Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nutr Neurosci ; 22(12): 840-849, 2019 Dec.
Article in English | MEDLINE | ID: mdl-29595091

ABSTRACT

Objectives: This study evaluated the bioactive composition of tempeh products and examined the effects of tempeh on BV-2 microglial cell cytotoxicity, neurotrophic effects, and expression of inflammatory genes.Methods: Tempeh products included soybean fermented by Rhizopus, soybean fermented through cocultivation with Rhizopus and Lactobacillus, and red bean fermented through cocultivation with Rhizopus and Lactobacillus (RT-C). We analyzed the bioactive contents of tempeh extracts and evaluated the effects of tempeh water extract on lipopolysaccharide (LPS)-treated BV-2 cells.Results: The results showed that RT-C water extract had the highest concentrations of γ-aminobutyric acid (GABA) and anthocyanin. The tempeh water extracts, especially RT-C, reduced the formation of LPS-induced reactive oxygen species, downregulated the levels of nitric oxide synthase and phospho-cyclic-AMP response element-binding protein, and upregulated the expression of brain-derived neurotrophic factor (BDNF).Discussion: Our data demonstrate that RT-C has the highest concentrations of GABA and anthocyanin, more effectively reduces oxidative stress and inflammation, and increases the expression of BDNF in LPS-induced BV-2 cells.


Subject(s)
Lipopolysaccharides/pharmacology , Microglia/drug effects , Neuroprotective Agents/administration & dosage , Plant Extracts/pharmacology , Soy Foods , Animals , Anthocyanins/analysis , Anthocyanins/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Brain-Derived Neurotrophic Factor/analysis , Cell Line , Cyclic AMP Response Element-Binding Protein/analysis , Fermentation , Lactobacillus/metabolism , Mice , Nitric Oxide Synthase Type II/analysis , Plant Extracts/chemistry , Rhizopus/metabolism , Glycine max , gamma-Aminobutyric Acid/analysis , gamma-Aminobutyric Acid/pharmacology
2.
J Biol Chem ; 289(29): 20170-81, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24898246

ABSTRACT

Cobra cardiotoxins (CTX) are a family of three-fingered basic polypeptides known to interact with diverse targets such as heparan sulfates, sulfatides, and integrins on cell surfaces. After CTX bind to the membrane surface, they are internalized to intracellular space and exert their cytotoxicity via an unknown mechanism. By the combined in vitro kinetic binding, three-dimensional x-ray structure determination, and cell biology studies on the naturally abundant CTX homologues from the Taiwanese cobra, we showed that slight variations on the spatial distribution of positively charged or hydrophobic domains among CTX A2, A3, and A4 could lead to significant changes in their endocytotic pathways and action mechanisms via distinct sulfated glycoconjugate-mediated processes. The intracellular locations of these structurally similar CTX after internalization are shown to vary between the mitochondria and lysosomes via either dynamin2-dependent or -independent processes with distinct membrane cholesterol sensitivity. Evidence is presented to suggest that the shifting between the sulfated glycoconjugates as distinct targets of CTX A2, A3, and A4 might play roles in the co-evolutionary arms race between venomous snake toxins to cope with different membrane repair mechanisms at the cellular levels. The sensitivity of endocytotic routes to the spatial distribution of positively charged or hydrophobic domains may provide an explanation for the diverse endocytosis pathways of other cell-penetrating basic polypeptides.


Subject(s)
Cobra Cardiotoxin Proteins/chemistry , Cobra Cardiotoxin Proteins/pharmacokinetics , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cell Line , Cell Membrane/metabolism , Cobra Cardiotoxin Proteins/genetics , Cricetinae , Cricetulus , Crystallography, X-Ray , Elapidae/genetics , Elapidae/metabolism , Endocytosis , Evolution, Molecular , Glycoconjugates/chemistry , Glycoconjugates/metabolism , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Rats , Static Electricity , Structural Homology, Protein
3.
Biochim Biophys Acta ; 1818(5): 1378-85, 2012 May.
Article in English | MEDLINE | ID: mdl-22387431

ABSTRACT

Cobra CTX A3, the major cardiotoxin (CTX) from Naja atra, is a cytotoxic, basic ß-sheet polypeptide that is known to induce a transient membrane leakage of cardiomyocytes through a sulfatide-dependent CTX membrane pore formation and internalization mechanism. The molecular specificity of CTX A3-sulfatide interaction at atomic levels has also been shown by both nuclear magnetic resonance (NMR) and X-ray diffraction techniques to reveal a role of CTX-induced sulfatide conformational changes for CTX A3 binding and dimer formation. In this study, we investigate the role of sulfatide lipid domains in CTX pore formation by various biophysical methods, including fluorescence imaging and atomic force microscopy, and suggest an important role of liquid-disordered (ld) and solid-ordered (so) phase boundary in lipid domains to facilitate the process. Fluorescence spectroscopic studies on the kinetics of membrane leakage and CTX oligomerization further reveal that, although most CTXs can oligomerize on membranes, only a small fraction of CTXs oligomerizations form leakage pores. We therefore suggest that CTX binding at the boundary between the so and so/ld phase coexistence sulfatide lipid domains could form effective pores to significantly enhance the CTX-induced membrane leakage of sulfatide-containing phosphatidylcholine vesicles. The model is consistent with our earlier observations that CTX may penetrate and lyse the bilayers into small aggregates at a lipid/protein molar ratio of about 20 in the ripple P(ß)' phase of phosphatidylcholine bilayers and suggest a novel mechanism for the synergistic action of cobra secretary phospholipase A2 and CTXs.


Subject(s)
Cell Membrane/chemistry , Cobra Cardiotoxin Proteins/chemistry , Elapidae , Lipid Bilayers/chemistry , Membranes, Artificial , Protein Multimerization , Animals , Cell Membrane/metabolism , Cobra Cardiotoxin Proteins/metabolism , Lipid Bilayers/metabolism , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/metabolism , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Protein Structure, Secondary , X-Ray Diffraction
4.
Biochem J ; 433(1): 127-38, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20964630

ABSTRACT

HDGF (hepatoma-derived growth factor) stimulates cell proliferation by functioning on both sides of the plasma membrane as a ligand for membrane receptor binding to trigger cell signalling and as a stimulator for DNA synthesis in the nucleus. Although HDGF was initially identified as a secretory heparin-binding protein, the biological significance of its heparin-binding ability remains to be determined. In the present study we demonstrate that cells devoid of surface HS (heparan sulfate) were unable to internalize HDGF, HATH (N-terminal domain of HDGF consisting of amino acid residues 1-100, including the PWWP motif) and HATH(K96A) (single-site mutant form of HATH devoid of receptor binding activity), suggesting that the binding of HATH to surface HS is important for HDGF internalization. We further demonstrate that both HATH and HATH(K96A) could be internalized through macropinocytosis after binding to the cell surface HS. Interestingly, HS-mediated HATH(K96A) internalization is found to exhibit an inhibitory effect on cell migration and proliferation in contrast with that observed for HATH action on NIH 3T3 cells, suggesting that HDGF exploits the innate properties of both cell surface HS and membrane receptor via the HATH domain to affect related cell signalling processes. The present study indicates that MAPK (mitogen-activated protein kinase) signalling pathways could be affected by the HS-mediated HATH internalization to regulate cell migration in NIH 3T3 fibroblasts, as judged from the differential effect of HATH and HATH(K96A) treatment on the expression level of matrix metalloproteases.


Subject(s)
Cell Movement , Fibroblasts/physiology , Heparitin Sulfate/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Pinocytosis , Signal Transduction/physiology , 3T3 Cells , Animals , Cell Membrane/chemistry , Cell Proliferation , Fibroblasts/cytology , Matrix Metalloproteinases/biosynthesis , Mice , Protein Structure, Tertiary
5.
J Struct Biol ; 169(3): 294-303, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19932752

ABSTRACT

The structures of snake venom metalloproteases (SVMPs) are proposed to be useful models to understand the structural and functional relationship of ADAM (a disintegrin and metalloprotease) which are membrane-anchored proteins involved in multiple human diseases. We have purified, sequenced and determined the structures of two new P-III SVMPs - atragin and kaouthiagin-like (K-like) from Naja atra. Atragin exhibits a known C-shaped topology, whereas K-like adopts an I-shaped conformation because of the distinct disulfide pattern in the disintegrin-like (D) domain. K-like exhibits an enzymatic specificity toward pro-TNFalpha with less inhibition of cell migration, but atragin shows the opposite effect. The specificity of the enzymatic activity is indicated to be dominated mainly by the local structures of SVMP in the metalloprotease (M) domain, whereas the hyper-variable region (HVR) in the cysteine-rich (C) domain is involved in a cell-migration activity. We demonstrate also a pH-dependent enzymatic activity of atragin that we correlate with the structural dynamics of a Zn(2+)-binding motif and the Met-turn based on the structures determined with a pH-jump method. The structural variations between the C- and I-shapes highlight the disulfide bond patterns in the D domain of the ADAM/adamalysin/reprolysins family proteins.


Subject(s)
Elapid Venoms/enzymology , Metalloproteases/chemistry , Metalloproteases/metabolism , Snakes/metabolism , Amino Acid Motifs/genetics , Amino Acid Motifs/physiology , Animals , Crystallography, X-Ray , Hydrogen-Ion Concentration , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Metalloproteases/genetics , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Spectrometry, Fluorescence , Substrate Specificity/genetics , Substrate Specificity/physiology
6.
Adv Exp Med Biol ; 677: 143-9, 2010.
Article in English | MEDLINE | ID: mdl-20687487

ABSTRACT

Cobra venom contains cardiotoxins (CTXs) that induce tissue necrosis and systolic heart arrest in bitten victims. CTX-induced membrane pore formation is one of the major mechanisms responsible for the venom's designated cytotoxicity. This chapter examines how glycoconjugates such as heparan sulfates (HS) and glycosphingolipids, located respectively in the extracellular matrix and lipid bilayers of the cell membranes, facilitate CTX pore formation. Evidences for HS-facilitated cell surface retention and glycosphingolipid-facilitated membrane bilayer insertion of CTX are reviewed. We suggest that similar physical steps could play a role in the mediation of other pore forming toxins (PFT). The membrane pores formed by PFT are expected to have limited lifetime on biological cell surface as a result of membrane dynamics during endocytosis and/or rearrangement of lipid rafts.


Subject(s)
Cobra Cardiotoxin Proteins/chemistry , Elapidae , Heparitin Sulfate/chemistry , Lipid Bilayers/chemistry , Membrane Microdomains/chemistry , Pore Forming Cytotoxic Proteins/chemistry , Sphingolipids/chemistry , Animals , Cobra Cardiotoxin Proteins/metabolism , Heart Arrest/metabolism , Heparitin Sulfate/metabolism , Humans , Lipid Bilayers/metabolism , Membrane Microdomains/metabolism , Necrosis/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Sphingolipids/metabolism
7.
Polymers (Basel) ; 12(8)2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32722124

ABSTRACT

Chitin, a polymer of N-acetyl-d-glucosamine (GlcNAc), can be degraded by chitinase, which is produced by higher plants, vertebrates, and bacteria. Chitinases are characterized by the ability to hydrolyze the beta-1,4-linkages in the chitin chain by either an endolytic or an exolytic mechanism. Chitinase 1198 is a novel endochitinase from the genome sequence of Chitinibacter tainanensis CT01. Herein, we report the findings of molecular simulations and bioassays for chitinase 1198. Our experimental results suggest that chitinase 1198 can recognize the nonreducing end of chitin and cleave the second or third glycosidic linkage from the nonreducing end of chitin oligomers. Furthermore, our simulations results revealed that chitinase 1198 is more likely to bind chitin oligomers with the main hydrogen bonds of the Asp440, the second GlcNAc unit of chitin oligomers, and degrade chitin oligomers to (GlcNAc)2 molecules. Moreover, chitinase 1198 is less likely to bind chitin oligomers with the main hydrogen bonds of the Asp440, the third GlcNAc unit of chitin oligomers, and degrade chitin oligomers to (GlcNAc)3 molecules. Lastly, chitinase 1198 can bind (GlcNAc)3 molecules with the main hydrogen bonds of the Asp440, the second GlcNAc of the (GlcNAc)3 molecules, and degrade chitin oligomers to GlcNAc and (GlcNAc)2 molecules.

8.
Toxicon ; 118: 1-12, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27090013

ABSTRACT

We have previously identified two new P-III type ADAM-like snake venom metalloproteinases (SVMPs), i.e., atragin and kaouthiagin-like, from Taiwan cobra venom and determined their 3D structures with a distinct C- and I-shaped metalloproteinase/disintegrin-like/cysteine-rich (MDC) modular architecture. Herein, we investigated their functional targets to elucidate the role of cobra SVMPs in perturbing wound healing in snakebite victims. We showed that the non-RGD (Arg-Gly-Asp) C-shaped SVMP atragin binds about ten-fold stronger than the RGD-containing I-shaped SVMP kaouthiagin-like to αvß3 integrin in the surface-immobilized form. Atragin binds to αvß3 integrin through a novel interaction mode involving distal M and C domains via the RRN sequence motif in the hyper variable loop. In a cell adhesion assay, the adhesion of fibroblasts to atragin was mediated by αvß3 integrin. Furthermore, atragin inhibited wound healing and suppressed cell migration in a αvß3 integrin-dependent manner. These results, together with our previous demonstration of non-cytotoxic cobra CTX A5 in targeting αvß3 integrin, suggest that cobra venom consists of several non-RGD toxins with integrin-binding specificity that could perturb wound healing in snakebite victims.


Subject(s)
ADAM Proteins/metabolism , Elapid Venoms/enzymology , Integrin alphaVbeta3/metabolism , Reptilian Proteins/metabolism , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAM Proteins/isolation & purification , Amino Acid Motifs , Animals , Becaplermin , Cell Adhesion , Cell Movement , Elapidae , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/genetics , Immobilized Proteins/metabolism , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/genetics , Ligands , Mice , Molecular Docking Simulation , NIH 3T3 Cells , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-sis/chemistry , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reptilian Proteins/chemistry , Reptilian Proteins/genetics , Reptilian Proteins/isolation & purification , Solubility , Surface Plasmon Resonance , Taiwan
9.
Mol Oncol ; 8(8): 1575-87, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25091737

ABSTRACT

Treatment with individual anti-androgens is associated with the development of hot-spot mutations in the androgen receptor (AR). Here, we found that anti-androgens-mt-ARs have similar binary structure to the 5α-dihydrotestosterone-wt-AR. Phage display revealed that these ARs bound to similar peptides, including BUD31, containing an Fxx(F/H/L/W/Y)Y motif cluster with Tyr in the +5 position. Structural analyses of the AR-LBD-BUD31 complex revealed formation of an extra hydrogen bond between the Tyr+5 residue of the peptide and the AR. Functional studies showed that BUD31-related peptides suppressed AR transactivation, interrupted AR N-C interaction, and suppressed AR-mediated cell growth. Combination of peptide screening and X-ray structure analysis may serve as a new strategy for developing anti-ARs that simultaneously suppress both wt and mutated AR function.


Subject(s)
Peptides/chemistry , Peptides/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Crystallography, X-Ray , Humans , Immunoprecipitation , Male , Peptides/pharmacology , Protein Binding , Surface Plasmon Resonance
10.
Biochemistry ; 46(43): 12111-23, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17918958

ABSTRACT

The major cardiotoxin from Taiwan cobra (CTX A3) is a pore forming beta-sheet polypeptide that requires sulfatide (sulfogalactosylceramide, SGC) on the plasma membrane of cardiomyocytes for CTX-induced membrane leakage and cell internalization. Herein, we demonstrate by fluorescence spectroscopic studies that sulfatides induce CTX A3 oligomerization in sulfatide containing phosphatidylcholine (PC) vesicles to form transient pores with pore size and lifetime in the range of about 30 A and 10(-2) s, respectively. These values are consistent with the CTX A3-induced conductance and mean lifetime determined previously by using patch-clamp electrophysiological experiments on the plasma membrane of H9C2 cells. We also derived the peripheral binding structural model of CTX A3-sulfatide complex in sulfatide containing PC micelles by NMR and molecular docking method and compared with other CTX A3-sulfatide complex structure determined previously by X-ray in membrane-like environment. The NMR results indicate that sulfatide head group conformation changes from a bent shovel (-sc/ap) to an extended (sc/ap) conformation upon initial binding of CTX A3. An additional global reorientation of sulfatide molecule is also needed for CTX A3 dimer formation as inferred by the difference between the X-ray and NMR complex structure. Since the overall folding of CTX A3 molecules remained the same, sulfatide in phospholipid bilayer is proposed to play an active role by involving its local and global conformational changes to promote both the oligomerization and reorientation of CTX A3 molecule for its transient pore formation and cell internalization.


Subject(s)
Cardiotoxins/chemistry , Glycosphingolipids/metabolism , Animals , Cell Line , Elapidae , Glycosphingolipids/chemistry , Models, Molecular , Molecular Conformation , Nuclear Magnetic Resonance, Biomolecular , Spectrometry, Fluorescence
11.
J Biol Chem ; 281(12): 7937-45, 2006 Mar 24.
Article in English | MEDLINE | ID: mdl-16407244

ABSTRACT

Severe tissue necrosis with a retarded wound healing process is a major symptom of a cobra snakebite. Cardiotoxins (CTXs) are major components of cobra venoms that belong to the Ly-6 protein family and are implicated in tissue damage. The interaction of the major CTX from Taiwan cobra, i.e. CTX A3, with sulfatides in the cell membrane has recently been shown to induce pore formation and cell internalization and to be responsible for cytotoxicity in cardiomyocytes (Wang, C.-H., Liu, J.-H., Lee, S.-C., Hsiao, C.-D., and Wu, W.-g. (2006) J. Biol. Chem. 281, 656-667). We show here that one of the non-cytotoxic CTXs, i.e. CTX A5 or cardiotoxin-like basic polypeptide, from Taiwan cobra specifically bound to alpha(v)beta3 integrin and inhibited bone resorption activity. We found that both membrane-bound and recombinant soluble alpha(v)beta3 integrins bound specifically to CTX A5 in a dose-dependent manner. Surface plasmon resonance analysis showed that human soluble alpha(v)beta3 bound to CTX A5 with an apparent affinity of approximately 0.3 microM. Calf pulmonary artery endothelial cells, which constitutively express alpha(v)beta3, showed a CTX A5 binding profile similar to that of membrane-bound and soluble alpha(v)beta3 integrins, suggesting that endothelial cells are a potential target for CTX action. We tested whether CTX A5 inhibits osteoclast differentiation and bone resorption, a process known to be involved in alpha(v)beta3 binding and inhibited by RGD-containing peptides. We demonstrate that CTX A5 inhibited both activities at a micromolar range by binding to murine alpha(v)beta3 integrin in osteoclasts and that CTX A5 co-localized with beta3 integrin. Finally, after comparing the integrin binding affinity among CTX homologs, we propose that the amino acid residues near the two loops of CTX A5 are involved in integrin binding. These results identify CTX A5 as a non-RGD integrin-binding protein with therapeutic potential as an integrin antagonist.


Subject(s)
Bone Resorption , Cobra Cardiotoxin Proteins/chemistry , Integrin alphaVbeta3/chemistry , Oligopeptides/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Cattle , Cell Adhesion , Cell Differentiation , Cell Line , Cell Proliferation , Cobra Cardiotoxin Proteins/metabolism , Dose-Response Relationship, Drug , Elapidae , Endothelial Cells/metabolism , Glycoproteins/chemistry , Inflammation , Integrin alphaVbeta3/metabolism , Integrins/chemistry , Integrins/metabolism , Ligands , Mice , Microscopy, Electron, Scanning , Molecular Sequence Data , Necrosis , Neoplasms/metabolism , Osteoclasts/metabolism , Protein Binding , Protein Structure, Secondary , Pulmonary Artery/metabolism , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Structure-Activity Relationship , Time Factors
12.
Biochemistry ; 44(20): 7414-26, 2005 May 24.
Article in English | MEDLINE | ID: mdl-15895985

ABSTRACT

Natural homologues of cobra cardiotoxins (CTXs) were classified into two structural subclasses of group I and II based on the amino acid sequence and circular dichroism analysis, but the exact differences in their three-dimensional structures and biological significance remain elusive. We show by circular dichroism, NMR spectroscopic, and X-ray crystallographic analyses of a newly purified group I CTX A6 from eastern Taiwan cobra (Naja atra) venoms that its loop I conformation adopts a type VIa turn with a cis peptide bond located between two proline residues of PPxY. A similar "banana-twisted" conformation can be observed in other group I CTXs and also in cyclolinopeptide A and its analogues. By binding to the membrane environment, group I CTX undergoes a conformational change to adopt a more extended hydrophobic domain with beta-sheet twisting closer to the one adopted by group II CTX. This result resolves a discrepancy in the CTX structural difference reported previously between solution as well as crystal state and shows that, in addition to the hydrophobicity, the exact loop I conformation also plays an important role in CTX-membrane interaction. Potential protein targets of group I CTXs after cell internalization are also discussed on the basis of the determined loop I conformation.


Subject(s)
Cobra Cardiotoxin Proteins/chemistry , Cobra Cardiotoxin Proteins/classification , Elapid Venoms/chemistry , Elapid Venoms/classification , Proline/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Circular Dichroism , Cobra Cardiotoxin Proteins/isolation & purification , Crystallization , Crystallography, X-Ray , Elapid Venoms/isolation & purification , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylserines/chemistry , Protein Binding , Protein Conformation , Solutions , Taiwan
13.
Biochemistry ; 42(24): 7457-66, 2003 Jun 24.
Article in English | MEDLINE | ID: mdl-12809502

ABSTRACT

Cobra cardiotoxin, a cytotoxic beta-sheet basic polypeptide, is known to cause membrane leakage in many cells including human erythrocytes. Herein, we demonstrate that the major cobra cardiotoxin from Naja atra, CTX A3, can cause leakage of vesicle contents in phosphatidylglycerol (PG) and phosphatidylserine containing, but not in pure phosphatidylcholine (PC), membrane bilayers. By the combined polarized attenuated total reflection infrared spectroscopy and computer simulation studies, CTX A3 is shown to peripherally bind to both zwitterionic and anionic monolayers in a similar edgewise manner with a tilted angle of approximately 48 +/- 20 degrees between the beta-sheet plane of the CTX molecule and the normal of the membrane surface. The average surface area expansion induced by CTX A3 binding to the PG monolayer, however, is two times larger than that of the PC monolayer as determined by the Langmuir minitrough method. Interaction energy considerations of CTX A3 on neutral and negatively charged membrane surfaces suggests that the electrostatic interaction between anionic lipid and cationic CTXs plays a role in modulating the penetration depth of CTX molecules on the initial peripheral binding mode and reveals a pathway leading to the formation of an inserted mode in negatively charged membrane bilayers.


Subject(s)
Cobra Cardiotoxin Proteins/chemistry , Cobra Cardiotoxin Proteins/metabolism , Glycerophospholipids/chemistry , Glycerophospholipids/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Lipids/metabolism , Animals , Cobra Cardiotoxin Proteins/pharmacology , Computer Simulation , Membrane Lipids/chemistry , Membrane Potentials , Membranes, Artificial , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Pressure , Protein Binding/drug effects , Protein Structure, Secondary , Spectroscopy, Fourier Transform Infrared/methods , Static Electricity , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL