Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Langmuir ; 36(5): 1258-1265, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31961695

ABSTRACT

The ability of amphipathic polymers to self-assemble with lipids and form nanodiscs has been a boon for the field of functional reconstitution of membrane proteins. In a field dominated by detergent micelles, a unique feature of polymer nanodiscs is their much-desired ability to align in the presence of an external magnetic field. Magnetic alignment facilitates the application of solid-state nuclear magnetic resonance (NMR) spectroscopy and aids in the measurement of residual dipolar couplings via well-established solution NMR spectroscopy. In this study, we comprehensively investigate the magnetic alignment properties of styrene maleimide quaternary ammonium (SMA-QA) polymer-based nanodiscs by using 31P and 14N solid-state NMR experiments under static conditions. The results reported herein demonstrate the spontaneous magnetic alignment of large-sized (≥20 nm diameter) SMA-QA nanodiscs (also called as macro-nanodiscs) with the lipid bilayer normal perpendicular to the magnetic field direction. Consequently, the orientation of macro-nanodiscs is further shown to flip the alignment axis parallel to the magnetic field direction upon the addition of a paramagnetic lanthanide salt. These results demonstrate the use of SMA-QA polymer nanodiscs for solid-state NMR applications including structural studies on membrane proteins.


Subject(s)
Lipid Bilayers/chemistry , Maleimides/chemistry , Nanostructures/chemistry , Polystyrenes/chemistry , Quaternary Ammonium Compounds/chemistry , Chlorides/chemistry , Dimyristoylphosphatidylcholine/chemistry , Magnetic Phenomena , Magnetic Resonance Spectroscopy/methods , Nitrogen Isotopes/chemistry , Phosphorus/chemistry , Ytterbium/chemistry
2.
Biochim Biophys Acta Biomembr ; 1860(9): 1793-1802, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29555190

ABSTRACT

Type II diabetes mellitus (T2DM) is characterized by the presence of amyloid deposits of the human islet amyloid polypeptide (hIAPP) in pancreatic ß-cells. A wealth of data supports the hypothesis that hIAPP's toxicity is related to an abnormal interaction of amyloids with islet cell membranes. Thus, many studies aimed at finding effective therapies for T2DM focus on the design of molecules that are able to inhibit hIAPP's amyloid growth and the related membrane damage as well. Based on this view and inspired by its known anti-amyloid properties, we have functionalized resveratrol with a phosphoryl moiety (4'-O-PR) that improves its solubility and pharmacological properties. A second resveratrol derivative has also been obtained by conjugating resveratrol with a dimyristoylphosphatidyl moiety (4'-DMPR). The use of both compounds resulted in abolishing both amyloid growth and amyloid mediated POPC/POPS membrane damage in tube tests. We propose that a mixture of a water-soluble anti-aggregating compound and its lipid-anchored derivative may be employed as a general strategy to prevent and/or to halt amyloid-related membrane damage.

3.
Biochemistry ; 54(10): 1897-907, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25715195

ABSTRACT

Multidrug resistance against the existing antibiotics is becoming a global threat, and any potential drug that can be designed using cationic antimicrobial peptides (AMP) could be an alternate solution to alleviate this existing problem. The mechanism of action of killing bacteria by an AMP differs drastically in comparison to that of small molecule antibiotics. The main target of AMPs is to interact with the lipid bilayer of the cell membrane and disrupt it to kill bacteria. Consequently, the modes of membrane interaction that lead to the selectivity of an AMP are very important to understand. Here, we have used different membrane compositions, such as negatively charged, zwitterionic, or mixed large unilamellar vesicles (LUVs), to study the interaction of four different synthetically designed cationic, linear antimicrobial peptides: MSI-78 (commercially known as pexiganan), MSI-367, MSI-594, and MSI-843. Our solid-state nuclear magnetic resonance (NMR) experiments confirmed that the MSI peptides fragmented LUVs through a detergent-like carpet mechanism depending on the amino acid sequence of the MSI peptide and/or the membrane composition of LUVs. Interestingly, the fragmented lipid aggregates such as SUVs or micelles are sufficiently small to produce an isotropic peak in the (31)P NMR spectrum. These fragmented lipid aggregates contain only MSI peptides bestowed with lipid molecules as confirmed by NMR in conjunction with circular dichroism spectroscopy. Our results also demonstrate that cholesterol, which is present only in the eukaryotic cell membrane, inhibits the MSI-induced fragmentation of LUVs, suggesting that the MSI peptides can discriminate the bacteria and the eukaryotic cell membranes, and this selectivity could be used for further development of novel antibiotics.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Cholesterol/chemistry , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Unilamellar Liposomes/chemistry , Bacteria , Cell Membrane/chemistry , Drug Resistance, Multiple, Bacterial
4.
Langmuir ; 31(4): 1496-504, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25565453

ABSTRACT

Three-dimensional structure determination of membrane proteins is important to fully understand their biological functions. However, obtaining a high-resolution structure has been a major challenge mainly due to the difficulties in retaining the native folding and function of membrane proteins outside of the cellular membrane environment. These challenges are acute if the protein contains a large soluble domain, as it needs bulk water unlike the transmembrane domains of an integral membrane protein. For structural studies on such proteins either by nuclear magnetic resonance (NMR) spectroscopy or X-ray crystallography, bicelles have been demonstrated to be superior to conventional micelles, yet their temperature restrictions attributed to their thermal instabilities are a major disadvantage. Here, we report an approach to overcome this drawback through searching for an optimum combination of bicellar compositions. We demonstrate that bicelles composed of 1,2-didecanoyl-sn-glycero-3-phosphocholine (DDPC) and 1,2-diheptanoyl-sn-glycero-3-phosphocholin (DHepPC), without utilizing additional stabilizing chemicals, are quite stable and are resistant to temperature variations. These temperature-resistant bicelles have a robust bicellar phase and magnetic alignment over a broad range of temperatures, between -15 and 80 °C, retain the native structure of a membrane protein, and increase the sensitivity of solid-state NMR experiments performed at low temperatures. Advantages of two-dimensional separated-local field (SLF) solid-state NMR experiments at a low temperature are demonstrated on magnetically aligned bicelles containing an electron carrier membrane protein, cytochrome b5. Morphological information on different DDPC-based bicellar compositions, varying q ratio/size, and hydration levels obtained from (31)P NMR experiments in this study is also beneficial for a variety of biophysical and spectroscopic techniques, including solution NMR and magic-angle-spinning (MAS) NMR for a wide range of temperatures.


Subject(s)
Membrane Proteins/chemistry , Micelles , Nuclear Magnetic Resonance, Biomolecular/methods , Temperature , Protein Conformation
5.
Biochemistry ; 52(19): 3254-63, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23590672

ABSTRACT

The potency and selectivity of many antimicrobial peptides (AMPs) are correlated with their ability to interact with and disrupt the bacterial cell membrane. In vitro experiments using model membranes have been used to determine the mechanism of membrane disruption of AMPs. Because the mechanism of action of an AMP depends on the ability of the model membrane to accurately mimic the cell membrane, it is important to understand the effect of membrane composition. Anionic lipids that are present in the outer membrane of prokaryotes but are less common in eukaryotic membranes are usually thought to be key for the bacterial selectivity of AMPs. We show by fluorescence measurements of peptide-induced membrane permeabilization that the presence of anionic lipids at high concentrations can actually inhibit membrane disruption by the AMP MSI-78 (pexiganan), a representative of a large class of highly cationic AMPs. Paramagnetic quenching studies suggest MSI-78 is in a surface-associated inactive mode in anionic sodium dodecyl sulfate micelles but is in a deeply buried and presumably more active mode in zwitterionic dodecylphosphocholine micelles. Furthermore, a switch in mechanism occurs with lipid composition. Membrane fragmentation with MSI-78 can be observed in mixed vesicles containing both anionic and zwitterionic lipids but not in vesicles composed of a single lipid of either type. These findings suggest membrane affinity and membrane permeabilization are not always correlated, and additional effects that may be more reflective of the actual cellular environment can be seen as the complexity of the model membranes is increased.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Membrane Lipids/chemistry , Antimicrobial Cationic Peptides/chemistry , Bacteria/chemistry , Bacteria/drug effects , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane Permeability , Lipid Bilayers/chemistry , Membranes, Artificial , Micelles , Nuclear Magnetic Resonance, Biomolecular , Static Electricity
6.
Biophys J ; 103(4): 702-10, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22947931

ABSTRACT

Disruption of cell membranes by Aß is believed to be one of the key components of Aß toxicity. However, the mechanism by which this occurs is not fully understood. Here, we demonstrate that membrane disruption by Aß occurs by a two-step process, with the initial formation of ion-selective pores followed by nonspecific fragmentation of the lipid membrane during amyloid fiber formation. Immediately after the addition of freshly dissolved Aß(1-40), defects form on the membrane that share many of the properties of Aß channels originally reported from single-channel electrical recording, such as cation selectivity and the ability to be blockaded by zinc. By contrast, subsequent amyloid fiber formation on the surface of the membrane fragments the membrane in a way that is not cation selective and cannot be stopped by zinc ions. Moreover, we observed that the presence of ganglioside enhances both the initial pore formation and the fiber-dependent membrane fragmentation process. Whereas pore formation by freshly dissolved Aß(1-40) is weakly observed in the absence of gangliosides, fiber-dependent membrane fragmentation can only be observed in their presence. These results provide insights into the toxicity of Aß and may aid in the design of specific compounds to alleviate the neurodegeneration of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Multimerization , Cell Membrane/chemistry , Cell Membrane Permeability/drug effects , Gangliosides/metabolism , Porosity , Protein Structure, Secondary , Solubility , Zinc/metabolism
7.
Biochemistry ; 51(39): 7676-84, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22970795

ABSTRACT

The toxicity of amyloid-forming peptides has been hypothesized to reside in the ability of protein oligomers to interact with and disrupt the cell membrane. Much of the evidence for this hypothesis comes from in vitro experiments using model membranes. However, the accuracy of this approach depends on the ability of the model membrane to accurately mimic the cell membrane. The effect of membrane composition has been overlooked in many studies of amyloid toxicity in model systems. By combining measurements of membrane binding, membrane permeabilization, and fiber formation, we show that lipids with the phosphatidylethanolamine (PE) headgroup strongly modulate the membrane disruption induced by IAPP (islet amyloid polypeptide protein), an amyloidogenic protein involved in type II diabetes. Our results suggest that PE lipids hamper the interaction of prefibrillar IAPP with membranes but enhance the membrane disruption correlated with the growth of fibers on the membrane surface via a detergent-like mechanism. These findings provide insights into the mechanism of membrane disruption induced by IAPP, suggesting a possible role of PE and other amyloids involved in other pathologies.


Subject(s)
Cell Membrane/metabolism , Islet Amyloid Polypeptide/metabolism , Liposomes/metabolism , Phosphatidylethanolamines/metabolism , Amino Acid Sequence , Cell Membrane/chemistry , Cell Membrane/pathology , Circular Dichroism , Humans , Islet Amyloid Polypeptide/chemistry , Liposomes/chemistry , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Permeability , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/chemistry
8.
J Clin Neurol ; 8(2): 139-45, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22787498

ABSTRACT

BACKGROUND AND PURPOSE: Charcot-Marie-Tooth disease (CMT) type 1A (CMT1A) is the demyelinating form of CMT that is significantly associated with PMP22 duplication. Some studies have found that the disease-related disabilities of these patients are correlated with their compound muscle action potentials (CMAPs), while others have suggested that they are related to the nerve conduction velocities. In the present study, we investigated the correlations between the disease-related disabilities and the electrophysiological values in a large cohort of Korean CMT1A patients. METHODS: We analyzed 167 CMT1A patients of Korean origin with PMP22 duplication using clinical and electrophysiological assessments, including the CMT neuropathy score and the functional disability scale. RESULTS: Clinical motor disabilities were significantly correlated with the CMAPs but not the motor nerve conduction velocities (MNCVs). Moreover, the observed sensory impairments matched the corresponding reductions in the sensory nerve action potentials (SNAPs) but not with slowing of the sensory nerve conduction velocities (SNCVs). In addition, CMAPs were strongly correlated with the disease duration but not with the age at onset. The terminal latency index did not differ between CMT1A patients and healthy controls. CONCLUSIONS: In CMT1A patients, disease-related disabilities such as muscle wasting and sensory impairment were strongly correlated with CMAPs and SNAPs but not with the MNCVs or SNCVs. Therefore, we suggest that the clinical disabilities of CMT patients are determined by the extent of axonal dysfunction.

9.
J Phys Chem B ; 115(2): 366-75, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21171655

ABSTRACT

Cationic, amphiphilic polymers are currently being used as antimicrobial agents that disrupt biomembranes, although their mechanisms remain poorly understood. Herein, membrane association and disruption by amphiphilic polymers bearing primary, tertiary, or quaternary ammonium salt groups reveal the role of cationic group structure in the polymer-membrane interaction. The dissociation constants of polymers to liposomes of POPC were obtained by a fluorometric assay, exploiting the environmental sensitivity of dansyl moieties in the polymer end groups. Dye leakage from liposomes and solid-state NMR provided further insights into the polymer-induced membrane disruption. Interestingly, the polymers with primary amine groups induced reorganization of the bilayer structure to align lipid headgroups perpendicular to the membrane. The results showed that polymers bearing primary amines exceed the tertiary and quaternary ammonium counterparts in membrane binding and disrupting abilities. This is likely due to enhanced complexation of primary amines to the phosphate groups in the lipids, through a combination of hydrogen bonding and electrostatic interactions.


Subject(s)
Anti-Infective Agents/chemistry , Lipid Bilayers/chemistry , Liposomes/chemistry , Phosphatidylcholines/chemistry , Polymers/chemistry , Surface-Active Agents/chemistry , Amines/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Binding Sites , Cations/chemistry , Cell Membrane/chemistry , Cell Membrane/drug effects , Drug Delivery Systems/methods , Fluorescent Dyes/analysis , Fluorometry , Hydrogen Bonding , Lipid Bilayers/metabolism , Liposomes/metabolism , Magnetic Resonance Spectroscopy , Phosphates/chemistry , Phosphatidylcholines/metabolism , Polymers/metabolism , Polymers/pharmacology , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/metabolism , Quaternary Ammonium Compounds/pharmacology , Surface-Active Agents/metabolism , Surface-Active Agents/pharmacology
10.
Biochemistry ; 49(50): 10595-605, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21062093

ABSTRACT

In a minimalist design approach, a synthetic peptide MSI-367 [(KFAKKFA)(3)-NH(2)] was designed and synthesized with the objective of generating cell-selective nonlytic peptides, which have a significant bearing on cell targeting. The peptide exhibited potent activity against both bacteria and fungi, but no toxicity to human cells at micromolar concentrations. Bacterial versus human cell membrane selectivity of the peptide was determined via membrane permeabilization assays. Circular dichroism investigations revealed the intrinsic helix propensity of the peptide, ß-turn structure in aqueous buffer and extended and turn conformations upon binding to lipid vesicles. Differential scanning calorimetry experiments with 1,2-dipalmitoleoyl-sn-glycero-3-phosphatidylethanolamine bilayers indicated the induction of positive curvature strain and repression of the fluid lamellar to inverted hexagonal phase transition by MSI-367. Results of isothermal titration calorimetry (ITC) experiments suggested the possibility of formation of specific lipid-peptide complexes leading to aggregation. (2)H nuclear magnetic resonance (NMR) of deuterated 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphatidylcholine (POPC) multilamellar vesicles confirmed the limited effect of the membrane-embedded peptide at the lipid-water interface. (31)P NMR data indicated changes in the lipid headgroup orientation of POPC, 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphatidylglycerol, and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphatidylethanolamine lipid bilayers upon peptide binding. Membrane-embedded and membrane-inserted states of the peptide were observed via sum frequency generation vibrational spectroscopy. Circular dichroism, ITC, and (31)P NMR data for Escherichia coli lipids agree with the hypothesis that strong electrostatic lipid-peptide interactions embrace the peptide at the lipid-water interface and provide the basis for bacterial cell selectivity.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Escherichia coli/drug effects , Lipid Bilayers/chemistry , Lipids/chemistry , Water/chemistry , Animals , Calorimetry , Calorimetry, Differential Scanning , Circular Dichroism , Erythrocytes/drug effects , Magnetic Resonance Spectroscopy , Phosphatidylethanolamines/chemistry , Sheep
11.
J Microbiol Biotechnol ; 20(4): 727-31, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20467245

ABSTRACT

The gene APE0743 encoding the superoxide dismutase (ApSOD) of a hyperthermophilic archaeon Aeropyrum pernix K1 was cloned and over-expressed as a GST fusion protein at a high level in Escherichia coli. The expressed protein was simply purified by the process of glutathione affinity chromatography and thrombin treatment. The ApSOD was a homodimer of 25 kDa subunits and a cambialistic SOD which was active with either Fe(II) or Mn(II) as a cofactor. The ApSOD was highly stable against high temperature. This thermostable ApSOD is expected to be applicable as a useful biocatalyst for medicine and bio-industrial processes.


Subject(s)
Aeropyrum/enzymology , Industrial Microbiology/methods , Superoxide Dismutase/biosynthesis , Aeropyrum/genetics , Amino Acid Sequence , Base Sequence , DNA, Archaeal/chemistry , DNA, Archaeal/genetics , Enzyme Activation , Escherichia coli/enzymology , Escherichia coli/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sequence Alignment , Superoxide Dismutase/genetics , Superoxide Dismutase/isolation & purification
12.
Biochim Biophys Acta ; 1798(2): 223-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19716800

ABSTRACT

While high-resolution 3D structures reveal the locations of all atoms in a molecule, it is the dynamics that correlates the structure with the function of a biological molecule. The complete characterization of dynamics of a membrane protein is in general complex. In this study, we report the influence of dynamics on the channel-forming function of pardaxin using chemical shifts and dipolar couplings measured from 2D broadband-PISEMA experiments on mechanically aligned phospholipids bilayers. Pardaxin is a 33-residue antimicrobial peptide originally isolated from the Red Sea Moses sole, Pardachirus marmoratus, which functions via either a carpet-type or barrel-stave mechanism depending on the membrane composition. Our results reveal that the presence of cholesterol significantly reduces the backbone motion and the tilt angle of the C-terminal amphipathic helix of pardaxin. In addition, a correlation between the dynamics-induced heterogeneity in the tilt of the C-terminal helix and the membrane disrupting activity of pardaxin by the barrel-stave mechanism is established. This correlation is in excellent agreement with the absence of hemolytic activity for the derivatives of pardaxin. These results explain the role of cholesterol in the selectivity of the broad-spectrum of antimicrobial activities of pardaxin.


Subject(s)
Anti-Infective Agents/chemistry , Cholesterol/chemistry , Fish Proteins/chemistry , Fish Venoms/chemistry , Lipid Bilayers/chemistry , Phospholipids/chemistry , Animals , Fishes , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Structure-Activity Relationship
13.
J Am Chem Soc ; 131(12): 4490-8, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19256547

ABSTRACT

Curcumin is the active ingredient of turmeric powder, a natural spice used for generations in traditional medicines. Curcumin's broad spectrum of antioxidant, anticarcinogenic, antimutagenic, and anti-inflammatory properties makes it particularly interesting for the development of pharmaceutical compounds. Because of curcumin's various effects on the function of numerous unrelated membrane proteins, it has been suggested that it affects the properties of the bilayer itself. However, a detailed atomic-level study of the interaction of curcumin with membranes has not been attempted. A combination of solid-state NMR and differential scanning calorimetry experiments shows curcumin has a strong effect on membrane structure at low concentrations. Curcumin inserts deep into the membrane in a transbilayer orientation, anchored by hydrogen bonding to the phosphate group of lipids in a manner analogous to cholesterol. Like cholesterol, curcumin induces segmental ordering in the membrane. Analysis of the concentration dependence of the order parameter profile derived from NMR results suggests curcumin forms higher order oligomeric structures in the membrane that span and likely thin the bilayer. Curcumin promotes the formation of the highly curved inverted hexagonal phase, which may influence exocytotic and membrane fusion processes within the cell. The experiments outlined here show promise for understanding the action of other drugs such as capsaicin in which drug-induced alterations of membrane structure have strong pharmacological effects.


Subject(s)
Antioxidants/chemistry , Antioxidants/pharmacology , Cell Membrane/drug effects , Curcumin/chemistry , Curcumin/pharmacology , Magnetic Resonance Spectroscopy/methods , Calorimetry, Differential Scanning , Cellular Structures/metabolism , Cholesterol/chemistry , Curcuma/metabolism , Lipids/chemistry , Membrane Proteins/chemistry , Membranes, Artificial , Micelles , Molecular Structure , Plant Extracts/metabolism
14.
Biochim Biophys Acta ; 1788(3): 686-95, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19071084

ABSTRACT

The second transmembrane (TM2) domain of GABA(A) receptor forms the inner-lining surface of chloride ion-channel and plays important roles in the function of the receptor protein. In this study, we report the first structure of TM2 in lipid bilayers determined using solid-state NMR and MD simulations. The interatomic (13)C-(15)N distances measured from REDOR magic angle spinning experiments on multilamellar vesicles, containing a TM2 peptide site specifically labeled with (13)C' and (15)N isotopes, were used to determine the secondary structure of the peptide. The (15)N chemical shift and (1)H-(15)N dipolar coupling parameters measured from PISEMA experiments on mechanically aligned phospholipid bilayers, containing a TM2 peptide site specifically labeled with (15)N isotopes, under static conditions were used to determine the membrane orientation of the peptide. Our results reveal that the TM2 peptide forms an alpha helical conformation with a tilted transmembrane orientation, which is unstable as a monomer but stable as pentameric oligomers as indicated by MD simulations. Even though the peptide consists of a number of hydrophilic residues, the transmembrane folding of the peptide is stabilized by intermolecular hydrogen bondings between the side chains of Ser and Thr residues as revealed by MD simulations. The results also suggest that peptide-peptide interactions in the tilted transmembrane orientation overcome the hydrophobic mismatch between the peptide and bilayer thickness.


Subject(s)
Receptors, GABA-A/physiology , Chloride Channels/chemistry , Chloride Channels/metabolism , Computer Simulation , Hydrogen Bonding , Lipid Bilayers , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Structure, Secondary , Receptors, GABA-A/chemistry
15.
Langmuir ; 24(23): 13598-604, 2008 Dec 02.
Article in English | MEDLINE | ID: mdl-18991419

ABSTRACT

Lipid-water interaction plays an important role in the properties of lipid bilayers, cryoprotectants, and membrane-associated peptides and proteins. The temperature at which water bound to lipid bilayers freezes is lower than that of free water. Here, we report a solid-state NMR investigation on the freezing point depression of water in phospholipid bilayers in the presence and absence of cholesterol. Deuterium NMR spectra at different temperatures ranging from -75 to + 10 degrees C were obtained from fully (2)H2O-hydrated POPC (1-palmitoyl-2-oleoylphosphatidylcholine) multilamellar vesicles (MLVs), prepared with and without cholesterol, to determine the freezing temperature of water and the effect of cholesterol on the freezing temperature of water in POPC bilayers. Our 2H NMR experiments reveal the motional behavior of unfrozen water molecules in POPC bilayers even at temperatures significantly below 0 degrees C and show that the presence of cholesterol further lowered the freezing temperature of water in POPC bilayers. These results suggest that in the presence of cholesterol the fluidity and dynamics of lipid bilayers can be retained even at very low temperatures as exist in the liquid crystalline phase of the lipid. Therefore, bilayer samples prepared with a cryoprotectant like cholesterol should enable the performance of multidimensional solid-state NMR experiments to investigate the structure, dynamics, and topology of membrane proteins at a very low temperature with enhanced sample stability and possibly a better sensitivity. Phosphorus-31 NMR data suggest that lipid bilayers can be aligned at low temperatures, while 15N NMR experiments demonstrate that such aligned samples can be used to enhance the signal-to-noise ratio of is 15N chemical shift spectra of a 37-residue human antimicrobial peptide, LL-37.


Subject(s)
Lipid Bilayers/chemistry , Magnetic Resonance Spectroscopy/methods , Phospholipids/chemistry , Transition Temperature , Water/chemistry , Surface Properties
16.
J Am Chem Soc ; 130(33): 11023-9, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18646853

ABSTRACT

Characterization of the oligomerization of membrane-associated peptides is important to understand the folding and function of biomolecules like antimicrobial peptides, fusion peptides, amyloid peptides, toxins, and ion channels. However, this has been considered to be very difficult, because the amphipathic properties of the constituents of the cell membrane pose tremendous challenges to most commonly used biophysical techniques. In this study, we present the application of a simple (14)N solid-state NMR spectroscopy of aligned model membranes containing a phosphatidyl choline lipid to investigate the oligomerization of membrane-associated peptides. Since the near-symmetric nature of the choline headgroup of a phosphocholine lipid considerably reduces the (14)N quadrupole coupling, there are significant practical advantages in using (14)N solid-state NMR experiments to probe the interaction of peptide or protein with the surface of model membranes. Experimental results for several membrane-associated peptides are presented in this paper. Our results suggest that the experimentally measured (14)N quadrupole splitting of the lipid depends on the peptide-induced changes in the electrostatic potential of the lipid bilayer surface and therefore on the nature of the peptide, peptide-membrane interaction, and peptide-peptide interaction. It is inferred that the membrane orientation and oligomerization of the membrane-associated peptides can be measured using (14)N solid-state NMR spectroscopy.


Subject(s)
Lipid Bilayers/chemistry , Lipids/chemistry , Magnetic Resonance Spectroscopy/methods , Nitrogen/chemistry , Peptides/chemistry , Phospholipids/chemistry , Magnetic Resonance Spectroscopy/standards , Membranes, Artificial , Molecular Structure , Phosphorus/chemistry , Reference Standards
17.
Biochemistry ; 46(4): 965-75, 2007 Jan 30.
Article in English | MEDLINE | ID: mdl-17240980

ABSTRACT

Cell-signaling peptides have been extensively used to transport functional molecules across the plasma membrane into living cells. These peptides consist of a hydrophobic sequence and a cationic nuclear localization sequence (NLS). It has been assumed that the hydrophobic region penetrates the hydrophobic lipid bilayer and delivers the NLS inside the cell. To better understand the transport mechanism of these peptides, in this study, we investigated the structure, orientation, tilt of the peptide relative to the bilayer normal, and the membrane interaction of two cell-signaling peptides, SA and SKP. Results from CD and solid-state NMR experiments combined with molecular dynamics simulations suggest that the hydrophobic region is helical and has a transmembrane orientation with the helical axis tilted away from the bilayer normal. The influence of the hydrophobic mismatch, between the hydrophobic length of the peptide and the hydrophobic thickness of the bilayer, on the tilt angle of the peptides was investigated using thicker POPC and thinner DMPC bilayers. NMR experiments showed that the hydrophobic domain of each peptide has a tilt angle of 15 +/- 3 degrees in POPC, whereas in DMPC, 25 +/- 3 degree and 30 +/- 3 degree tilts were observed for SA and SKP peptides, respectively. These results are in good agreement with molecular dynamics simulations, which predict a tilt angle of 13.3 degrees (SA in POPC), 16.4 degrees (SKP in POPC), 22.3 degrees (SA in DMPC), and 31.7 degrees (SKP in DMPC). These results and simulations on the hydrophobic fragment of SA or SKP suggest that the tilt of helices increases with a decrease in bilayer thickness without changing the phase, order, and structure of the lipid bilayers.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Amino Acid Sequence , Animals , Circular Dichroism , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Lipid Bilayers/chemistry , Macromolecular Substances , Molecular Sequence Data , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Thermodynamics
18.
Biophys J ; 91(1): 206-16, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16603496

ABSTRACT

The mechanism of membrane interaction of two amphipathic antimicrobial peptides, MSI-78 and MSI-594, derived from magainin-2 and melittin, is presented. Both the peptides show excellent antimicrobial activity. The 8-anilinonaphthalene-1-sulfonic acid uptake experiment using Escherichia coli cells suggests that the outer membrane permeabilization is mainly due to electrostatic interactions. The interaction of MSI-78 and MSI-594 with lipid membranes was studied using 31P and 2H solid-state NMR, circular dichroism, and differential scanning calorimetry techniques. The binding of MSI-78 and MSI-594 to the lipid membrane is associated with a random coil to alpha-helix structural transition. MSI-78 and MSI-594 also induce the release of entrapped dye from POPC/POPG (3:1) vesicles. Measurement of the phase-transition temperature of peptide-DiPoPE dispersions shows that both MSI-78 and MSI-594 repress the lamellar-to-inverted hexagonal phase transition by inducing positive curvature strain. 15N NMR data suggest that both the peptides are oriented nearly perpendicular to the bilayer normal, which infers that the peptides most likely do not function via a barrel-stave mechanism of membrane-disruption. Data obtained from 31P NMR measurements using peptide-incorporated POPC and POPG oriented lamellar bilayers show a disorder in the orientation of lipids up to a peptide/lipid ratio of 1:20, and the formation of nonbilayer structures at peptide/lipid ratio>1:8. 2H-NMR experiments with selectively deuterated lipids reveal peptide-induced disorder in the methylene units of the lipid acyl chains. These results are discussed in light of lipid-peptide interactions leading to the disruption of membrane via either a carpet or a toroidal-type mechanism.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Cell Membrane/chemistry , Escherichia coli/chemistry , Magnetic Resonance Spectroscopy , Membrane Fluidity , Peptides/chemistry , Cell Membrane Permeability , Magainins , Melitten/chemistry , Molecular Conformation , Xenopus Proteins/chemistry
19.
Biochim Biophys Acta ; 1758(2): 154-63, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16579960

ABSTRACT

A 15-residue peptide dimer G15 derived from the cell lytic protein granulysin has been shown to exert potent activity against microbes, including E. coli, but not against human Jurkat cells [Z. Wang, E. Choice, A. Kaspar, D. Hanson, S. Okada, S.C. Lyu, A.M. Krensky, C. Clayberger, Bactericidal and tumoricidal activities of synthetic peptides derived from granulysin. J. Immunol. 165 (2000) 1486-1490]. We investigated the target membrane selectivity of G15 using fluorescence, circular dichroism and 31P NMR methods. The ANS uptake assay shows that the extent of E. coli outer membrane disruption depends on G15 concentration. 31P NMR spectra obtained from E. coli total lipid bilayers incorporated with G15 show disruption of lipid bilayers. Fluorescence binding studies on the interaction of G15 with synthetic liposomes formed of E. coli lipids suggest a tight binding of the peptide at the membrane interface. The peptide also binds to negatively charged POPC/POPG (3:1) lipid vesicles but fails to insert deep into the membrane interior. These results are supported by the peptide-induced changes in the measured isotropic chemical shift and T1 values of POPG in 3:1 POPC:POPG multilamellar vesicles while neither a non-lamellar phase nor a fragmentation of bilayers was observed from NMR studies. The circular dichroism studies reveal that the peptide exists as a random coil in solution but folds into a less ordered conformation upon binding to POPC/POPG (3:1) vesicles. However, G15 does not bind to lipid vesicles made of POPC/POPG/Chl (9:1:1) mixture, mimicking tumor cell membrane. These results explain the susceptibility of E. coli and the resistance of human Jurkat cells to G15, and may have implications in designing membrane-selective therapeutic agents.


Subject(s)
Anti-Infective Agents/isolation & purification , Anti-Infective Agents/pharmacology , Antigens, Differentiation, T-Lymphocyte/chemistry , Amino Acid Sequence , Anti-Infective Agents/chemistry , Antigens, Differentiation, T-Lymphocyte/genetics , Cell Membrane/chemistry , Cell Membrane/drug effects , Circular Dichroism , Escherichia coli/chemistry , Escherichia coli/drug effects , Humans , In Vitro Techniques , Jurkat Cells , Lipid Bilayers/chemistry , Liposomes , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Fragments/pharmacology , Spectrometry, Fluorescence
20.
Biophys J ; 89(6): 4043-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16183881

ABSTRACT

The interaction of an antimicrobial peptide, MSI-78, with phospholipid bilayers has been investigated using atomic force microscopy, circular dichroism, and nuclear magnetic resonance (NMR). Binding of amphipathic peptide helices with their helical axis parallel to the membrane surface leads to membrane thinning. Atomic force microscopy of supported 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) bilayers in the presence of MSI-78 provides images of the membrane thinning process at a high spatial resolution. This data reveals that the membrane thickness is not reduced uniformly over the entire bilayer area. Instead, peptide binding leads to the formation of distinct domains where the bilayer thickness is reduced by 1.1 +/- 0.2 nm. The data is interpreted using a previously published geometric model for the structure of the peptide-lipid domains. In this model, the peptides reside at the hydrophilic-hydrophobic boundary in the lipid headgroup region, which leads to an increased distance between lipid headgroups. This picture is consistent with concentration-dependent 31P and 2H NMR spectra of MSI-78 in mechanically aligned DMPC bilayers. Furthermore, 2H NMR experiments on DMPC-d54 multilamellar vesicles indicate that the acyl chains of DMPC are highly disordered in the presence of the peptide as is to be expected for the proposed structure of the peptide-lipid assembly.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Lipid Bilayers/chemistry , Liposomes/chemistry , Membrane Fluidity , Membrane Microdomains/chemistry , Membrane Microdomains/ultrastructure , Microscopy, Atomic Force/methods , Molecular Conformation , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...