Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Biophys J ; 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38444158

ABSTRACT

Many membrane proteins are modulated by cholesterol. Here we report profound effects of cholesterol depletion and restoration on the human voltage-gated proton channel, hHV1, in excised patches but negligible effects in the whole-cell configuration. Despite the presence of a putative cholesterol-binding site, a CARC motif in hHV1, mutation of this motif did not affect cholesterol effects. The murine HV1 lacks a CARC sequence but displays similar cholesterol effects. These results argue against a direct effect of cholesterol on the HV1 protein. However, the data are fully explainable if HV1 preferentially associates with cholesterol-dependent lipid domains, or "rafts." The rafts would be expected to concentrate in the membrane/glass interface and to be depleted from the electrically accessible patch membrane. This idea is supported by evidence that HV1 channels can diffuse between seal and patch membranes when suction is applied. Simultaneous truncation of the large intracellular N and C termini of hHV1 greatly attenuated the cholesterol effect, but C truncation alone did not; this suggests that the N terminus is the region of attachment to lipid domains. Searching for abundant raft-associated proteins led to stomatin. Co-immunoprecipitation experiment results were consistent with hHV1 binding to stomatin. The stomatin-mediated association of HV1 with cholesterol-dependent lipid domains provides a mechanism for cells to direct HV1 to subcellular locations where it is needed, such as the phagosome in leukocytes.

2.
Soft Matter ; 19(29): 5487-5501, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37434554

ABSTRACT

The effect of cholesterol on biological membranes is important in biochemistry. In this study, a polymer system is used to simulate the consequences of varying cholesterol content in membranes. The system consists of an AB-diblock copolymer, a hydrophilic homopolymer hA, and a hydrophobic rigid homopolymer C, corresponding to phospholipid, water, and cholesterol, respectively. The effect of the C-polymer content on the membrane is studied within the framework of a self-consistent field model. The results show that the liquid-crystal behavior of B and C has a great influence on the chemical potential of cholesterol in bilayer membranes. The effects of the interaction strength between components, characterized by the Flory-Huggins parameters and the Maier-Saupe parameter, were studied. Some consequences of adding a coil headgroup to the C-rod are presented. Results of our model are compared to experimental findings for cholesterol-containing lipid bilayer membranes.


Subject(s)
Molecular Mimicry , Cholesterol/chemistry , Lipid Bilayers/chemistry , Polymers/chemistry , Hydrophobic and Hydrophilic Interactions , Crystallins/chemistry
3.
PLoS Pathog ; 17(9): e1009488, 2021 09.
Article in English | MEDLINE | ID: mdl-34492091

ABSTRACT

Arenavirus entry into host cells occurs through a low pH-dependent fusion with late endosomes that is mediated by the viral glycoprotein complex (GPC). The mechanisms of GPC-mediated membrane fusion and of virus targeting to late endosomes are not well understood. To gain insights into arenavirus fusion, we examined cell-cell fusion induced by the Old World Lassa virus (LASV) GPC complex. LASV GPC-mediated cell fusion is more efficient and occurs at higher pH with target cells expressing human LAMP1 compared to cells lacking this cognate receptor. However, human LAMP1 is not absolutely required for cell-cell fusion or LASV entry. We found that GPC-induced fusion progresses through the same lipid intermediates as fusion mediated by other viral glycoproteins-a lipid curvature-sensitive intermediate upstream of hemifusion and a hemifusion intermediate downstream of acid-dependent steps that can be arrested in the cold. Importantly, GPC-mediated fusion and LASV pseudovirus entry are specifically augmented by an anionic lipid, bis(monoacylglycero)phosphate (BMP), which is highly enriched in late endosomes. This lipid also specifically promotes cell fusion mediated by Junin virus GPC, an unrelated New World arenavirus. We show that BMP promotes late steps of LASV fusion downstream of hemifusion-the formation and enlargement of fusion pores. The BMP-dependence of post-hemifusion stages of arenavirus fusion suggests that these viruses evolved to use this lipid as a cofactor to selectively fuse with late endosomes.


Subject(s)
Endosomes/metabolism , Lassa Fever/metabolism , Lassa virus/physiology , Lysophospholipids/metabolism , Monoglycerides/metabolism , Virus Internalization , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Viral Envelope Proteins/metabolism
4.
Sci Rep ; 10(1): 7499, 2020 May 05.
Article in English | MEDLINE | ID: mdl-32372013

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Sci Rep ; 10(1): 4746, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32179788

ABSTRACT

Ginkgolic acids (GA) are alkylphenol constituents of the leaves and fruits of Ginkgo biloba. GA has shown pleiotropic effects in vitro, including: antitumor effects through inhibition of lipogenesis; decreased expression of invasion associated proteins through AMPK activation; and potential rescue of amyloid-ß (Aß) induced synaptic impairment. GA was also reported to have activity against Escherichia coli and Staphylococcus aureus. Several mechanisms for this activity have been suggested including: SUMOylation inhibition; blocking formation of the E1-SUMO intermediate; inhibition of fatty acid synthase; non-specific SIRT inhibition; and activation of protein phosphatase type-2C. Here we report that GA inhibits Herpes simplex virus type 1 (HSV-1) by inhibition of both fusion and viral protein synthesis. Additionally, we report that GA inhibits human cytomegalovirus (HCMV) genome replication and Zika virus (ZIKV) infection of normal human astrocytes (NHA). We show a broad spectrum of fusion inhibition by GA of all three classes of fusion proteins including HIV, Ebola virus (EBOV), influenza A virus (IAV) and Epstein Barr virus (EBV). In addition, we show inhibition of a non-enveloped adenovirus. Our experiments suggest that GA inhibits virion entry by blocking the initial fusion event. Data showing inhibition of HSV-1 and CMV replication, when GA is administered post-infection, suggest a possible secondary mechanism targeting protein and DNA synthesis. Thus, in light of the strong effect of GA on viral infection, even after the infection begins, it may potentially be used to treat acute infections (e.g. Coronavirus, EBOV, ZIKV, IAV and measles), and also topically for the successful treatment of active lesions (e.g. HSV-1, HSV-2 and varicella-zoster virus (VZV)).


Subject(s)
Antiviral Agents/pharmacology , DNA Virus Infections/metabolism , DNA Viruses/drug effects , RNA Virus Infections/metabolism , RNA Viruses/drug effects , Salicylates/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Viral Fusion Proteins/antagonists & inhibitors , Animals , Astrocytes/metabolism , Chlorocebus aethiops , DNA Replication/drug effects , DNA Virus Infections/virology , DNA Viruses/genetics , DNA, Viral/genetics , HEK293 Cells , Humans , RNA Virus Infections/virology , RNA Viruses/genetics , Vero Cells , Viral Envelope Proteins/biosynthesis , Viral Fusion Proteins/biosynthesis , Virion/drug effects , Virus Internalization/drug effects , Virus Replication/drug effects
6.
PLoS Pathog ; 15(1): e1007532, 2019 01.
Article in English | MEDLINE | ID: mdl-30640957

ABSTRACT

Late endosome-resident interferon-induced transmembrane protein 3 (IFITM3) inhibits fusion of diverse viruses, including Influenza A virus (IAV), by a poorly understood mechanism. Despite the broad antiviral activity of IFITM3, viruses like Lassa virus (LASV), are fully resistant to its inhibitory effects. It is currently unclear whether resistance arises from a highly efficient fusion machinery that is capable of overcoming IFITM3 restriction or the ability to enter from cellular sites devoid of this factor. Here, we constructed and validated a functional IFITM3 tagged with EGFP or other fluorescent proteins. This breakthrough allowed live cell imaging of virus co-trafficking and fusion with endosomal compartments in cells expressing fluorescent IFITM3. Three-color single virus and endosome tracking revealed that sensitive (IAV), but not resistant (LASV), viruses become trapped within IFITM3-positive endosomes where they underwent hemifusion but failed to release their content into the cytoplasm. IAV fusion with IFITM3-containing compartments could be rescued by amphotericin B treatment, which has been previously shown to antagonize the antiviral activity of this protein. By comparison, virtually all LASV particles trafficked and fused with endosomes lacking detectable levels of fluorescent IFITM3, implying that this virus escapes restriction by utilizing endocytic pathways that are distinct from the IAV entry pathways. The importance of virus uptake and transport pathways is further reinforced by the observation that LASV glycoprotein-mediated cell-cell fusion is inhibited by IFITM3 and other members of the IFITM family expressed in target cells. Together, our results strongly support a model according to which IFITM3 accumulation at the sites of virus fusion is a prerequisite for its antiviral activity and that this protein traps viral fusion at a hemifusion stage by preventing the formation of fusion pores. We conclude that the ability to utilize alternative endocytic pathways for entry confers IFITM3-resistance to otherwise sensitive viruses.


Subject(s)
Endosomes/metabolism , Membrane Proteins/metabolism , Membrane Proteins/physiology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , A549 Cells/metabolism , Animals , Antiviral Agents/metabolism , COS Cells/metabolism , Chlorocebus aethiops , Endosomes/virology , HEK293 Cells/metabolism , Host-Pathogen Interactions , Humans , Influenza A virus/pathogenicity , Interferons/metabolism , Lassa virus/pathogenicity , Optical Imaging/methods , Protein Transport , Virus Internalization
7.
Biophys J ; 114(4): 904-918, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29490250

ABSTRACT

Cholesterol is abundant in plasma membranes and exhibits a variety of interactions throughout the membrane. Chemical potential accounts for thermodynamic consequences of molecular interactions, and quantifies the effective concentration (i.e., activity) of any substance participating in a process. We have developed, to our knowledge, the first method to measure cholesterol chemical potential in plasma membranes. This was accomplished by complexing methyl-ß-cyclodextrin with cholesterol in an aqueous solution and equilibrating it with an organic solvent containing dissolved cholesterol. The chemical potential of cholesterol was thereby equalized in the two phases. Because cholesterol is dilute in the organic phase, here activity and concentration were equivalent. This equivalence allowed the amount of cholesterol bound to methyl-ß-cyclodextrin to be converted to cholesterol chemical potential. Our method was used to determine the chemical potential of cholesterol in erythrocytes and in plasma membranes of nucleated cells in culture. For erythrocytes, the chemical potential did not vary when the concentration was below a critical value. Above this value, the chemical potential progressively increased with concentration. We used standard cancer lines to characterize cholesterol chemical potential in plasma membranes of nucleated cells. This chemical potential was significantly greater for highly metastatic breast cancer cells than for nonmetastatic breast cancer cells. Chemical potential depended on density of the cancer cells. A method to alter and fix the cholesterol chemical potential to any value (i.e., a cholesterol chemical potential clamp) was also developed. Cholesterol content did not change when cells were clamped for 24-48 h. It was found that the level of activation of the transcription factor STAT3 increased with increasing cholesterol chemical potential. The cholesterol chemical potential may regulate signaling pathways.


Subject(s)
Breast Neoplasms/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Erythrocytes/metabolism , beta-Cyclodextrins/metabolism , Breast Neoplasms/pathology , Cell Membrane/chemistry , Cholesterol/chemistry , Female , Humans , Neoplasm Metastasis , Signal Transduction , Thermodynamics , Tumor Cells, Cultured , beta-Cyclodextrins/chemistry
8.
Nat Microbiol ; 1(6): 16050, 2016 04 18.
Article in English | MEDLINE | ID: mdl-27572837

ABSTRACT

Influenza A virus haemagglutinin conformational change drives the membrane fusion of viral and endosomal membranes at low pH. Membrane fusion proceeds through an intermediate called hemifusion(1,2). For viral fusion, the hemifusion structures are not determined(3). Here, influenza virus-like particles(4) carrying wild-type haemagglutinin or haemagglutinin hemifusion mutant G1S(5) and liposome mixtures were studied at low pH by Volta phase plate cryo-electron tomography, which improves the signal-to-noise ratio close to focus. We determined two distinct hemifusion structures: a hemifusion diaphragm and a novel structure termed a 'lipidic junction'. Liposomes with lipidic junctions were ruptured with membrane edges stabilized by haemagglutinin. The rupture frequency and hemifusion diaphragm diameter were not affected by G1S mutation, but decreased when the cholesterol level in the liposomes was close to physiological concentrations. We propose that haemagglutinin induces a merger between the viral and target membranes by one of two independent pathways: a rupture-insertion pathway leading to the lipidic junction and a hemifusion-stalk pathway leading to a fusion pore. The latter is relevant under the conditions of influenza virus infection of cells. Cholesterol concentration functions as a pathway switch because of its negative spontaneous curvature in the target bilayer, as determined by continuum analysis.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Membrane Fusion , Membranes/chemistry , Cholesterol/analysis , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hydrogen-Ion Concentration , Lipid Bilayers/chemistry , Liposomes/chemistry , Membranes/virology , Mutation , Physical Phenomena , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism
9.
Biophys J ; 110(5): 1028-32, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26958878
10.
Biophys J ; 110(5): 1110-24, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26958888

ABSTRACT

We use continuum mechanics to calculate an entire least energy pathway of membrane fusion, from stalk formation, to pore creation, and through fusion pore enlargement. The model assumes that each structure in the pathway is axially symmetric. The static continuum stalk structure agrees quantitatively with experimental stalk architecture. Calculations show that in a stalk, the distal monolayer is stretched and the stored stretching energy is significantly less than the tilt energy of an unstretched distal monolayer. The string method is used to determine the energy of the transition barriers that separate intermediate states and the dynamics of two bilayers as they pass through them. Hemifusion requires a small amount of energy independently of lipid composition, while direct transition from a stalk to a fusion pore without a hemifusion intermediate is highly improbable. Hemifusion diaphragm expansion is spontaneous for distal monolayers containing at least two lipid components, given sufficiently negative diaphragm spontaneous curvature. Conversely, diaphragms formed from single-component distal monolayers do not expand without the continual injection of energy. We identify a diaphragm radius, below which central pore expansion is spontaneous. For larger diaphragms, prior studies have shown that pore expansion is not axisymmetric, and here our calculations supply an upper bound for the energy of the barrier against pore formation. The major energy-requiring deformations in the steps of fusion are: widening of a hydrophobic fissure in bilayers for stalk formation, splay within the expanding hemifusion diaphragm, and fissure widening initiating pore formation in a hemifusion diaphragm.


Subject(s)
Membrane Fusion , Hydrophobic and Hydrophilic Interactions , Lipid Bilayers/chemistry , Porosity , Thermodynamics
12.
PLoS Pathog ; 12(1): e1005373, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26730950

ABSTRACT

Ebola virus (EBOV) is a highly pathogenic filovirus that causes hemorrhagic fever in humans and animals. Currently, how EBOV fuses its envelope membrane within an endosomal membrane to cause infection is poorly understood. We successfully measure cell-cell fusion mediated by the EBOV fusion protein, GP, assayed by the transfer of both cytoplasmic and membrane dyes. A small molecule fusion inhibitor, a neutralizing antibody, as well as mutations in EBOV GP known to reduce viral infection, all greatly reduce fusion. By monitoring redistribution of small aqueous dyes between cells and by electrical capacitance measurements, we discovered that EBOV GP-mediated fusion pores do not readily enlarge-a marked difference from the behavior of other viral fusion proteins. EBOV GP must be cleaved by late endosome-resident cathepsins B or L in order to become fusion-competent. Cleavage of cell surface-expressed GP appears to occur in endosomes, as evidenced by the fusion block imposed by cathepsin inhibitors, agents that raise endosomal pH, or an inhibitor of anterograde trafficking. Treating effector cells with a recombinant soluble cathepsin B or thermolysin, which cleaves GP into an active form, increases the extent of fusion, suggesting that a fraction of surface-expressed GP is not cleaved. Whereas the rate of fusion is increased by a brief exposure to acidic pH, fusion does occur at neutral pH. Importantly, the extent of fusion is independent of external pH in experiments in which cathepsin activity is blocked and EBOV GP is cleaved by thermolysin. These results imply that low pH promotes fusion through the well-known pH-dependent activity of cathepsins; fusion induced by cleaved EBOV GP is a process that is fundamentally independent of pH. The cell-cell fusion system has revealed some previously unappreciated features of EBOV entry, which could not be readily elucidated in the context of endosomal entry.


Subject(s)
Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Viral Fusion Proteins/metabolism , Virus Internalization , Animals , Blotting, Western , COS Cells , Cathepsins/metabolism , Chlorocebus aethiops , Flow Cytometry , Fluorescent Antibody Technique , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Immunohistochemistry , Patch-Clamp Techniques
13.
Virology ; 488: 202-15, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26655238

ABSTRACT

Ebola virus (EBOV) is a highly pathogenic filovirus that causes hemorrhagic fever in humans and animals. Here we provide evidence that cell-cell contact promotes infection mediated by the glycoprotein (GP) of EBOV. Interestingly, expression of EBOV GP alone, even in the absence of retroviral Gag-Pol, is sufficient to transfer a retroviral vector encoding Tet-off from cell to cell. Cell-to-cell infection mediated by EBOV GP is blocked by inhibitors of actin polymerization, but appears to be less sensitive to KZ52 neutralization. Treatment of co-cultured cells with cathepsin B/L inhibitors, or an entry inhibitor 3.47 that targets the receptor NPC1 for virus binding, also blocks cell-to-cell infection. Cell-cell contact also enhances spread of rVSV bearing GP in monocytes and macrophages, the primary targets of natural EBOV infection. Altogether, our study reveals that cell-cell contact promotes EBOV GP-mediated infection, and provides new insight into understanding EBOV spread and viral pathogenesis.


Subject(s)
Ebolavirus/physiology , Viral Envelope Proteins/metabolism , Virus Internalization , Carrier Proteins/metabolism , Cell Line , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Niemann-Pick C1 Protein , Receptors, Virus/metabolism
14.
Phys Rev Lett ; 115(8): 088101, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26340212

ABSTRACT

The mechanism responsible for domain registration in two membrane leaflets has thus far remained enigmatic. Using continuum elasticity theory, we show that minimum line tension is achieved along the rim between thicker (ordered) and thinner (disordered) domains by shifting the rims in opposing leaflets by a few nanometers relative to each other. Increasing surface tension yields an increase in line tension, resulting in larger domains. Because domain registration is driven by lipid deformation energy, it does not require special lipid components or interactions at the membrane midplane.


Subject(s)
Membrane Lipids/chemistry , Models, Biological , Models, Chemical , Cell Membrane/chemistry , Cell Membrane/metabolism , Cholesterol/chemistry , Cholesterol/metabolism , Elasticity , Membrane Lipids/metabolism , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Surface Tension , Thermodynamics
15.
Phys Fluids (1994) ; 26(2): 023101, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24711690

ABSTRACT

A problem for fluid flow around an axisymmetric spherical surface with a hole is presented to characterize pore dynamics in liposomes. A rotational stream function for the contraction of a punctured plane region is obtained and is used in the perturbation expansion for a stream function in the case of a spherical surface with a hole of small radius compared to the spherical radius. The Rayleigh dissipation function is calculated and used to infer the aqueous friction induced by the contraction of the hole. The theoretical aqueous friction coefficient is compared with one derived from experimental data, and they are in agreement.

16.
PLoS One ; 8(10): e76174, 2013.
Article in English | MEDLINE | ID: mdl-24124539

ABSTRACT

Voltage dependence of fusion induced by class II and class III viral fusion proteins was investigated. Class II proteins from Ross River and Sindbus virus and a mutant class III protein from Epstein Barr virus were found to induce cell-cell fusion that is voltage dependent. Combined with previous studies, in all, four class II and two class III protein have now been shown to exhibit voltage-dependent fusion, demonstrating that this is probably a general phenomenon for these two classes of viral fusion proteins. In the present study, monitoring fusion of pseudovirus expressing Vesicular Stomatitis virus (VSV) G within endosomes shows that here, too, fusion is voltage dependent. This supports the claim that voltage dependence of fusion is biologically relevant and that cell-cell fusion reliably models the voltage dependence. Fusion induced by class I viral proteins is independent of voltage; chimeras expressing the ectodomain of a class I fusion protein and the transmembrane domain of VSV G could therefore be used to explore the location within the protein responsible for voltage dependence. Results showed that the transmembrane domain is the region associated with voltage dependence. Experiments in which cells were enriched with acidic lipids led to the conclusion that it is the flip-flop of acidic lipids that carries the charge responsible for the observed voltage dependence of fusion. This flip-flop occurred downstream of hemifusion, in accord with previous findings that the voltage dependent steps of fusion occur at a stage subsequent to hemifusion.


Subject(s)
Viral Proteins/chemistry , Viral Proteins/metabolism , Cell Line , Humans , Vesicular stomatitis Indiana virus/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Viral Proteins/genetics
17.
PLoS Pathog ; 9(1): e1003124, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23358889

ABSTRACT

The interferon-inducible transmembrane (IFITM) protein family represents a new class of cellular restriction factors that block early stages of viral replication; the underlying mechanism is currently not known. Here we provide evidence that IFITM proteins restrict membrane fusion induced by representatives of all three classes of viral membrane fusion proteins. IFITM1 profoundly suppressed syncytia formation and cell-cell fusion induced by almost all viral fusion proteins examined; IFITM2 and IFITM3 also strongly inhibited their fusion, with efficiency somewhat dependent on cell types. Furthermore, treatment of cells with IFN also markedly inhibited viral membrane fusion and entry. By using the Jaagsiekte sheep retrovirus envelope and influenza A virus hemagglutinin as models for study, we showed that IFITM-mediated restriction on membrane fusion is not at the steps of receptor- and/or low pH-mediated triggering; instead, the creation of hemifusion was essentially blocked by IFITMs. Chlorpromazine (CPZ), a chemical known to promote the transition from hemifusion to full fusion, was unable to rescue the IFITM-mediated restriction on fusion. In contrast, oleic acid (OA), a lipid analog that generates negative spontaneous curvature and thereby promotes hemifusion, virtually overcame the restriction. To explore the possible effect of IFITM proteins on membrane molecular order and fluidity, we performed fluorescence labeling with Laurdan, in conjunction with two-photon laser scanning and fluorescence-lifetime imaging microscopy (FLIM). We observed that the generalized polarizations (GPs) and fluorescence lifetimes of cell membranes expressing IFITM proteins were greatly enhanced, indicating higher molecularly ordered and less fluidized membranes. Collectively, our data demonstrated that IFITM proteins suppress viral membrane fusion before the creation of hemifusion, and suggested that they may do so by reducing membrane fluidity and conferring a positive spontaneous curvature in the outer leaflets of cell membranes. Our study provides novel insight into the understanding of how IFITM protein family restricts viral membrane fusion and infection.


Subject(s)
Antigens, Differentiation/immunology , Influenza A virus/immunology , Virus Internalization , Animals , Antigens, Viral/immunology , COS Cells , Cricetinae , Cricetulus , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Jaagsiekte sheep retrovirus/immunology , Pulmonary Adenomatosis, Ovine/immunology , Sheep , Viral Envelope Proteins
18.
Article in English | MEDLINE | ID: mdl-24483480

ABSTRACT

A numerical gradient flow procedure was devised to characterize minimal energy shapes of fusion pores connecting two parallel planar bilayer membranes. Pore energy, composed of splay, tilt, and stretching, was obtained by modeling each bilayer as two monolayers and treating each monolayer of a bilayer membrane as a freely deformable surface described with a mean lipid orientation field. Voids between the two monolayers were prevented by a steric penalty formulation. Pore shapes were assumed to possess both axial and reflectional symmetry. For fixed pore radius and bilayer separation, the gradient flow procedure was applied to initially toroidal pore shapes. Using initially elliptical pore shapes yielded the same final shape. The resulting minimal pore shapes and energies were analyzed as a function of pore dimension and lipid composition. Previous studies either assumed or confined pore shapes, thereby tacitly supplying an unspecified amount of energy to maintain shape. The shapes derived in the present study were outputs of calculations and an externally provided energy was not supplied. Our procedure therefore yielded energy minima significantly lower than those reported in prior studies. The membrane of minimal energy pores bowed outward near the pore lumen, yielding a pore length that exceeded the distance between the two fusing membranes.


Subject(s)
Cell Membrane/metabolism , Lipid Bilayers/metabolism , Mechanical Phenomena , Membrane Fusion , Models, Biological , Biomechanical Phenomena , Porosity , Thermodynamics
19.
Biophys J ; 100(8): 1960-8, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21504732

ABSTRACT

T-20/Fuzeon/Enfuvirtide (ENF), a peptide inhibitor of HIV-1 infection, targets the grooves created by heptad repeat 2 (HR2) of Env's coiled-coil, but mutants resistant to ENF emerge. In this study, ENF-resistant mutants--V38A, N43D, N43D/S138A, Q40H/L45M--were combined with modified inhibitory peptides to identify what we believe to be novel ways to improve peptide efficacy. V38A did not substantially reduce infectivity, but was relatively resistant to inhibitory peptides. N43D was more resistant to inhibitory peptides than wild-type, but infectivity was reduced. The additional mutation S138A (N43D/S138A) increased infectivity and further reduced peptide inhibitory potency. It is concluded that S138A increased binding of HR2/ENF into grooves and that S138A compensated for electrostatic repulsion between N43D and HR2. The six-helix bundle structure indicated that E148A should increase hydrophobic interactions between the coiled-coil and peptide. Importantly, the modifications S138A and E148A in the same peptide retained potency against ENF-escape mutants. The double mutant's increase in potency was greater than the increases from the sum of S138A and E148A individually, showing that these two altered residues synergistically contributed to peptide binding. Isothermal titration calorimetry established that hydrophobic substitutions at positions S138 and E148 improved potency of inhibitory peptides against escape mutants by increasing enthalpic release of energy upon peptide binding.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/pharmacology , HIV-1/drug effects , Hydrophobic and Hydrophilic Interactions , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , env Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Amino Acid Sequence , Amino Acid Substitution , Anti-HIV Agents/metabolism , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Drug Synergism , Enfuvirtide , Entropy , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/metabolism , HIV-1/genetics , HIV-1/pathogenicity , Models, Molecular , Molecular Sequence Data , Mutation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Stability , Protein Structure, Secondary , Repetitive Sequences, Amino Acid , Sequence Homology, Amino Acid , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...