Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Rep ; 12(1): 17520, 2022 Oct 20.
Article in English | MEDLINE | ID: covidwho-2077118

ABSTRACT

SiRNA is a new generation of drug molecules and a new approach for treating a variety of diseases such as cancer and viral infections. SiRNA delivery to cells and translocation into cytoplasm are the main challenges in the clinical application of siRNA. Lipid carriers are one of the most successful carriers for siRNA delivery. In this study, we investigated the interaction of siRNA with a zwitterionic bilayer and how ion concentration and lipid conjugation can affect it. The divalent cation such as Mg2+ ions could promote the siRNA adsorption on the bilayer surface. The cation ions can bind to the head groups of lipids and the grooves of siRNA molecules and form bridges between the siRNA and bilayer surface. Our findings demonstrated the bridges formed by divalent ions could facilitate the attachment of siRNA to the membrane surface. We showed that the divalent cations can regulate the bridging-driven membrane attachment and it seems the result of this modulation can be used for designing biomimetic devices. In the following, we examined the effect of cations on the interaction between siRNA modified by cholesterol and the membrane surface. Our MD simulations showed that in the presence of Mg2+, the electrostatic and vdW energy between the membrane and siRNA were higher compared to those in the presence of NA+. We showed that the electrostatic interaction between membrane and siRNA cannot be facilitated only by cholesterol conjugated. Indeed, cations are essential to create coulomb repulsion and enable membrane attachment. This study provides important insight into liposome carriers for siRNA delivery and could help us in the development of siRNA-based therapeutics. Due to the coronavirus pandemic outbreak, these results may shed light on the new approach for treating these diseases and their molecular details.


Subject(s)
Lipid Bilayers , Molecular Dynamics Simulation , RNA, Small Interfering/genetics , Lipid Bilayers/metabolism , Liposomes , Cations, Divalent , Cell Membrane/metabolism , Cations , Cholesterol
2.
ACS Sens ; 7(7): 2084-2092, 2022 07 22.
Article in English | MEDLINE | ID: covidwho-1900429

ABSTRACT

With the rapid spread and multigeneration variation of coronavirus, rapid drug development has become imperative. A major obstacle to addressing this issue is adequately constructing the cell membrane at the molecular level, which enables in vitro observation of the cell response to virus and drug molecules quantitatively, shortening the drug experiment cycle. Herein, we propose a rapid and label-free supported lipid bilayer-based lab-on-a-chip biosensor for the screening of effective inhibition drugs. An extended gate electrode was prepared and functionalized by an angiotensin-converting enzyme II (ACE2) receptor-incorporated supported lipid bilayer (SLB). Such an integrated system can convert the interactions of targets and membrane receptors into real-time charge signals. The platform can simulate the cell membrane microenvironment in vitro and accurately capture the interaction signal between the target and the cell membrane with minimized interference, thus observing the drug action pathway quantitatively and realizing drug screening effectively. Due to these label-free, low-cost, convenient, and integrated advantages, it is a suitable candidate method for the rapid drug screening for the early treatment and prevention of worldwide spread of coronavirus.


Subject(s)
Biosensing Techniques , Coronavirus , Cell Membrane/metabolism , Coronavirus/metabolism , Lab-On-A-Chip Devices , Lipid Bilayers/metabolism
3.
Nucleic Acid Ther ; 32(5): 361-368, 2022 10.
Article in English | MEDLINE | ID: covidwho-1864945

ABSTRACT

RNA therapeutics, including siRNAs, antisense oligonucleotides, and other oligonucleotides, have great potential to selectively treat a multitude of human diseases, from cancer to COVID to Parkinson's disease. RNA therapeutic activity is mechanistically driven by Watson-Crick base pairing to the target gene RNA without the requirement of prior knowledge of the protein structure, function, or cellular location. However, before widespread use of RNA therapeutics becomes a reality, we must overcome a billion years of evolutionary defenses designed to keep invading RNAs from entering cells. Unlike small-molecule therapeutics that are designed to passively diffuse across the cell membrane, macromolecular RNA therapeutics are too large, too charged, and/or too hydrophilic to passively diffuse across the cellular membrane and are instead taken up into cells by endocytosis. However, similar to the cell membrane, endosomes comprise a lipid bilayer that entraps 99% or more of RNA therapeutics, even in semipermissive tissues such as the liver, central nervous system, and muscle. Consequently, before RNA therapeutics can achieve their ultimate clinical potential to treat widespread human disease, the rate-limiting delivery problem of endosomal escape must be solved in a clinically acceptable manner.


Subject(s)
COVID-19 , Lipid Bilayers , Humans , Lipid Bilayers/metabolism , COVID-19/genetics , COVID-19/therapy , Endosomes/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , RNA, Small Interfering/chemistry , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Oligonucleotides, Antisense/metabolism , Oligonucleotides/metabolism
4.
Commun Biol ; 5(1): 507, 2022 05 26.
Article in English | MEDLINE | ID: covidwho-1864775

ABSTRACT

Protein-lipid interactions are vital for numerous transmembrane signaling pathways. However, simple tools to characterize these interactions remain scarce and are much needed to advance our understanding of signal transduction across lipid bilayers. To tackle this challenge, we herein engineer nanodisc as a robust fluorescent sensor for reporting membrane biochemical reactions. We circularize nanodiscs via split GFP and thereby create an intensity-based fluorescent sensor (isenND) for detecting membrane binding and remodeling events. We show that isenND responds robustly and specifically to the action of a diverse array of membrane-interacting proteins and peptides, ranging from synaptotagmin and synuclein involved in neurotransmission to viral fusion peptides of HIV-1 and SARS-CoV-2. Together, isenND can serve as a versatile biochemical reagent useful for basic and translational research of membrane biology.


Subject(s)
COVID-19 , Nanostructures , Biophysical Phenomena , Coloring Agents , Humans , Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Nanostructures/chemistry , SARS-CoV-2
5.
Int J Mol Sci ; 23(5)2022 Feb 23.
Article in English | MEDLINE | ID: covidwho-1736941

ABSTRACT

The fourth enzymatic reaction in the de novo pyrimidine biosynthesis, the oxidation of dihydroorotate to orotate, is catalyzed by dihydroorotate dehydrogenase (DHODH). Enzymes belonging to the DHODH Class II are membrane-bound proteins that use ubiquinones as their electron acceptors. We have designed this study to understand the interaction of an N-terminally truncated human DHODH (HsΔ29DHODH) and the DHODH from Escherichia coli (EcDHODH) with ubiquinone (Q10) in supported lipid membranes using neutron reflectometry (NR). NR has allowed us to determine in situ, under solution conditions, how the enzymes bind to lipid membranes and to unambiguously resolve the location of Q10. Q10 is exclusively located at the center of all of the lipid bilayers investigated, and upon binding, both of the DHODHs penetrate into the hydrophobic region of the outer lipid leaflet towards the Q10. We therefore show that the interaction between the soluble enzymes and the membrane-embedded Q10 is mediated by enzyme penetration. We can also show that EcDHODH binds more efficiently to the surface of simple bilayers consisting of 1-palmitoyl, 2-oleoyl phosphatidylcholine, and tetraoleoyl cardiolipin than HsΔ29DHODH, but does not penetrate into the lipids to the same degree. Our results also highlight the importance of Q10, as well as lipid composition, on enzyme binding.


Subject(s)
Dihydroorotate Dehydrogenase/chemistry , Dihydroorotate Dehydrogenase/metabolism , Escherichia coli/enzymology , Lipid Bilayers/metabolism , Ubiquinone/metabolism , Cardiolipins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Phosphatidylcholines/metabolism , Protein Conformation , Protein Domains
6.
J Am Chem Soc ; 144(7): 2968-2979, 2022 02 23.
Article in English | MEDLINE | ID: covidwho-1683928

ABSTRACT

Coronavirus disease-2019 (COVID-19), a potentially lethal respiratory illness caused by the coronavirus SARS-CoV-2, emerged in the end of 2019 and has since spread aggressively across the globe. A thorough understanding of the molecular mechanisms of cellular infection by coronaviruses is therefore of utmost importance. A critical stage in infection is the fusion between viral and host membranes. Here, we present a detailed investigation of the role of selected SARS-CoV-2 Spike fusion peptides, and the influence of calcium and cholesterol, in this fusion process. Structural information from specular neutron reflectometry and small angle neutron scattering, complemented by dynamics information from quasi-elastic and spin-echo neutron spectroscopy, revealed strikingly different functions encoded in the Spike fusion domain. Calcium drives the N-terminal of the Spike fusion domain to fully cross the host plasma membrane. Removing calcium, however, reorients the peptide back to the lipid leaflet closest to the virus, leading to significant changes in lipid fluidity and rigidity. In conjunction with other regions of the fusion domain, which are also positioned to bridge and dehydrate viral and host membranes, the molecular events leading to cell entry by SARS-CoV-2 are proposed.


Subject(s)
Lipid Bilayers/metabolism , Peptide Fragments/metabolism , SARS-CoV-2/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Amino Acid Sequence , Cholesterol/chemistry , Lipid Bilayers/chemistry , Membrane Fluidity , Neutron Diffraction , Protein Domains , Scattering, Small Angle , Spike Glycoprotein, Coronavirus/chemistry , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism
7.
J Extracell Vesicles ; 10(14): e12170, 2021 12.
Article in English | MEDLINE | ID: covidwho-1555701

ABSTRACT

SARS-CoV-2 spike protein (S) binds to human angiotensin-converting enzyme 2 (hACE2), allowing virus to dock on cell membrane follow by viral entry. Here, we use high-speed atomic force microscopy (HS-AFM) for real-time visualization of S, and its interaction with hACE2 and small extracellular vesicles (sEVs). Results show conformational heterogeneity of S, flexibility of S stalk and receptor-binding domain (RBD), and pH/temperature-induced conformational change of S. S in an S-ACE2 complex appears as an all-RBD up conformation. The complex acquires a distinct topology upon acidification. S and S2 subunit demonstrate different membrane docking mechanisms on sEVs. S-hACE2 interaction facilitates S to dock on sEVs, implying the feasibility of ACE2-expressing sEVs for viral neutralization. In contrary, S2 subunit docks on lipid layer and enters sEV using its fusion peptide, mimicking the viral entry scenario. Altogether, our study provides a platform that is suitable for real-time visualization of various entry inhibitors, neutralizing antibodies, and sEV-based decoy in blocking viral entry. Teaser: Comprehensive observation of SARS-CoV-2 spike and its interaction with receptor ACE2 and sEV-based decoy in real time using HS-AFM.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Extracellular Vesicles/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Humans , Hydrogen-Ion Concentration , Lipid Bilayers/metabolism , Microscopy, Atomic Force , Protein Binding , Protein Conformation , Protein Domains , Protein Subunits , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/chemistry , Temperature , Virus Internalization
8.
Sci Rep ; 11(1): 22288, 2021 11 15.
Article in English | MEDLINE | ID: covidwho-1517638

ABSTRACT

Numerous repositioned drugs have been sought to decrease the severity of SARS-CoV-2 infection. It is known that among its physicochemical properties, Ursodeoxycholic Acid (UDCA) has a reduction in surface tension and cholesterol solubilization, it has also been used to treat cholesterol gallstones and viral hepatitis. In this study, molecular docking was performed with the SARS-CoV-2 Spike protein and UDCA. In order to confirm this interaction, we used Molecular Dynamics (MD) in "SARS-CoV-2 Spike protein-UDCA". Using another system, we also simulated MD with six UDCA residues around the Spike protein at random, naming this "SARS-CoV-2 Spike protein-6UDCA". Finally, we evaluated the possible interaction between UDCA and different types of membranes, considering the possible membrane conformation of SARS-CoV-2, this was named "SARS-CoV-2 membrane-UDCA". In the "SARS-CoV-2 Spike protein-UDCA", we found that UDCA exhibits affinity towards the central region of the Spike protein structure of - 386.35 kcal/mol, in a region with 3 alpha helices, which comprises residues from K986 to C1032 of each monomer. MD confirmed that UDCA remains attached and occasionally forms hydrogen bonds with residues R995 and T998. In the presence of UDCA, we observed that the distances between residues atoms OG1 and CG2 of T998 in the monomers A, B, and C in the prefusion state do not change and remain at 5.93 ± 0.62 and 7.78 ± 0.51 Å, respectively, compared to the post-fusion state. Next, in "SARS-CoV-2 Spike protein-6UDCA", the three UDCA showed affinity towards different regions of the Spike protein, but only one of them remained bound to the region between the region's heptad repeat 1 and heptad repeat 2 (HR1 and HR2) for 375 ps of the trajectory. The RMSD of monomer C was the smallest of the three monomers with a value of 2.89 ± 0.32, likewise, the smallest RMSF was also of the monomer C (2.25 ± 056). In addition, in the simulation of "SARS-CoV-2 membrane-UDCA", UDCA had a higher affinity toward the virion-like membrane; where three of the four residues remained attached once they were close (5 Å, to the centre of mass) to the membrane by 30 ns. However, only one of them remained attached to the plasma-like membrane and this was in a cluster of cholesterol molecules. We have shown that UDCA interacts in two distinct regions of Spike protein sequences. In addition, UDCA tends to stay bound to the membrane, which could potentially reduce the internalization of SARS-CoV-2 in the host cell.


Subject(s)
Antiviral Agents/metabolism , Drug Repositioning/methods , Lipid Bilayers/metabolism , Molecular Docking Simulation/methods , Phospholipids/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Ursodeoxycholic Acid/metabolism , Antiviral Agents/chemistry , COVID-19/metabolism , COVID-19/virology , Humans , Hydrogen Bonding , Membrane Fusion , Molecular Dynamics Simulation , Protein Binding , Protein Conformation, alpha-Helical , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Ursodeoxycholic Acid/chemistry , Virion/metabolism
9.
Sci Rep ; 11(1): 14867, 2021 07 21.
Article in English | MEDLINE | ID: covidwho-1320237

ABSTRACT

SARS-CoV-2 spike proteins are responsible for the membrane fusion event, which allows the virus to enter the host cell and cause infection. This process starts with the binding of the spike extramembrane domain to the angiotensin-converting enzyme 2 (ACE2), a membrane receptor highly abundant in the lungs. In this study, the extramembrane domain of SARS-CoV-2 Spike (sSpike) was injected on model membranes formed by supported lipid bilayers in presence and absence of the soluble part of receptor ACE2 (sACE2), and the structural features were studied at sub-nanometer level by neutron reflection. In all cases the presence of the protein produced a remarkable degradation of the lipid bilayer. Indeed, both for membranes from synthetic and natural lipids, a significant reduction of the surface coverage was observed. Quartz crystal microbalance measurements showed that lipid extraction starts immediately after sSpike protein injection. All measurements indicate that the presence of proteins induces the removal of membrane lipids, both in the presence and in the absence of ACE2, suggesting that sSpike molecules strongly associate with lipids, and strip them away from the bilayer, via a non-specific interaction. A cooperative effect of sACE2 and sSpike on lipid extraction was also observed.


Subject(s)
COVID-19/virology , Lipid Bilayers/metabolism , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Binding Sites , COVID-19/metabolism , COVID-19/physiopathology , Cell Line , Humans , Membrane Fusion/physiology , Neutron Diffraction/methods , Protein Binding , Protein Domains , Spike Glycoprotein, Coronavirus/chemistry , Virus Internalization
10.
J Biol Chem ; 297(2): 100940, 2021 08.
Article in English | MEDLINE | ID: covidwho-1293905

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 envelope protein (S2-E) is a conserved membrane protein that is important for coronavirus (CoV) assembly and budding. Here, we describe the recombinant expression and purification of S2-E in amphipol-class amphipathic polymer solutions, which solubilize and stabilize membrane proteins, but do not disrupt membranes. We found that amphipol delivery of S2-E to preformed planar bilayers results in spontaneous membrane integration and formation of viroporin cation channels. Amphipol delivery of the S2-E protein to human cells results in plasma membrane integration, followed by retrograde trafficking to the trans-Golgi network and accumulation in swollen perinuclear lysosomal-associated membrane protein 1-positive vesicles, likely lysosomes. CoV envelope proteins have previously been proposed to manipulate the luminal pH of the trans-Golgi network, which serves as an accumulation station for progeny CoV particles prior to cellular egress via lysosomes. Delivery of S2-E to cells will enable chemical biological approaches for future studies of severe acute respiratory syndrome coronavirus 2 pathogenesis and possibly even development of "Trojan horse" antiviral therapies. Finally, this work also establishes a paradigm for amphipol-mediated delivery of membrane proteins to cells.


Subject(s)
Cell Membrane/drug effects , Coronavirus Envelope Proteins/metabolism , Polymers/pharmacology , Propylamines/pharmacology , Surface-Active Agents/pharmacology , trans-Golgi Network/metabolism , Cell Membrane/metabolism , Coronavirus Envelope Proteins/genetics , HeLa Cells , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Lysosomes/metabolism , Polymers/chemistry , Propylamines/chemistry , Protein Transport , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface-Active Agents/chemistry
11.
J Chem Phys ; 154(24): 245101, 2021 Jun 28.
Article in English | MEDLINE | ID: covidwho-1293030

ABSTRACT

Ethanol is highly effective against various enveloped viruses and can disable the virus by disintegrating the protective envelope surrounding it. The interactions between the coronavirus envelope (E) protein and its membrane environment play key roles in the stability and function of the viral envelope. By using molecular dynamics simulation, we explore the underlying mechanism of ethanol-induced disruption of a model coronavirus membrane and, in detail, interactions of the E-protein and lipids. We model the membrane bilayer as N-palmitoyl-sphingomyelin and 1-palmitoyl-2-oleoylphosphatidylcholine lipids and the coronavirus E-protein. The study reveals that ethanol causes an increase in the lateral area of the bilayer along with thinning of the bilayer membrane and orientational disordering of lipid tails. Ethanol resides at the head-tail region of the membrane and enhances bilayer permeability. We found an envelope-protein-mediated increase in the ordering of lipid tails. Our simulations also provide important insights into the orientation of the envelope protein in a model membrane environment. At ∼25 mol. % of ethanol in the surrounding ethanol-water phase, we observe disintegration of the lipid bilayer and dislocation of the E-protein from the membrane environment.


Subject(s)
Cell Membrane/drug effects , Cell Membrane/metabolism , Coronavirus/metabolism , Disinfectants/pharmacology , Ethanol/pharmacology , Viral Envelope Proteins/metabolism , Coronavirus/physiology , Lipid Bilayers/metabolism , Molecular Conformation , Molecular Dynamics Simulation , Permeability
12.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: covidwho-1143517

ABSTRACT

The interactions at the atomic level between small molecules and the main components of cellular plasma membranes are crucial for elucidating the mechanisms allowing for the entrance of such small species inside the cell. We have performed molecular dynamics and metadynamics simulations of tryptophan, serotonin, and melatonin at the interface of zwitterionic phospholipid bilayers. In this work, we will review recent computer simulation developments and report microscopic properties, such as the area per lipid and thickness of the membranes, atomic radial distribution functions, angular orientations, and free energy landscapes of small molecule binding to the membrane. Cholesterol affects the behaviour of the small molecules, which are mainly buried in the interfacial regions. We have observed a competition between the binding of small molecules to phospholipids and cholesterol through lipidic hydrogen-bonds. Free energy barriers that are associated to translational and orientational changes of melatonin have been found to be between 10-20 kJ/mol for distances of 1 nm between melatonin and the center of the membrane. Corresponding barriers for tryptophan and serotonin that are obtained from reversible work methods are of the order of 10 kJ/mol and reveal strong hydrogen bonding between such species and specific phospholipid sites. The diffusion of tryptophan and melatonin is of the order of 10-7 cm2/s for the cholesterol-free and cholesterol-rich setups.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/chemistry , Cholesterol/chemistry , Dimyristoylphosphatidylcholine/chemistry , Melatonin/chemistry , Serotonin/chemistry , Tryptophan/chemistry , 1,2-Dipalmitoylphosphatidylcholine/metabolism , Cholesterol/metabolism , Dimyristoylphosphatidylcholine/metabolism , Hydrogen Bonding , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Melatonin/metabolism , Molecular Dynamics Simulation , Serotonin/metabolism , Solutions , Static Electricity , Thermodynamics , Tryptophan/metabolism , Water/chemistry
13.
Biochim Biophys Acta Biomembr ; 1863(6): 183584, 2021 06 01.
Article in English | MEDLINE | ID: covidwho-1071093

ABSTRACT

This work investigates how docosahexaenoic acid (DHA) modifies the effect of Cholesterol (Chol) on the structural and dynamical properties of dipalmitoylphosphatidylcholine (DPPC) membrane. We employ low-cost and non-invasive methods: zeta potential (ZP), conductivity, density, and ultrasound velocity, complemented by molecular dynamics simulations. Our studies reveal that 30% of DHA added to the DPPC-Chol system tends to revert Chol action on a model lipid bilayer. Results obtained in this work shed light on the effect of polyunsaturated fatty acids - particularly DHA - on lipid membranes, with potential preventive applications in many diseases, e.g. neuronal as, Alzheimer's disease, and viral, as Covid-19.


Subject(s)
Cholesterol/metabolism , Docosahexaenoic Acids/metabolism , Lipid Bilayers/metabolism , Phosphatidylcholines/metabolism , Liposomes , Molecular Structure , Temperature , Ultrasonic Waves
SELECTION OF CITATIONS
SEARCH DETAIL