | | - Edward A. Dennis
- Department of Chemistry and Biochemistry, University of California San Diego; 9500 Gilman Dr., La Jolla, CA 92093, USA
- Department of Pharmacology, University of California San Diego Health Sciences; 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Edward Lyman
- Department of Physics and Astronomy, University of Delaware; 210 South College Ave., Newark, DE 19716, USA
| | - Steven H. D. Haddock
- Monterey Bay Aquarium Research Institute; 7700 Sandholdt Rd., Moss Landing, CA 95039, USA
- Department of Ecology and Evolutionary Biology, University of California Santa Cruz; 1156 High St., Santa Cruz, CA 95064, USA
| | - Itay Budin
- Department of Chemistry and Biochemistry, University of California San Diego; 9500 Gilman Dr., La Jolla, CA 92093, USA
| |
Collapse
|
2
|
Stępień P, Świątek S, Robles MYY, Markiewicz-Mizera J, Balakrishnan D, Inaba-Inoue S, De Vries AH, Beis K, Marrink SJ, Heddle JG. CRAFTing Delivery of Membrane Proteins into Protocells using Nanodiscs. ACS APPLIED MATERIALS & INTERFACES 2023; 15. [PMID: 38015973 PMCID: PMC10726305 DOI: 10.1021/acsami.3c11894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/30/2023]
Abstract
For the successful generative engineering of functional artificial cells, a convenient and controllable means of delivering membrane proteins into membrane lipid bilayers is necessary. Here we report a delivery system that achieves this by employing membrane protein-carrying nanodiscs and the calcium-dependent fusion of phosphatidylserine lipid membranes. We show that lipid nanodiscs can fuse a transported lipid bilayer with the lipid bilayers of small unilamellar vesicles (SUVs) or giant unilamellar vesicles (GUVs) while avoiding recipient vesicles aggregation. This is triggered by a simple, transient increase in calcium concentration, which results in efficient and rapid fusion in a one-pot reaction. Furthermore, nanodiscs can be loaded with membrane proteins that can be delivered into target SUV or GUV membranes in a detergent-independent fashion while retaining their functionality. Nanodiscs have a proven ability to carry a wide range of membrane proteins, control their oligomeric state, and are highly adaptable. Given this, our approach may be the basis for the development of useful tools that will allow bespoke delivery of membrane proteins to protocells, equipping them with the cell-like ability to exchange material across outer/subcellular membranes.
Collapse
Affiliation(s)
- Piotr Stępień
- Malopolska
Centre of Biotechnology, Jagiellonian University, Krakow 30-387, Poland
| | - Sylwia Świątek
- Malopolska
Centre of Biotechnology, Jagiellonian University, Krakow 30-387, Poland
| | | | | | - Dhanasekaran Balakrishnan
- Malopolska
Centre of Biotechnology, Jagiellonian University, Krakow 30-387, Poland
- Postgraduate
School of Molecular Medicine, Żwirki i Wigury 61, Warsaw 02-091, Poland
| | - Satomi Inaba-Inoue
- Department
of Life Sciences, Imperial College London, Exhibition Road, South Kensington, London SW7 2AZ, U.K.
- Rutherford
Appleton Laboratory, Research Complex at
Harwell, Didcot, Oxfordshire OX11 0FA, U.K.
| | - Alex H. De Vries
- Groningen
Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen 9747 AG, The Netherlands
| | - Konstantinos Beis
- Department
of Life Sciences, Imperial College London, Exhibition Road, South Kensington, London SW7 2AZ, U.K.
- Rutherford
Appleton Laboratory, Research Complex at
Harwell, Didcot, Oxfordshire OX11 0FA, U.K.
| | - Siewert J. Marrink
- Groningen
Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen 9747 AG, The Netherlands
| | - Jonathan G. Heddle
- Malopolska
Centre of Biotechnology, Jagiellonian University, Krakow 30-387, Poland
| |
Collapse
|
3
|
Niort K, Dancourt J, Boedec E, Al Amir Dache Z, Lavieu G, Tareste D. Cholesterol and Ceramide Facilitate Membrane Fusion Mediated by the Fusion Peptide of the SARS-CoV-2 Spike Protein. ACS OMEGA 2023; 8:32729-32739. [PMID: 37720777 PMCID: PMC10500581 DOI: 10.1021/acsomega.3c03610] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/17/2023] [Indexed: 09/19/2023]
Abstract
SARS-CoV-2 entry into host cells is mediated by the Spike (S) protein of the viral envelope. The S protein is composed of two subunits: S1 that induces binding to the host cell via its interaction with the ACE2 receptor of the cell surface and S2 that triggers fusion between viral and cellular membranes. Fusion by S2 depends on its heptad repeat domains that bring membranes close together and its fusion peptide (FP) that interacts with and perturbs the membrane structure to trigger fusion. Recent studies have suggested that cholesterol and ceramide lipids from the cell surface may facilitate SARS-CoV-2 entry into host cells, but their exact mode of action remains unknown. We have used a combination of in vitro liposome-liposome and in situ cell-cell fusion assays to study the lipid determinants of S-mediated membrane fusion. Our findings reveal that both cholesterol and ceramide lipids facilitate fusion, suggesting that targeting these lipids could be effective against SARS-CoV-2. As a proof of concept, we examined the effect of chlorpromazine (CPZ), an antipsychotic drug known to perturb membrane structure. Our results show that CPZ effectively inhibits S-mediated membrane fusion, thereby potentially impeding SARS-CoV-2 entry into the host cell.
Collapse
Affiliation(s)
- Kristina Niort
- Université
Paris Cité, Inserm UMR-S 1266, Institute of Psychiatry and
Neuroscience of Paris (IPNP), Paris 75014, France
| | - Julia Dancourt
- Université
Paris Cité, Inserm U 1316, CNRS UMR 7057, Laboratoire Matières
et Systèmes Complexes (MSC), Paris 75006, France
| | - Erwan Boedec
- Université
Paris Cité, Inserm UMR-S 1266, Institute of Psychiatry and
Neuroscience of Paris (IPNP), Paris 75014, France
| | - Zahra Al Amir Dache
- Université
Paris Cité, Inserm U 1316, CNRS UMR 7057, Laboratoire Matières
et Systèmes Complexes (MSC), Paris 75006, France
| | - Grégory Lavieu
- Université
Paris Cité, Inserm U 1316, CNRS UMR 7057, Laboratoire Matières
et Systèmes Complexes (MSC), Paris 75006, France
| | - David Tareste
- Université
Paris Cité, Inserm UMR-S 1266, Institute of Psychiatry and
Neuroscience of Paris (IPNP), Paris 75014, France
| |
Collapse
|
4
|
Canepa E, Bochicchio D, Brosio G, Silva PHJ, Stellacci F, Dante S, Rossi G, Relini A. Cholesterol-Containing Liposomes Decorated With Au Nanoparticles as Minimal Tunable Fusion Machinery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207125. [PMID: 36899445 DOI: 10.1002/smll.202207125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/30/2023] [Indexed: 06/08/2023]
Abstract
Membrane fusion is essential for the basal functionality of eukaryotic cells. In physiological conditions, fusion events are regulated by a wide range of specialized proteins, operating with finely tuned local lipid composition and ionic environment. Fusogenic proteins, assisted by membrane cholesterol and calcium ions, provide the mechanical energy necessary to achieve vesicle fusion in neuromediator release. Similar cooperative effects must be explored when considering synthetic approaches for controlled membrane fusion. We show that liposomes decorated with amphiphilic Au nanoparticles (AuLips) can act as minimal tunable fusion machinery. AuLips fusion is triggered by divalent ions, while the number of fusion events dramatically changes with, and can be finely tuned by, the liposome cholesterol content. We combine quartz-crystal-microbalance with dissipation monitoring (QCM-D), fluorescence assays, and small-angle X-ray scattering (SAXS) with molecular dynamics (MD) at coarse-grained (CG) resolution, revealing new mechanistic details on the fusogenic activity of amphiphilic Au nanoparticles (AuNPs) and demonstrating the ability of these synthetic nanomaterials to induce fusion regardless of the divalent ion used (Ca2+ or Mg2+ ). The results provide a novel contribution to developing new artificial fusogenic agents for next-generation biomedical applications that require tight control of the rate of fusion events (e.g., targeted drug delivery).
Collapse
Affiliation(s)
- Ester Canepa
- Department of Physics, University of Genoa, Genoa, 16146, Italy
- Institute of Materials Science & Engineering, EPFL, Lausanne, 1015, Switzerland
| | | | - Giorgia Brosio
- Department of Physics, University of Genoa, Genoa, 16146, Italy
| | | | - Francesco Stellacci
- Materials Characterization Facility, Istituto Italiano di Tecnologia, Genoa, 16163, Italy
| | - Silvia Dante
- Institute of Materials Science & Engineering, EPFL, Lausanne, 1015, Switzerland
| | - Giulia Rossi
- Department of Physics, University of Genoa, Genoa, 16146, Italy
| | - Annalisa Relini
- Department of Physics, University of Genoa, Genoa, 16146, Italy
| |
Collapse
|
5
|
Sule K, Anikovskiy M, Prenner EJ. Lipid Structure Determines the Differential Impact of Single Metal Additions and Binary Mixtures of Manganese, Calcium and Magnesium on Membrane Fluidity and Liposome Size. Int J Mol Sci 2023; 24:1066. [PMID: 36674581 PMCID: PMC9860990 DOI: 10.3390/ijms24021066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/23/2022] [Accepted: 01/02/2023] [Indexed: 01/08/2023] Open
Abstract
Unilamellar vesicles of the biologically relevant lipids phosphatidic acid (PA) and phosphatidylserine (PS) with fully saturated (DM-) or partly unsaturated (PO-) acyl side chains were exposed to Ca, Mn and Mg in single metal additions; in equimolar mixtures or by sequential additions of one metal at a time. Laurdan generalized polarization measured the membrane fluidity, while dynamic light scattering reported liposome size changes complemented by zeta potential. All metals induced membrane rigidity and increased liposome sizes across all systems. Mn had the strongest effect overall, but Mg was comparable for DMPS. Lipid side chain architecture was important as GP values for binary mixtures were higher than expected from the sum of values for single additions added to POPS but smaller for DMPS. Sequential additions were predominantly different for Ca:Mg mixtures. Mn induced the strongest increase of liposome size in saturated lipids whereas Ca effects dominated unsaturated matrices. Binary additions induced larger sizes than the sum of single additions for POPS, but much lower changes in DMPA. The order of addition was relevant for PS systems. Thus, lipid structure determines metal effects, but their impact is modulated by other ions. Thus, metal effects may differ with the local lipid architecture and metal concentrations within cells.
Collapse
Affiliation(s)
- Kevin Sule
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Max Anikovskiy
- Department of Chemistry, Nanoscience Program, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Elmar J. Prenner
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
6
|
Mechanisms of Co, Ni, and Mn toxicity: From exposure and homeostasis to their interactions with and impact on lipids and biomembranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183250. [DOI: 10.1016/j.bbamem.2020.183250] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/12/2020] [Accepted: 02/24/2020] [Indexed: 01/21/2023]
|
7
|
Lipidomic profiling analysis of the phospholipid molecules in SCAP-induced lipid droplet formation in bovine mammary epithelial cells. Prostaglandins Other Lipid Mediat 2020; 149:106420. [PMID: 31953015 DOI: 10.1016/j.prostaglandins.2020.106420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/15/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
Abstract
The accumulation of lipid droplets (LDs) in the cytoplasm plays an important role in energy balance, membrane synthesis and cell signal transduction. The aim of this study was to investigate the profile of phospholipids after SCAP-induced LD formation in bovine mammary epithelial cells (BMECs). A shRNA-SCAP vector and a SCAP/SREBP vector were used to knock down and overexpress the SCAP gene in BMECs prior to evaluating the effects on LDs using Western blotting, real-time PCR, LD staining and liquid chromatography-tandem mass spectrometry (LC-MS/MS). The average LD diameter was determined following oil red O staining. The overexpression of SCAP increased the abundance of SCD, ACACA and FASN genes and nuclear SREBP1a. In contrast, knocking down SCAP decreased the abundance of the nuclear SREBP1a protein and downregulated the abundance of target genes. Lipid droplet staining revealed that knocking down SCAP reduced LD formation and average LD diameter. In contrast, overexpression of SCAP increased the formation and size of the LDs. The results from an analysis of cellular lipids revealed that phospholipids are the predominant species in the profile of cell lipids. phosphatidylethanolamine (PE) and phosphatidylcholine (PC) are important for determining the size of LDs. The LD formation induced by SCAP gene overexpression and knockdown underscored the role of phospholipids involved in lipid droplet formation and fusion.
Collapse
|
8
|
Kim B, Park JH, Sailor MJ. Rekindling RNAi Therapy: Materials Design Requirements for In Vivo siRNA Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1903637. [PMID: 31566258 PMCID: PMC6891135 DOI: 10.1002/adma.201903637] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/12/2019] [Indexed: 05/07/2023]
Abstract
With the recent FDA approval of the first siRNA-derived therapeutic, RNA interference (RNAi)-mediated gene therapy is undergoing a transition from research to the clinical space. The primary obstacle to realization of RNAi therapy has been the delivery of oligonucleotide payloads. Therefore, the main aims is to identify and describe key design features needed for nanoscale vehicles to achieve effective delivery of siRNA-mediated gene silencing agents in vivo. The problem is broken into three elements: 1) protection of siRNA from degradation and clearance; 2) selective homing to target cell types; and 3) cytoplasmic release of the siRNA payload by escaping or bypassing endocytic uptake. The in vitro and in vivo gene silencing efficiency values that have been reported in publications over the past decade are quantitatively summarized by material type (lipid, polymer, metal, mesoporous silica, and porous silicon), and the overall trends in research publication and in clinical translation are discussed to reflect on the direction of the RNAi therapeutics field.
Collapse
Affiliation(s)
- Byungji Kim
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Michael J Sailor
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| |
Collapse
|
9
|
Pannwitt S, Slama K, Depoix F, Helm M, Schneider D. Against Expectations: Unassisted RNA Adsorption onto Negatively Charged Lipid Bilayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:14704-14711. [PMID: 31626734 DOI: 10.1021/acs.langmuir.9b02830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The composition and physicochemical properties of biological membranes can be altered by diverse membrane integral and peripheral proteins as well as by small molecules, natural and synthetic. Diverse oligonucleotides have been shown to electrostatically interact with cationic and bivalent ion loaded zwitterionic liposomes, leading to the formation of oligonucleotide-liposome aggregates. However, interaction of RNAs with other membrane surfaces remains ill understood. We used the nonnatural RNA10 to investigate RNA binding to anionic and net-uncharged membrane surfaces. RNA10 had initially been selected in a screen for nonnatural RNA motives that bind to phosphatidylcholine liposomes in the presence of Mg2+. Here we show that interaction of defined RNA molecules with membrane surfaces crucially depends on electrostatic surface properties. Furthermore, RNA10 electrostatically binds to anionic lipid bilayers in the absence of Mg2+ or other bivalent cations, and this interaction leads to measurably changed physicochemical properties of the bilayer and the oligonucleotide. Thus, the structure of polyanionic RNA can be modulated via contact with negatively charged membrane surfaces and vice versa.
Collapse
Affiliation(s)
- Stefanie Pannwitt
- Institute of Pharmacy and Biochemistry , Johannes Gutenberg University , Johann-Joachim-Becherweg 30 , 55128 Mainz , Germany
| | - Kaouthar Slama
- Institute of Pharmacy and Biochemistry , Johannes Gutenberg University , Staudinger Weg 5 , 55128 Mainz , Germany
| | - Frank Depoix
- Institute of Molecular Physiology , Johannes Gutenberg University , Johann-Joachim-Becherweg 9-11 , 55128 Mainz , Germany
| | - Mark Helm
- Institute of Pharmacy and Biochemistry , Johannes Gutenberg University , Staudinger Weg 5 , 55128 Mainz , Germany
| | - Dirk Schneider
- Institute of Pharmacy and Biochemistry , Johannes Gutenberg University , Johann-Joachim-Becherweg 30 , 55128 Mainz , Germany
| |
Collapse
|
10
|
Yue K, Trung TN, Zhu Y, Kaldenhoff R, Kai L. Co-Translational Insertion of Aquaporins into Liposome for Functional Analysis via an E. coli Based Cell-Free Protein Synthesis System. Cells 2019; 8:E1325. [PMID: 31717877 PMCID: PMC6912355 DOI: 10.3390/cells8111325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 01/06/2023] Open
Abstract
Aquaporins are important and well-studied water channel membrane proteins. However, being membrane proteins, sample preparation for functional analysis is tedious and time-consuming. In this paper, we report a new approach for the co-translational insertion of two aquaporins from Escherichia coli and Nicotiana tabacum using the CFPS system. This was done in the presence of liposomes with a modified procedure to form homogenous proteo-liposomes suitable for functional analysis of water permeability using stopped-flow spectrophotometry. Two model aquaporins, AqpZ and NtPIP2;1, were successfully incorporated into the liposome in their active forms. Shifted green fluorescent protein was fused to the C-terminal part of AqpZ to monitor its insertion and status in the lipid environment. This new fast approach offers a fast and straightforward method for the functional analysis of aquaporins in both prokaryotic and eukaryotic organisms.
Collapse
Affiliation(s)
- Ke Yue
- The Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Sciences, Jiangsu Normal University, Xuzhou 22116, China;
| | - Tran Nam Trung
- Department of Biology, Applied Plant Sciences, Technische Universität Darmstadt, Schnittspahn Strasse 10, D-64287 Darmstadt, Germany; (T.N.T.); (R.K.)
| | - Yiyong Zhu
- Jiangsu Provincial Key Lab for Organic Solid Waste Utilization, National Engineering Research Center for Organic-based Fertilizers, Jiangsu Collaborative Innovation Center for Solid Organic Waste Resource Utilization, Nanjing Agricultural University, Nanjing 210095, China;
| | - Ralf Kaldenhoff
- Department of Biology, Applied Plant Sciences, Technische Universität Darmstadt, Schnittspahn Strasse 10, D-64287 Darmstadt, Germany; (T.N.T.); (R.K.)
| | - Lei Kai
- The Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Sciences, Jiangsu Normal University, Xuzhou 22116, China;
- Department of Biology, Applied Plant Sciences, Technische Universität Darmstadt, Schnittspahn Strasse 10, D-64287 Darmstadt, Germany; (T.N.T.); (R.K.)
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, D-82152 Martinsried, Germany
| |
Collapse
|
11
|
Argov-Argaman N. Symposium review: Milk fat globule size: Practical implications and metabolic regulation. J Dairy Sci 2019; 102:2783-2795. [PMID: 30639008 DOI: 10.3168/jds.2018-15240] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
Abstract
Milk fat globule (MFG) size ranges over 3 orders of magnitude, from less than 200 nm to over 15 µm. The significance of MFG size derives from its tight association with its lipidome and proteome. More specifically, small MFG have relatively higher content of membrane compared with large globules, and this membrane exerts diverse positive health effects, as reported in human and animal studies. In addition, MFG size has industrial significance, as it affects the physicochemical and sensory characteristics of dairy products. Studies on the size regulation of MFG are scarce, mainly because various confounders indirectly affect MFG size. Because MFG size is determined before and during its secretion from mammary epithelial cells, studies on the size regulation of its precursors, the intracellular lipid droplets (LD), have been used as a proxy for understanding the mechanisms controlling MFG size. In this review, we provide evidence for 2 distinct mechanisms regulating LD size in mammary epithelial cells: co-regulation of fat content and triglyceride-synthesis capacity of the cells, and fusion between LD. The latter is controlled by the membrane's polar lipid composition and involves mitochondrial enzymes. Accordingly, this review also discusses MFG size regulation in the in vivo metabolic context, as MFG morphometric features are often modulated under conditions that involve animals' altered energy status.
Collapse
Affiliation(s)
- Nurit Argov-Argaman
- Department of Animal Science, the Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University of Jerusalem, Israel, POB 76100.
| |
Collapse
|
12
|
Abstract
Mitochondria undergo frequent fusion and fission events to adapt their morphology to cellular needs. Homotypic docking and fusion of outer mitochondrial membranes are controlled by Mitofusins, a set of large membrane-anchored GTPase proteins belonging to the dynamin superfamily. Mitofusins include, in addition to their GTPase and transmembrane domains, two heptad repeat domains, HR1 and HR2. All four regions are crucial for Mitofusin function, but their precise contribution to mitochondrial docking and fusion events has remained elusive until very recently. In this commentary, we first give an overview of the established strategies employed by various protein machineries distinct from Mitofusins to mediate membrane fusion. We then present recent structure–function data on Mitofusins that provide important novel insights into their mode of action in mitochondrial fusion.
Collapse
Affiliation(s)
- Mickael M Cohen
- Sorbonne Université, CNRS UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, Paris, France
| | - David Tareste
- Université Paris Descartes, Sorbonne Paris Cité, INSERM ERL U950, Trafic Membranaire dans le Cerveau Normal et Pathologique, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, INSERM UMR 894, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| |
Collapse
|
13
|
Allolio C, Magarkar A, Jurkiewicz P, Baxová K, Javanainen M, Mason PE, Šachl R, Cebecauer M, Hof M, Horinek D, Heinz V, Rachel R, Ziegler CM, Schröfel A, Jungwirth P. Arginine-rich cell-penetrating peptides induce membrane multilamellarity and subsequently enter via formation of a fusion pore. Proc Natl Acad Sci U S A 2018; 115:11923-11928. [PMID: 30397112 PMCID: PMC6255155 DOI: 10.1073/pnas.1811520115] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Arginine-rich cell-penetrating peptides do not enter cells by directly passing through a lipid membrane; they instead passively enter vesicles and live cells by inducing membrane multilamellarity and fusion. The molecular picture of this penetration mode, which differs qualitatively from the previously proposed direct mechanism, is provided by molecular dynamics simulations. The kinetics of vesicle agglomeration and fusion by an iconic cell-penetrating peptide-nonaarginine-are documented via real-time fluorescence techniques, while the induction of multilamellar phases in vesicles and live cells is demonstrated by a combination of electron and fluorescence microscopies. This concert of experiments and simulations reveals that the identified passive cell penetration mechanism bears analogy to vesicle fusion induced by calcium ions, indicating that the two processes may share a common mechanistic origin.
Collapse
Affiliation(s)
- Christoph Allolio
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, CZ-166 10 Prague 6, Czech Republic
- Institute of Physical and Theoretical Chemistry, University of Regensburg, D-93040 Regensburg, Germany
- Fritz Haber Research Center, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
- Department of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Aniket Magarkar
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, CZ-166 10 Prague 6, Czech Republic
- Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Piotr Jurkiewicz
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, 182 23 Prague 8, Czech Republic
| | - Katarína Baxová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, CZ-166 10 Prague 6, Czech Republic
| | - Matti Javanainen
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, CZ-166 10 Prague 6, Czech Republic
| | - Philip E Mason
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, CZ-166 10 Prague 6, Czech Republic
| | - Radek Šachl
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, 182 23 Prague 8, Czech Republic
| | - Marek Cebecauer
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, 182 23 Prague 8, Czech Republic
| | - Martin Hof
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, 182 23 Prague 8, Czech Republic
| | - Dominik Horinek
- Institute of Physical and Theoretical Chemistry, University of Regensburg, D-93040 Regensburg, Germany
| | - Veronika Heinz
- Institute of Biophysics and Biophysical Chemistry, University of Regensburg, D-93040 Regensburg, Germany
| | - Reinhard Rachel
- Microbiology and Archaea Centre, University of Regensburg, D-93040 Regensburg, Germany
| | - Christine M Ziegler
- Institute of Biophysics and Biophysical Chemistry, University of Regensburg, D-93040 Regensburg, Germany
- Institute of Biophysics and Biophysical Chemistry, University of Regensburg, D-93040 Regensburg, Germany
| | - Adam Schröfel
- Imaging Methods Core Facility at Biocev, Faculty of Sciences, Charles University, 242 50 Vestec, Czech Republic
| | - Pavel Jungwirth
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, CZ-166 10 Prague 6, Czech Republic;
| |
Collapse
|
14
|
Aronson MR, Simonson AW, Orchard LM, Llinás M, Medina SH. Lipopeptisomes: Anticancer peptide-assembled particles for fusolytic oncotherapy. Acta Biomater 2018; 80:269-277. [PMID: 30240951 DOI: 10.1016/j.actbio.2018.09.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/28/2018] [Accepted: 09/17/2018] [Indexed: 12/31/2022]
Abstract
Anticancer peptides (ACPs) are cationic amphiphiles that preferentially kill cancer cells through folding-dependent membrane disruption. Although ACPs represent attractive therapeutic candidates, particularly against drug-resistant cancers, their successful translation into clinical practice has gone unrealized due to their poor bioavailability, serum instability and, most importantly, severe hemolytic toxicity. Here, we exploit the membrane-specific interactions of ACPs to prepare a new class of peptide-lipid particle, we term a lipopeptisome (LP). This design sequesters loaded ACPs within a lipid lamellar corona to avoid contact with red blood cells and healthy tissues, while affording potent lytic destruction of cancer cells following LP-membrane fusion. Biophysical studies show ACPs rapidly fold at, and integrate into, liposomal membranes to form stable LPs with high loading efficiencies (>80%). Rational design of the particles to possess lipid combinations mimicking that of the aberrant cancer cell outer leaflet allows LPs to rapidly fuse with tumor cell membranes and afford localized assembly of loaded ACPs within the bilayer. This leads to preferential fusolytic killing of cancer cells with minimal collateral toxicity towards non-cancerous cells and erythrocytes, thereby imparting clinically relevant therapeutic indices to otherwise toxic ACPs. Thus, integration of ACPs into self-assembled LPs represents a new delivery strategy to improve the therapeutic utility of oncolytic agents, and suggests this technology may be added to targeted combinatorial approaches in precision medicine. STATEMENT OF SIGNIFICANCE: Despite their significant clinical potential, the therapeutic utility of many ACPs has been limited by their collateral hemolysis during administration. Leveraging the membrane-specific interactions of ACPs, here we prepare self-assembled peptide-lipid nanoparticles, or 'lipopeptisomes' (LPs), capable of preferentially fusing with and lysing cancer cell membranes. Key to this fusolytic action is the construction of LPs from lipids simulating the cancer cell outer leaflet. This design recruits the oncolytic peptide payload into the carrier lamella and allows for selective destruction of cancer cells without disrupting healthy cells. Consequently, LPs impart clinically relevant therapeutic indexes to previously toxic ACPs, and thus open new opportunities to improve the clinical translation of oncolytics challenged by narrow therapeutic windows.
Collapse
Affiliation(s)
- Matthew R Aronson
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Andrew W Simonson
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Lindsey M Orchard
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA; Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA; Huck Center for Malaria Research, The Pennsylvania State University, University Park, PA 16802, USA
| | - Scott H Medina
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
15
|
Rahnfeld L, Thamm J, Steiniger F, van Hoogevest P, Luciani P. Study on the in situ aggregation of liposomes with negatively charged phospholipids for use as injectable depot formulation. Colloids Surf B Biointerfaces 2018; 168:10-17. [PMID: 29478769 DOI: 10.1016/j.colsurfb.2018.02.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 01/09/2023]
Abstract
Compared to conventional parenteral formulations injectable depot formulations, owing to a sustained drug release, offer several advantages, such as a reduced dosing frequency - and consequent improved compliance - or a predictable release profile. Additionally, fluctuations in the drug blood level may be smoothened and consequently side effects reduced. Because of their capability to encapsulate water soluble drugs and their very low toxicity profile liposomes comprising phospholipids, most certainly represent a vehicle of choice for subcutaneous (s.c.) or intramuscular (i.m.) administration typical for depot injections too. In the past, especially liposomes containing negatively charged phosphatidylserines were investigated regarding their aggregation and fusion behavior upon addition of calcium ions. Liposomes need to have a large size to prevent fast removal from the s.c. or i.m. injection site to make them useful as depot vehicle. In order to obtain such large liposomes, aggregation of smaller liposomes may be considered. Aim of the present study was to induce and investigate controlled aggregation of vesicles containing other negatively charged phospholipids besides phosphatidylserines upon mixing with a solution of divalent cations. L-α-phosphatidylcholine from egg (EPC) liposomes formulated with 25 mol% of 1,2-dipalmitoyl-sn-glycero-3-phosphate (DPPA) or 1,2-distearoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (DSPG) proved to be possible lipid-based depot candidates due to their strong aggregation induced by calcium and magnesium cations. Different aggregation profiles with both cations could be observed. Morphological investigations of the aggregates showed that individual liposomes remain stable in the aggregates and no fusion occurred. A fluorescence-based fusion assay confirmed these results. Differential scanning calorimetry measurements supported the findings of the diverse aggregation profiles with calcium or magnesium owing to different binding sites of the cations to the lipid molecules.
Collapse
Affiliation(s)
- Lisa Rahnfeld
- Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Lessingstrasse 8, 07743 Jena, Germany
| | - Jana Thamm
- Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Lessingstrasse 8, 07743 Jena, Germany
| | - Frank Steiniger
- Electron Microscopy Center, University Hospital Jena, Friedrich Schiller University Jena, Ziegelmuehlenweg 1, 07743 Jena, Germany
| | - Peter van Hoogevest
- Phospholipid Research Center, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Paola Luciani
- Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Lessingstrasse 8, 07743 Jena, Germany.
| |
Collapse
|
16
|
Cobalt and nickel affect the fluidity of negatively-charged biomimetic membranes. Chem Phys Lipids 2018; 210:28-37. [DOI: 10.1016/j.chemphyslip.2017.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/02/2017] [Accepted: 11/24/2017] [Indexed: 01/28/2023]
|
17
|
Cohen BC, Raz C, Shamay A, Argov-Argaman N. Lipid Droplet Fusion in Mammary Epithelial Cells is Regulated by Phosphatidylethanolamine Metabolism. J Mammary Gland Biol Neoplasia 2017; 22:235-249. [PMID: 29188493 DOI: 10.1007/s10911-017-9386-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022] Open
Abstract
Mammary epithelial cells (MEC) secrete fat in the form of milk fat globules (MFG) which are found in milk in diverse sizes. MFG originate from intracellular lipid droplets, and the mechanism underlying their size regulation is still elusive. Two main mechanisms have been suggested to control lipid droplet size. The first is a well-documented pathway, which involves regulation of cellular triglyceride content. The second is the fusion pathway, which is less-documented, especially in mammalian cells, and its importance in the regulation of droplet size is still unclear. Using biochemical and molecular inhibitors, we provide evidence that in MEC, lipid droplet size is determined by fusion, independent of cellular triglyceride content. The extent of fusion is determined by the cell membrane's phospholipid composition. In particular, increasing phosphatidylethanolamine (PE) content enhances fusion between lipid droplets and hence increases lipid droplet size. We further identified the underlying biochemical mechanism that controls this content as the mitochondrial enzyme phosphatidylserine decarboxylase; siRNA knockdown of this enzyme reduced the number of large lipid droplets threefold. Further, inhibition of phosphatidylserine transfer to the mitochondria, where its conversion to PE occurs, diminished the large lipid droplet phenotype in these cells. These results reveal, for the first time to our knowledge in mammalian cells and specifically in mammary epithelium, the missing biochemical link between the metabolism of cellular complex lipids and lipid-droplet fusion, which ultimately defines lipid droplet size.
Collapse
Affiliation(s)
- Bat-Chen Cohen
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel.
| | - Chen Raz
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel
| | - Avi Shamay
- Department of Ruminant Science, Agricultural Research Organization, Volcani Center, Bet Dagan, Israel
| | - Nurit Argov-Argaman
- The Animal Science Department, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, PO Box 12, Rehovot, 76100, Israel
| |
Collapse
|
18
|
Graber ZT, Shi Z, Baumgart T. Cations induce shape remodeling of negatively charged phospholipid membranes. Phys Chem Chem Phys 2017; 19:15285-15295. [PMID: 28569910 PMCID: PMC5562360 DOI: 10.1039/c7cp00718c] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The divalent cation Ca2+ is a key component in many cell signaling and membrane trafficking pathways. Ca2+ signal transduction commonly occurs through interaction with protein partners. However, in this study we show a novel mechanism by which Ca2+ may impact membrane structure. We find an asymmetric concentration of Ca2+ across the membrane triggers deformation of membranes containing negatively charged lipids such as phosphatidylserine (PS) and phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2). Membrane invaginations in vesicles were observed forming away from the leaflet with higher Ca2+ concentration, showing that Ca2+ induces negative curvature. We hypothesize that the negative curvature is produced by Ca2+-induced clustering of PS and PI(4,5)P2. In support of this notion, we find that Ca2+-induced membrane deformation is stronger for membranes containing PI(4,5)P2, which is known to more readily cluster in the presence of Ca2+. The observed Ca2+-induced membrane deformation is strongly influenced by Na+ ions. A high symmetric [Na+] across the membrane reduces Ca2+ binding by electrostatic shielding, inhibiting Ca2+-induced membrane deformation. An asymmetric [Na+] across the membrane, however, can either oppose or support Ca2+-induced deformation, depending on the direction of the gradient in [Na+]. At a sufficiently high asymmetric Na+ concentration it can impact membrane structure in the absence of Ca2+. We propose that Ca2+ works in concert with curvature generating proteins to modulate membrane curvature and shape transitions. This novel structural impact of Ca2+ could be important for Ca2+-dependent cellular processes that involve the creation of membrane curvature, including exocytosis, invadopodia, and cell motility.
Collapse
Affiliation(s)
- Z T Graber
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA.
| | | | | |
Collapse
|
19
|
Roqanian S, Meratan AA, Ahmadian S, Shafizadeh M, Ghasemi A, Karami L. Polyphenols protect mitochondrial membrane against permeabilization induced by HEWL oligomers: Possible mechanism of action. Int J Biol Macromol 2017; 103:709-720. [PMID: 28545969 DOI: 10.1016/j.ijbiomac.2017.05.130] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 01/08/2023]
Abstract
Increasing body of evidence suggests that polyphenols frequently interacting with amyloid aggregates and/or interfering with aggregate species to bind biomembranes may serve as a therapeutic approach for the treatment of amyloid-related diseases. Hence, in the present study, the possible effects of three naturally occurring polyphenols including Curcumin, Quercetin, and Resveratrol on mitochondrial membrane permeabilization induced by Hen Egg White Lysozyme (HEWL) oligomers were investigated. Our results indicated that pre-incubation of mitochondrial homogenate with polyphenols considerably inhibit membrane permeabilization in a concentration dependent manner. In parallel, HEWL oligomers, which were co-incubated with the polyphenols, showed less effectiveness on membrane permeabilization, suggesting that toxicity of oligomers was hindered. Using a range of techniques including fluorescence quenching, Nile red binding assay, zeta potential and size measurements, CD (far- and near-UV) spectroscopy, and molecular docking, we found that the polyphenols, structure-dependently, interact with and induce conformational changes in HEWL oligomers, thereby inhibit their toxicity. We proposed a mechanism by which selected polyphenols induce their protective effects through binding to mitochondria and interfering with HEWL oligomer-membrane interactions and/or by direct interaction with HEWL oligomers, induction of conformational changes, and generating far less toxic species. However, additional studies are needed to elucidate the detailed mechanisms involved.
Collapse
Affiliation(s)
- Shaqayeq Roqanian
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, 1417614411 Tehran, Iran
| | - Ali Akbar Meratan
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran.
| | - Shahin Ahmadian
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, 1417614411 Tehran, Iran.
| | - Mahshid Shafizadeh
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, 1417614411 Tehran, Iran
| | - Atiyeh Ghasemi
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, 1417614411 Tehran, Iran
| | - Leila Karami
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
20
|
Phosphatidylethanolamine dynamics are required for osteoclast fusion. Sci Rep 2017; 7:46715. [PMID: 28436434 PMCID: PMC5402267 DOI: 10.1038/srep46715] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/23/2017] [Indexed: 11/08/2022] Open
Abstract
Osteoclasts, responsible for bone resorption, are multinucleated cells formed by cell-cell fusion of mononuclear pre-osteoclasts. Although osteoclast fusion is a pivotal step for osteoclastogenesis, little is known about the mechanism involved. To clarify the underlying process, we investigated dynamics of membrane phospholipids during osteoclastogenesis in vitro. We found that the cellular content of phospholipids, phosphatidylethanolamine (PE) in particular, was increased during osteoclast differentiation. Furthermore, PE was greatly increased in the outer leaflet of the plasma membrane bilayer during osteoclastogenesis, being concentrated in filopodia involved in cell-cell fusion. Immobilisation of the cell surface PE blocked osteoclast fusion, revealing the importance of PE abundance and distribution. To identify the molecules responsible for these PE dynamics, we screened a wide array of lipid-related genes by quantitative PCR and shRNA-mediated knockdown. Among them, a PE-biosynthetic enzyme, acyl-CoA:lysophosphatidylethanolamine acyltransferase 2 (LPEAT2), and two ATP-binding cassette (ABC) transporters, ABCB4 and ABCG1, were markedly increased during osteoclastogenesis, and their knockdown in pre-osteoclasts led to reduction in PE exposure on the cell surface and subsequent osteoclast fusion. These findings demonstrate that the PE dynamics play an essential role in osteoclast fusion, in which LPEAT2, ABCB4 and ABCG1 are key players for PE biosynthesis and redistribution.
Collapse
|
21
|
Yingchoncharoen P, Kalinowski DS, Richardson DR. Lipid-Based Drug Delivery Systems in Cancer Therapy: What Is Available and What Is Yet to Come. Pharmacol Rev 2016; 68:701-87. [PMID: 27363439 PMCID: PMC4931871 DOI: 10.1124/pr.115.012070] [Citation(s) in RCA: 438] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer is a leading cause of death in many countries around the world. However, the efficacy of current standard treatments for a variety of cancers is suboptimal. First, most cancer treatments lack specificity, meaning that these treatments affect both cancer cells and their normal counterparts. Second, many anticancer agents are highly toxic, and thus, limit their use in treatment. Third, a number of cytotoxic chemotherapeutics are highly hydrophobic, which limits their utility in cancer therapy. Finally, many chemotherapeutic agents exhibit short half-lives that curtail their efficacy. As a result of these deficiencies, many current treatments lead to side effects, noncompliance, and patient inconvenience due to difficulties in administration. However, the application of nanotechnology has led to the development of effective nanosized drug delivery systems known commonly as nanoparticles. Among these delivery systems, lipid-based nanoparticles, particularly liposomes, have shown to be quite effective at exhibiting the ability to: 1) improve the selectivity of cancer chemotherapeutic agents; 2) lower the cytotoxicity of anticancer drugs to normal tissues, and thus, reduce their toxic side effects; 3) increase the solubility of hydrophobic drugs; and 4) offer a prolonged and controlled release of agents. This review will discuss the current state of lipid-based nanoparticle research, including the development of liposomes for cancer therapy, different strategies for tumor targeting, liposomal formulation of various anticancer drugs that are commercially available, recent progress in liposome technology for the treatment of cancer, and the next generation of lipid-based nanoparticles.
Collapse
Affiliation(s)
- Phatsapong Yingchoncharoen
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
22
|
Koyanagi T, Leriche G, Yep A, Onofrei D, Holland GP, Mayer M, Yang J. Effect of Headgroups on Small-Ion Permeability across Archaea-Inspired Tetraether Lipid Membranes. Chemistry 2016; 22:8074-7. [DOI: 10.1002/chem.201601326] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Takaoki Koyanagi
- Department of Chemistry and Biochemistry; University of California San Diego; La Jolla CA 92093-0358 USA
| | - Geoffray Leriche
- Department of Chemistry and Biochemistry; University of California San Diego; La Jolla CA 92093-0358 USA
| | - Alvin Yep
- Department of Chemistry and Biochemistry; University of California San Diego; La Jolla CA 92093-0358 USA
| | - David Onofrei
- Department of Chemistry and Biochemistry; San Diego State University; San Diego CA 92182-1030 USA
| | - Gregory P. Holland
- Department of Chemistry and Biochemistry; San Diego State University; San Diego CA 92182-1030 USA
| | - Michael Mayer
- Adolphe Merkle Institute; University of Fribourg; Chemin des Verdiers 4 1700 Fribourg Switzerland
| | - Jerry Yang
- Department of Chemistry and Biochemistry; University of California San Diego; La Jolla CA 92093-0358 USA
| |
Collapse
|
23
|
Hollander A, Danino D. Cochleate characterization by cryogenic electron microscopy methods: Cryo-TEM and Cryo-SEM. Colloids Surf A Physicochem Eng Asp 2015. [DOI: 10.1016/j.colsurfa.2015.07.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
24
|
Meriney SD, Umbach JA, Gundersen CB. Fast, Ca2+-dependent exocytosis at nerve terminals: shortcomings of SNARE-based models. Prog Neurobiol 2014; 121:55-90. [PMID: 25042638 DOI: 10.1016/j.pneurobio.2014.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 04/14/2014] [Accepted: 07/03/2014] [Indexed: 11/30/2022]
Abstract
Investigations over the last two decades have made major inroads in clarifying the cellular and molecular events that underlie the fast, synchronous release of neurotransmitter at nerve endings. Thus, appreciable progress has been made in establishing the structural features and biophysical properties of the calcium (Ca2+) channels that mediate the entry into nerve endings of the Ca2+ ions that trigger neurotransmitter release. It is now clear that presynaptic Ca2+ channels are regulated at many levels and the interplay of these regulatory mechanisms is just beginning to be understood. At the same time, many lines of research have converged on the conclusion that members of the synaptotagmin family serve as the primary Ca2+ sensors for the action potential-dependent release of neurotransmitter. This identification of synaptotagmins as the proteins which bind Ca2+ and initiate the exocytotic fusion of synaptic vesicles with the plasma membrane has spurred widespread efforts to reveal molecular details of synaptotagmin's action. Currently, most models propose that synaptotagmin interfaces directly or indirectly with SNARE (soluble, N-ethylmaleimide sensitive factor attachment receptors) proteins to trigger membrane fusion. However, in spite of intensive efforts, the field has not achieved consensus on the mechanism by which synaptotagmins act. Concurrently, the precise sequence of steps underlying SNARE-dependent membrane fusion remains controversial. This review considers the pros and cons of the different models of SNARE-mediated membrane fusion and concludes by discussing a novel proposal in which synaptotagmins might directly elicit membrane fusion without the intervention of SNARE proteins in this final fusion step.
Collapse
Affiliation(s)
- Stephen D Meriney
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Joy A Umbach
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Cameron B Gundersen
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA.
| |
Collapse
|
25
|
Rüger R, Tansi FL, Rabenhold M, Steiniger F, Kontermann RE, Fahr A, Hilger I. In vivo near-infrared fluorescence imaging of FAP-expressing tumors with activatable FAP-targeted, single-chain Fv-immunoliposomes. J Control Release 2014; 186:1-10. [PMID: 24810115 DOI: 10.1016/j.jconrel.2014.04.050] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/17/2014] [Accepted: 04/24/2014] [Indexed: 12/01/2022]
Abstract
Molecular and cellular changes that precede the invasive growth of solid tumors include the release of proteolytic enzymes and peptides in the tumor stroma, the recruitment of phagocytic and lymphoid infiltrates and alteration of the extracellular matrix. The reactive tumor stroma consists of a large number of myofibroblasts, characterized by high expression of fibroblast activation protein alpha (FAP). FAP, a type-II transmembrane sialoglycoprotein is an attractive target in diagnosis and therapy of several pathologic disorders especially cancer. In the underlying work, a fluorescence-activatable liposome (fluorescence-quenched during circulation and fluorescence activation upon cellular uptake), bearing specific single-chain Fv fragments directed against FAP (scFv'FAP) was developed, and its potential for use in fluorescence diagnostic imaging of FAP-expressing tumor cells was evaluated by whole body fluorescence imaging. The liposomes termed anti-FAP-IL were prepared via post-insertion of ligand-phospholipid-conjugates into preformed DY-676-COOH-containing liposomes. The anti-FAP-IL revealed a homogeneous size distribution and showed specific interaction and binding with FAP-expressing cells in vitro. The high level of fluorescence quenching of the near-infrared fluorescent dye sequestered in the aqueous interior of the liposomes enables fluorescence imaging exclusively upon uptake and degradation by cells, which results in fluorescence activation. Only FAP-expressing cells were able to take up and activate fluorescence of anti-FAP-IL in vitro. Furthermore, anti-FAP-IL accumulated selectively in FAP-expressing xenograft models in vivo, as demonstrated by blocking experiments using free scFv'FAP. The local tumor fluorescence intensities were in agreement with the intrinsic degree of FAP-expression in different xenograft models. Thus, anti-FAP-IL can serve as a suitable in vivo diagnostic tool for pathological disorders accompanied by high FAP-expression.
Collapse
Affiliation(s)
- Ronny Rüger
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany.
| | - Felista L Tansi
- Dept. of Experimental Radiology, Institute of Diagnostic and Interventional Radiology I, Jena University Hospital-Friedrich Schiller University Jena, Erlanger Allee 101, 07747 Jena, Germany.
| | - Markus Rabenhold
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany
| | - Frank Steiniger
- Center for Electron Microscopy, Jena University Hospital-Friedrich Schiller University Jena, Ziegelmuehlenweg 1, 07743 Jena, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Alfred Fahr
- Department of Pharmaceutical Technology, Friedrich-Schiller-University Jena, Lessingstrasse 8, 07743 Jena, Germany.
| | - Ingrid Hilger
- Dept. of Experimental Radiology, Institute of Diagnostic and Interventional Radiology I, Jena University Hospital-Friedrich Schiller University Jena, Erlanger Allee 101, 07747 Jena, Germany.
| |
Collapse
|
26
|
Bahreman A, Rabe M, Kros A, Bruylants G, Bonnet S. Binding of a ruthenium complex to a thioether ligand embedded in a negatively charged lipid bilayer: a two-step mechanism. Chemistry 2014; 20:7429-38. [PMID: 24782232 DOI: 10.1002/chem.201400377] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Indexed: 01/14/2023]
Abstract
The interaction between the ruthenium polypyridyl complex [Ru(terpy)(dcbpy)(H2O)](2+) (terpy = 2,2';6',2"-terpyridine, dcbpy = 6,6'-dichloro-2,2'-bipyridine) and phospholipid membranes containing either thioether ligands or cholesterol were investigated using UV-visible spectroscopy, Langmuir-Blodgett monolayer surface pressure measurements, and isothermal titration calorimety (ITC). When embedded in a membrane, the thioether ligand coordinated to the dicationic metal complex only when the phospholipids of the membrane were negatively charged, that is, in the presence of attractive electrostatic interaction. In such a case coordination is much faster than in homogeneous conditions. A two-step model for the coordination of the metal complex to the membrane-embedded sulfur ligand is proposed, in which adsorption of the complex to the negative surface of the monolayers or bilayers occurs within minutes, whereas formation of the coordination bond between the surface-bound metal complex and ligand takes hours. Finally, adsorption of the aqua complex to the membrane is driven by entropy. It does not involve insertion of the metal complex into the hydrophobic lipid layer, but rather simple electrostatic adsorption at the water-bilayer interface.
Collapse
Affiliation(s)
- Azadeh Bahreman
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, Leiden, 2300 RA (The Netherlands)
| | | | | | | | | |
Collapse
|
27
|
Yasuhara K, Tsukamoto M, Tsuji Y, Kikuchi JI. Unique concentration dependence on the fusion of anionic liposomes induced by polyethyleneimine. Colloids Surf A Physicochem Eng Asp 2012. [DOI: 10.1016/j.colsurfa.2012.01.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Tsai HHG, Lai WX, Lin HD, Lee JB, Juang WF, Tseng WH. Molecular dynamics simulation of cation–phospholipid clustering in phospholipid bilayers: Possible role in stalk formation during membrane fusion. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:2742-55. [DOI: 10.1016/j.bbamem.2012.05.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 05/11/2012] [Accepted: 05/29/2012] [Indexed: 10/28/2022]
|
29
|
Kapoor M, Burgess DJ. Efficient and safe delivery of siRNA using anionic lipids: Formulation optimization studies. Int J Pharm 2012; 432:80-90. [DOI: 10.1016/j.ijpharm.2012.04.058] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 04/16/2012] [Accepted: 04/21/2012] [Indexed: 10/28/2022]
|
30
|
Mondal Roy S, Sarkar M. Membrane fusion induced by small molecules and ions. J Lipids 2011; 2011:528784. [PMID: 21660306 PMCID: PMC3108104 DOI: 10.1155/2011/528784] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 01/28/2011] [Accepted: 02/25/2011] [Indexed: 01/11/2023] Open
Abstract
Membrane fusion is a key event in many biological processes. These processes are controlled by various fusogenic agents of which proteins and peptides from the principal group. The fusion process is characterized by three major steps, namely, inter membrane contact, lipid mixing forming the intermediate step, pore opening and finally mixing of inner contents of the cells/vesicles. These steps are governed by energy barriers, which need to be overcome to complete fusion. Structural reorganization of big molecules like proteins/peptides, supplies the required driving force to overcome the energy barrier of the different intermediate steps. Small molecules/ions do not share this advantage. Hence fusion induced by small molecules/ions is expected to be different from that induced by proteins/peptides. Although several reviews exist on membrane fusion, no recent review is devoted solely to small moleculs/ions induced membrane fusion. Here we intend to present, how a variety of small molecules/ions act as independent fusogens. The detailed mechanism of some are well understood but for many it is still an unanswered question. Clearer understanding of how a particular small molecule can control fusion will open up a vista to use these moleucles instead of proteins/peptides to induce fusion both in vivo and in vitro fusion processes.
Collapse
Affiliation(s)
- Sutapa Mondal Roy
- Chemical Sciences Division, Saha Institute of Nuclear Physics, Sector 1, Block AF, Bidhannagar, Kolkata 700064, India
| | - Munna Sarkar
- Chemical Sciences Division, Saha Institute of Nuclear Physics, Sector 1, Block AF, Bidhannagar, Kolkata 700064, India
| |
Collapse
|
31
|
Sasai M, Tadokoro S, Hirashima N. Artificial exocytotic system that secretes intravesicular contents upon Ca2+ influx. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2010; 26:14788-14792. [PMID: 20722459 DOI: 10.1021/la102737e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Exocytosis is a crucial process of secreting various signaling molecules such as neurotransmitters, hormones, and other chemical mediators into the extracellular space. Exocytotic release is caused by membrane fusion of intracellular vesicles with the plasma membrane triggered by an increase in intracellular Ca(2+). In the present study, we developed an artificial system of exocytosis that secretes intravesicular contents upon Ca(2+) influx. We prepared artificial secretory cells using cell-sized giant unilamellar liposomal vesicles (GUVs) that contain small liposomes (SUVs) that correspond to secretory vesicles. To observe exocytosis-like secretion in an artificial system, we labeled both an intra-SUV solution and an SUV membrane with a soluble fluorescent dye and a rhodamine-labeled phospholipid, respectively. To induce membrane fusion between SUVs and a GUV as observed in exocytosis, the Ca(2+) concentration of intra-GUV was elevated by incorporating ionomycin (a Ca(2+) ionophore) into the GUV membrane. We succeeded in inducing exocytosis-like secretion by Ca(2+) elevation in a GUV together with the osmolarity difference between the intra-GUV and extra-GUV solutions.
Collapse
Affiliation(s)
- Masao Sasai
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | | | | |
Collapse
|
32
|
Paukner S, Kohl G, Jalava K, Lubitz W. Sealed Bacterial Ghosts—Novel Targeting Vehicles for Advanced Drug Delivery of Water-soluble Substances. J Drug Target 2010. [DOI: 10.3109/10611860310001593366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
33
|
Quinn PJ. A lipid matrix model of membrane raft structure. Prog Lipid Res 2010; 49:390-406. [PMID: 20478335 DOI: 10.1016/j.plipres.2010.05.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 05/06/2010] [Indexed: 12/12/2022]
Abstract
Domains in cell membranes are created by lipid-lipid interactions and are referred to as membrane rafts. Reliable isolation methods have been developed which have shown that rafts from the same membranes have different proteins and can be sub-fractionated by immunoaffinity methods. Analysis of these raft subfractions shows that they are also comprised of different molecular species of lipids. The major lipid classes present are phospholipids, glycosphingolipids and cholesterol. Model studies show that mixtures of phospholipids, particularly sphingomyelin, and cholesterol form liquid-ordered phase with properties intermediate between a gel and fluid phase. This type of liquid-ordered phase dominates theories of domain formation and raft structure in biological membranes. Recently it has been shown that sphingolipids with long (22-26C) N-acyl fatty acids form quasi-crystalline bilayer structures with diacylphospholipids that have well-defined stoichiometries. A two tier heuristic model of membrane raft structure is proposed in which liquid-ordered phase created by a molecular complex between sphingolipids with hydrocarbon chains of approximately equal length and cholesterol acts as a primary staging area for selecting raft proteins. Tailoring of the lipid anchors of raft proteins takes place at this site. Assembly of lipid-anchored proteins on a scaffold of sphingolipids with asymmetric hydrocarbon chains and phospholipids arranged in a quasi-crystalline bilayer structure serves to concentrate and orient the proteins in a manner that couples them functionally within the membrane. Specificity is inherent in the quasi-crystalline lipid structure of liquid-ordered matrices formed by both types of complex into which protein lipid anchors are interpolated. An interaction between the sugar residues of the glycolipids and the raft proteins provides an additional level of specificity that distinguishes one raft from another.
Collapse
Affiliation(s)
- Peter J Quinn
- Biochemistry Department, King's College London, 150 Stamford Street, London, UK.
| |
Collapse
|
34
|
Morris RJ. Ionic control of the metastable inner leaflet of the plasma membrane: Fusions natural and artefactual. FEBS Lett 2009; 584:1665-9. [PMID: 19913542 DOI: 10.1016/j.febslet.2009.11.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 11/02/2009] [Accepted: 11/07/2009] [Indexed: 10/20/2022]
Abstract
The phospholipids of the inner and outer leaflets of the plasma membrane face chemically very different environments, and are specialized to serve different needs. While lipids of the outer leaflet are inherently stable in a lamellar (bilayer) phase, the main lipid of the inner layer, phosphatidylethanolamine (PE), does not form a lamellar phase unless evenly mixed with phosphatidylserine (PS(-)). This mixture can be readily perturbed by factors that include an influx of Ca(2+) that chelates the negatively charged PS(-), thereby destabilizing PE. The implications of this metastability of the inner leaflet for vesicular trafficking, and experimentally for the isolation of detergent-resistant membrane domains (DRMs) at physiological temperature, are considered.
Collapse
Affiliation(s)
- Roger J Morris
- Wolfson Centre for Age-Related Disease, Guy's Campus, King's College London, UK.
| |
Collapse
|
35
|
Björkbom A, Ohvo-Rekilä H, Kankaanpää P, Nyholm TKM, Westerlund B, Slotte JP. Characterization of membrane properties of inositol phosphorylceramide. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1798:453-60. [PMID: 19913494 DOI: 10.1016/j.bbamem.2009.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 10/05/2009] [Accepted: 11/04/2009] [Indexed: 12/11/2022]
Abstract
Inositol phosphorylceramides (IPCs) are a class of anionic sphingolipids with a single inositol-phosphate head group coupled to ceramide. IPCs and more complex glycosylated IPCs have been identified in fungi, plants and protozoa but not in mammals. IPCs have also been identified in detergent resistant membranes in several organisms. Here we report on the membrane properties of the saturated N-palmitoyl-IPC (P-IPC) in one component bilayers as well as in complex bilayers together with 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine and cholesterol. The membrane properties of P-IPC were shown to be affected by calcium. According to anisotropy changes reported by DPH, the gel-to-liquid transition temperature (T(m)) of P-IPC was 48 degrees C. Addition of 5 mM CaCl(2) during vesicle preparation markedly increased the T(m) (65 degrees C). According to fluorescence quenching experiments in complex lipid mixtures, P-IPC formed sterol containing domains in an otherwise fluid environment. The P-IPC containing domains melted at a lower temperature and appeared to contain less sterol as compared to domains containing N-palmitoyl-sphingomyelin. Calcium further reduced the sterol content of the ordered domains and also increased the thermal stability of the domains. Calcium also induced vesicle aggregation of unilamellar vesicles containing P-IPC, as was observed by 4D confocal microscopy and dynamic light scattering. We believe that IPCs and the calcium induced effects could be important in numerous membrane associated cellular processes such as membrane fusion and in membrane raft linked processes.
Collapse
Affiliation(s)
- Anders Björkbom
- Department of Biochemistry and Pharmacy, Abo Akademi University, Tykistökatu 6 A, FI-20520, Finland.
| | | | | | | | | | | |
Collapse
|
36
|
Zhu L, Michel V, Bakovic M. Regulation of the mouse CTP: Phosphoethanolamine cytidylyltransferase gene Pcyt2 during myogenesis. Gene 2009; 447:51-9. [DOI: 10.1016/j.gene.2009.07.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 07/22/2009] [Accepted: 07/22/2009] [Indexed: 10/20/2022]
|
37
|
Abstract
Exocytosis is a highly conserved and essential process. Although numerous proteins are involved throughout the exocytotic process, the defining membrane fusion step appears to occur through a lipid-dominated mechanism. Here we review and integrate the current literature on protein and lipid roles in exocytosis, with emphasis on the multiple roles of cholesterol in exocytosis and membrane fusion, in an effort to promote a more molecular systems-level view of the as yet poorly understood process of Ca2+-triggered membrane mergers.
Collapse
|
38
|
Schultz ZD, Pazos IM, McNeil-Watson FK, Lewis EN, Levin IW. Magnesium-induced lipid bilayer microdomain reorganizations: implications for membrane fusion. J Phys Chem B 2009; 113:9932-41. [PMID: 19603842 PMCID: PMC2754194 DOI: 10.1021/jp9011944] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interactions between dipalmitoylphosphatidylcholine (DPPC) and dipalmitoylphosphatidylserine (DPPS), combined both as binary lipid bilayer assemblies and separately, under the influence of divalent Mg2+, a membrane bilayer fusogenic agent, are reported. Infrared vibrational spectroscopic analyses of the lipid acyl chain methylene symmetric stretching modes indicate that aggregates of the two phospholipid components exist as domains heterogeneously distributed throughout the binary bilayer system. In the presence of Mg2+, DPPS maintains an ordered orthorhombic subcell gel phase structure through the phase transition temperature, while the DPPC component is only minimally perturbed with respect to the gel to liquid crystalline phase change. The addition of Mg2+ induces a reorganization of the lipid domains in which the gel phase acyl chain planes rearrange from a hexagonal configuration toward a triclinic, parallel chain subcell. Examination of the acyl chain methylene deformation modes at low temperatures allows a determination of DPPS microdomain sizes, which decrease upon the addition of DPPC-d62 in the absence of Mg2+. On adding Mg2+, a uniform DPPS domain size is observed in the binary mixtures. In either the presence or absence of Mg2+, DPPC-d62 aggregates remain in a configuration for which microdomain sizes are not spectroscopically measurable. Analysis of the acyl chain methylene deformation modes for DPPC-d62 in the binary system suggests that clusters of the deuterated lipids are distributed throughout the DPPS matrix. Light scattering and fluorescence measurements indicate that Mg2+ induces both the aggregation and the fusion of the lipid assemblies as a function of the ratio of DPPS to DPPC. The structural reorganizations of the lipid microdomains within the DPPS-DPPC bilayer are interpreted in the context of current concepts regarding lipid bilayer fusion.
Collapse
Affiliation(s)
- Zachary D. Schultz
- Laboratory of Chemical Physics, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892
| | - Ileana M. Pazos
- Laboratory of Chemical Physics, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892
| | | | - E. Neil Lewis
- Novel Measurements Group, Malvern Instruments, Ltd., Malvern WR14 1XZ, United Kingdom
| | - Ira W. Levin
- Laboratory of Chemical Physics, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892
| |
Collapse
|
39
|
Isolation at physiological temperature of detergent-resistant membranes with properties expected of lipid rafts: the influence of buffer composition. Biochem J 2008; 417:525-33. [DOI: 10.1042/bj20081385] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The failure of most non-ionic detergents to release patches of DRM (detergent-resistant membrane) at 37 °C undermines the claim that DRMs consist of lipid nanodomains that exist in an Lo (liquid ordered) phase on the living cell surface. In the present study, we have shown that inclusion of cations (Mg2+, K+) to mimic the intracellular environment stabilizes membranes during solubilization sufficiently to allow the isolation of DRMs at 37 °C, using either Triton X-100 or Brij 96. These DRMs are sensitive to chelation of cholesterol, maintain outside-out orientation of membrane glycoproteins, have prolonged (18 h) stability at 37 °C, and are vesicles or sheets up to 150–200 nm diameter. DRMs containing GPI (glycosylphosphatidylinositol)-anchored proteins PrP (prion protein) and Thy-1 can be separated by immunoaffinity isolation, in keeping with their separate organization and trafficking on the neuronal surface. Thy-1, but not PrP, DRMs are associated with actin. EM (electron microscopy) immunohistochemistry shows most PrP, and some Thy-1, to be clustered on DRMs, again maintaining their organization on the neuronal surface. For DRMs labelled for either protein, the bulk of the surface of the DRM is not labelled, indicating that the GPI-anchored protein is a minor component of its lipid domain. These 37 °C DRMs thus have properties expected of raft membrane, yet pose more questions about how proteins are organized within these nanodomains.
Collapse
|
40
|
Chen T, Choi LS, Einstein S, Klippenstein MA, Scherrer P, Cuhis PR. Proton-Induced Permeability and Fusion of Large Unilamellar Vesicles by Covalently Conjugated Poly(2-Ethylacrylic Acid). J Liposome Res 2008. [DOI: 10.3109/08982109909018658] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Yaradanakul A, Wang TM, Lariccia V, Lin MJ, Shen C, Liu X, Hilgemann DW. Massive Ca-induced membrane fusion and phospholipid changes triggered by reverse Na/Ca exchange in BHK fibroblasts. ACTA ACUST UNITED AC 2008; 132:29-50. [PMID: 18562498 PMCID: PMC2442179 DOI: 10.1085/jgp.200709865] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Baby hamster kidney (BHK) fibroblasts increase their cell capacitance by 25-100% within 5 s upon activating maximal Ca influx via constitutively expressed cardiac Na/Ca exchangers (NCX1). Free Ca, measured with fluo-5N, transiently exceeds 0.2 mM with total Ca influx amounting to approximately 5 mmol/liter cell volume. Capacitance responses are half-maximal when NCX1 promotes a free cytoplasmic Ca of 0.12 mM (Hill coefficient approximately 2). Capacitance can return to baseline in 1-3 min, and responses can be repeated several times. The membrane tracer, FM 4-64, is taken up during recovery and can be released at a subsequent Ca influx episode. Given recent interest in signaling lipids in membrane fusion, we used green fluorescent protein (GFP) fusions with phosphatidylinositol 4,5-bisphosphate (PI(4,5)P(2)) and diacylglycerol (DAG) binding domains to analyze phospholipid changes in relation to these responses. PI(4,5)P(2) is rapidly cleaved upon activating Ca influx and recovers within 2 min. However, PI(4,5)P(2) depletion by activation of overexpressed hM1 muscarinic receptors causes only little membrane fusion, and subsequent fusion in response to Ca influx remains massive. Two results suggest that DAG may be generated from sources other than PI(4,5)P in these protocols. First, acylglycerols are generated in response to elevated Ca, even when PI(4,5)P(2) is metabolically depleted. Second, DAG-binding C1A-GFP domains, which are brought to the cell surface by exogenous ligands, translocate rapidly back to the cytoplasm in response to Ca influx. Nevertheless, inhibitors of PLCs and cPLA2, PI(4,5)P(2)-binding peptides, and PLD modification by butanol do not block membrane fusion. The cationic agents, FM 4-64 and heptalysine, bind profusely to the extracellular cell surface during membrane fusion. While this binding might reflect phosphatidylserine (PS) "scrambling" between monolayers, it is unaffected by a PS-binding protein, lactadherin, and by polylysine from the cytoplasmic side. Furthermore, the PS indicator, annexin-V, binds only slowly after fusion. Therefore, we suggest that the luminal surfaces of membrane vesicles that fuse to the plasmalemma may be rather anionic. In summary, our results provide no support for any regulatory or modulatory role of phospholipids in Ca-induced membrane fusion in fibroblasts.
Collapse
Affiliation(s)
- Alp Yaradanakul
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Seguí-Simarro J, Nuez F. Pathways to doubled haploidy: chromosome doubling during androgenesis. Cytogenet Genome Res 2008; 120:358-69. [DOI: 10.1159/000121085] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2007] [Indexed: 01/04/2023] Open
|
43
|
Aguilar PS, Engel A, Walter P. The plasma membrane proteins Prm1 and Fig1 ascertain fidelity of membrane fusion during yeast mating. Mol Biol Cell 2006; 18:547-56. [PMID: 17151357 PMCID: PMC1783792 DOI: 10.1091/mbc.e06-09-0776] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
As for most cell-cell fusion events, the molecular details of membrane fusion during yeast mating are poorly understood. The multipass membrane protein Prm1 is the only known component that acts at the step of bilayer fusion. In its absence, mutant mating pairs lyse or arrest in the mating reaction with tightly apposed plasma membranes. We show that deletion of FIG 1, which controls pheromone-induced Ca(2+) influx, yields similar cell fusion defects. Although extracellular Ca(2+) is not required for efficient cell fusion of wild-type cells, cell fusion in prm1 mutant mating pairs is dramatically reduced when Ca(2+) is removed. This enhanced fusion defect is due to lysis. Time-lapse microscopy reveals that fusion and lysis events initiate with identical kinetics, suggesting that both outcomes result from engagement of the fusion machinery. The yeast synaptotagmin orthologue and Ca(2+) binding protein Tcb3 has a role in reducing lysis of prm1 mutants, which opens the possibility that the observed role of Ca(2+) is to engage a wound repair mechanism. Thus, our results suggest that Prm1 and Fig1 have a role in enhancing membrane fusion and maintaining its fidelity. Their absence results in frequent mating pair lysis, which is counteracted by Ca(2+)-dependent membrane repair.
Collapse
Affiliation(s)
- Pablo S Aguilar
- Howard Hughes Medical Institute, and Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA 94158, USA.
| | | | | |
Collapse
|
44
|
Portereiko MF, Sandaklie-Nikolova L, Lloyd A, Dever CA, Otsuga D, Drews GN. NUCLEAR FUSION DEFECTIVE1 encodes the Arabidopsis RPL21M protein and is required for karyogamy during female gametophyte development and fertilization. PLANT PHYSIOLOGY 2006; 141:957-65. [PMID: 16698901 PMCID: PMC1489897 DOI: 10.1104/pp.106.079319] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Karyogamy, or nuclear fusion, is essential for sexual reproduction. In angiosperms, karyogamy occurs three times: twice during double fertilization of the egg cell and the central cell and once during female gametophyte development when the two polar nuclei fuse to form the diploid central cell nucleus. The molecular mechanisms controlling karyogamy are poorly understood. We have identified nine female gametophyte mutants in Arabidopsis (Arabidopsis thaliana), nuclear fusion defective1 (nfd1) to nfd9, that are defective in fusion of the polar nuclei. In the nfd1 to nfd6 mutants, failure of fusion of the polar nuclei is the only defect detected during megagametogenesis. nfd1 is also affected in karyogamy during double fertilization. Using transmission electron microscopy, we showed that nfd1 nuclei fail to undergo fusion of the outer nuclear membranes. nfd1 contains a T-DNA insertion in RPL21M that is predicted to encode the mitochondrial 50S ribosomal subunit L21, and a wild-type copy of this gene rescues the mutant phenotype. Consistent with the predicted function of this gene, an NFD1-green fluorescent protein fusion protein localizes to mitochondria and the NFD1/RPL21M gene is expressed throughout the plant. The nfd3, nfd4, nfd5, and nfd6 mutants also contain T-DNA insertions in genes predicted to encode proteins that localize to mitochondria, suggesting a role for this organelle in nuclear fusion.
Collapse
|
45
|
Chapter 2: Surface Properties of Liposomes Depending on Their Composition. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/s1554-4516(06)04002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
46
|
Abstract
We have recently proposed a mechanism to describe secretion, a fundamental process in all cells. That hypothesis, called porocytosis, embodies all available data and encompasses both forms of secretion, i.e., vesicular and constitutive. The current accepted view of exocytotic secretion involves the physical fusion of vesicle and plasma membranes; however, that hypothesized mechanism does not fit all available physiological data. Energetics of apposed lipid bilayers do not favor unfacilitated fusion. We consider that calcium ions (e.g., 10(-4) to 10(-3) M calcium in microdomains when elevated for 1 ms or less), whose mobility is restricted in space and time, establish salt bridges among adjacent lipid molecules. This establishes transient pores that span both the vesicle and plasma membrane lipid bilayers; the diameter of this transient pore would be approximately 1 nm (the diameter of a single lipid molecule). The lifetime of the transient pore is completely dependent on the duration of sufficient calcium ion levels. This places the porocytosis hypothesis for secretion squarely in the realm of the physical and physical chemical interactions of calcium and phospholipids and places mass action as the driving force for release of secretory material. The porocytosis hypothesis that we propose satisfies all of the observations and provides a framework to integrate our combined knowledge of vesicular and constitutive secretion.
Collapse
Affiliation(s)
- Robert B Silver
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | | |
Collapse
|
47
|
Epand RF, Martinou JC, Montessuit S, Epand RM. Fatty acids enhance membrane permeabilization by pro-apoptotic Bax. Biochem J 2004; 377:509-16. [PMID: 14535847 PMCID: PMC1223875 DOI: 10.1042/bj20030938] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2003] [Revised: 10/01/2003] [Accepted: 10/09/2003] [Indexed: 11/17/2022]
Abstract
Fatty acids are known promoters of apoptosis. In the present study, the direct role of fatty acids with regard to their ability to cause membrane permeabilization by Bax was explored. Addition of fatty acids to liposomes in the presence of cations greatly enhanced the permeabilizing activity of Bax, a pro-apoptotic Bcl-2 protein. This provides a putative mechanism for the role of fatty acids in apoptosis. It is not a result of detergent-like properties of fatty acids, since a different micelle-forming amphiphile, dilysocardiolipin, was strongly inhibitory. We also demonstrate that there is a synergistic effect on Bax-induced permeabilization between Ca(2+) and Mg(2+), both on the binding of Bax to liposomes as well as on the induction of the leakage of liposomal contents. Micromolar concentrations of Ca(2+) added externally or submicromolar concentrations of free Ca(2+) present in the medium were sufficient to promote Bax-induced permeabilization synergistically with externally added Mg(2+). These results indicate that Bax can induce leakage from liposomes at ion concentrations resembling those found physiologically. The synergistic effects of Ca(2+) and Mg(2+) were observed with liposomes with different lipid compositions. Thus the action of Bax is strongly modulated by the presence of bivalent cations that can act synergistically, as well as by micelle-forming lipid components that can be either stimulatory or inhibitory.
Collapse
Affiliation(s)
- Raquel F Epand
- Department of Biochemistry, McMaster University Health Sciences Centre, Hamilton, ON, Canada L8N 3Z5.
| | | | | | | |
Collapse
|
48
|
Affiliation(s)
- Andreas Hofmann
- Macromolecular Crystallography Laboratory, NCI at Frederick, 539 Boyles Street, Frederick, Maryland 21702, USA
| | | |
Collapse
|
49
|
Abstract
Itraconazole is a drug which is poorly soluble in both the water and oil phases of emulsions. Incorporation in parenteral emulsions was performed applying the SolEmuls Technology, i.e. localising the drug in the interfacial lecithin layer of the emulsions by homogenising a hybrid dispersion of oil droplets and drug nanocrystals in water. The maximum loading capacity of the emulsion system was found to be 10 mg/ml; at 20 mg/ml the loading capacity was exceeded leading to remaining drug nanocrystals in the emulsion. Incorporation of itraconazole into the lecithin layer led to an enhanced dispersion effect, i.e. with increasing drug concentration the droplet size of the emulsions decreased. Physical long-term stability of the optimum emulsion with 10 mg/ml could be shown over a period of 3 months at room temperature.
Collapse
Affiliation(s)
- Aslihan Akkar
- Department of Pharmaceutical Technology, Biotechnology and Quality management, Free University of Berlin, Berlin, Germany.
| | | |
Collapse
|
50
|
Abstract
Disparate biological processes involve fusion of two membranes into one and fission of one membrane into two. To formulate the possible job description for the proteins that mediate remodeling of biological membranes, we analyze the energy price of disruption and bending of membrane lipid bilayers at the different stages of bilayer fusion. The phenomenology and the pathways of the well-characterized reactions of biological remodeling, such as fusion mediated by influenza hemagglutinin, are compared with those studied for protein-free bilayers. We briefly consider some proteins involved in fusion and fission, and the dependence of remodeling on the lipid composition of the membranes. The specific hypothetical mechanisms by which the proteins can lower the energy price of the bilayer rearrangement are discussed in light of the experimental data and the requirements imposed by the elastic properties of the bilayer.
Collapse
Affiliation(s)
- Leonid V Chernomordik
- Section on Membrane Biology, Laboratory of Cellular and Molecular Biophysics, NICHD, National Institutes of Health, 10 Center Drive, Bethesda, Maryland 20892-1855, USA.
| | | |
Collapse
|