Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
3.
Anal Biochem ; 292(1): 102-6, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11319823

ABSTRACT

Hydrophobicity, a term used to describe a fundamental physicochemical property of local anesthetics, was in the past obtained by octanol/buffer partitioning. It has been suggested that the octanol method, despite its obvious advantages, also has some drawbacks. HPLC has become an attractive alternative for the measurement of hydrophobicity and has been applied to local anesthetics recently. However, the methods in current use for measuring the hydrophobicity of local anesthetics suffer from a number of limitations and remain obscure. This study introduces a new HPLC method for measuring the hydrophobicity of eight local anesthetics in current clinical use. Using a C(18) derivatized polystyrene-divinylbenzene stationary phase HPLC column, the log k'(w) values of local anesthetics were determined by measuring the capacity factor k'(i) in the process of chromatographic separation using a hydrophobic stationary phase and a hydrophilic mobile phase. A rapid reversed-phase HPLC method was developed to directly measure log k'(w) of eight local anesthetics. A high correlation between log k'(w) and hydrophobicity (log P(oct)) from the traditional shake-flask method was obtained for the local anesthetics, demonstrating the reliability of the method. The results reveal an improved method for measuring the hydrophobicity of the local anesthetic agents in the unionized form. This simple, sensitive and reproducible approach may serve as a valuable tool for describing the physicochemical properties of novel local anesthetics.


Subject(s)
Anesthetics, Local/chemistry , Chromatography, High Pressure Liquid/methods , Procaine/chemistry , Alkanes/chemistry , Silicon Dioxide/chemistry
4.
Biochemistry ; 40(16): 5075-80, 2001 Apr 24.
Article in English | MEDLINE | ID: mdl-11305924

ABSTRACT

Xenon and dichloromethane are inhalational anesthetic agents whose binding to myoglobin has been demonstrated by X-ray crystallography. We explore the thermodynamic significance of such binding using differential scanning calorimetry, circular dichroism spectroscopy, and hydrogen-tritium exchange measurements to study the effect of these agents on myoglobin folding stability. Though specific binding of these anesthetics might be expected to stabilize myoglobin against unfolding, dichloromethane actually destabilized myoglobin at all examined concentrations of this anesthetic (15, 40, and 200 mM). On the other hand, xenon (1 atm) stabilized myoglobin. Thus, dichloromethane and xenon have opposite effects on myoglobin stability despite localization in comparably folded X-ray crystallographic structures. These results suggest a need for solution measurements to complement crystallography if the consequences of weak binding to proteins are to be appreciated.


Subject(s)
Anesthetics, Inhalation/chemistry , Crystallography, X-Ray , Methylene Chloride/chemistry , Myoglobin/chemistry , Animals , Calorimetry, Differential Scanning , Circular Dichroism , Crystallography, X-Ray/methods , Horses , Hydrogen/chemistry , Protein Binding , Protein Folding , Recombinant Proteins/chemistry , Thermodynamics , Tritium/chemistry , Whales , Xenon/chemistry
5.
J Biol Chem ; 276(18): 14861-6, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11278529

ABSTRACT

The stability and equilibrium unfolding of a model three-helix bundle protein, alpha(3)-1, by guanidine hydrochloride (GdnHCl), hydrostatic pressure, and temperature have been investigated. The combined use of these denaturing agents allowed detection of two partially folded states of alpha(3)-1, as monitored by circular dichroism, intrinsic fluorescence emission, and fluorescence of the hydrophobic probe bis-ANS (4,4'-dianilino-1,1'-binaphthyl-5,5'-disulfonic acid). The overall free-energy change for complete unfolding of alpha(3)-1, determined from GdnHCl unfolding data, is +4.6 kcal/mol. The native state is stabilized by -1.4 kcal/mol relative to a partially folded pressure-denatured intermediate (I(1)). Cold denaturation at high pressure gives rise to a second partially (un)folded conformation (I(2)), suggesting a significant contribution of hydrophobic interactions to the stability of alpha(3)-1. The free energy of stabilization of the native-like state relative to I(2) is evaluated to be -2.5 kcal/mol. Bis-ANS binding to the pressure- and cold-denatured states indicates the existence of significant residual hydrophobic structure in the partially (un)folded states of alpha(3)-1. The demonstration of folding intermediates of alpha(3)-1 lends experimental support to a number of recent protein folding simulation studies of other three-helix bundle proteins that predicted the existence of such intermediates. The results are discussed in terms of the significance of de novo designed proteins for protein folding studies.


Subject(s)
Protein Folding , Amino Acid Sequence , Circular Dichroism , Cold Temperature , Fluorescence , Molecular Sequence Data , Pressure , Protein Conformation , Protein Denaturation , Thermodynamics
7.
Biophys J ; 78(2): 982-93, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10653811

ABSTRACT

The structural features of volatile anesthetic binding sites on proteins are being examined with the use of a defined model system consisting of a four-alpha-helix bundle scaffold with a hydrophobic core. Previous work has suggested that introducing a cavity into the hydrophobic core improves anesthetic binding affinity. The more polarizable methionine side chain was substituted for a leucine, in an attempt to enhance the dispersion forces between the ligand and the protein. The resulting bundle variant has an improved affinity (K(d) = 0.20 +/- 0.01 mM) for halothane binding, compared with the leucine-containing bundle (K(d) = 0.69 +/- 0.06 mM). Photoaffinity labeling with (14)C-halothane reveals preferential labeling of the W15 residue in both peptides, supporting the view that fluorescence quenching by bound anesthetic reports both the binding energetics and the location of the ligand in the hydrophobic core. The rates of amide hydrogen exchange were similar for the two bundles, suggesting that differences in binding affinity were not due to changes in protein stability. Binding of halothane to both four-alpha-helix bundle proteins stabilized the native folded conformations. Molecular dynamics simulations of the bundles illustrate the existence of the hydrophobic core, containing both W15 residues. These results suggest that in addition to packing defects, enhanced dispersion forces may be important in providing higher affinity anesthetic binding sites. Alternatively, the effect of the methionine substitution on halothane binding energetics may reflect either improved access to the binding site or allosteric optimization of the dimensions of the binding pocket. Finally, preferential stabilization of folded protein conformations may represent a fundamental mechanism of inhaled anesthetic action.


Subject(s)
Anesthetics, Inhalation/chemistry , Protein Structure, Secondary , Amino Acid Sequence , Binding Sites , Circular Dichroism , Computer Simulation , Fluorescence , Models, Molecular , Molecular Sequence Data , Peptides/chemical synthesis , Photoaffinity Labels/chemistry , Protein Binding , Protein Conformation , Protein Denaturation , Protein Folding , Protons , Sequence Analysis , Thermodynamics
9.
Mol Pharmacol ; 56(2): 414-8, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10419562

ABSTRACT

A loss of potency as one ascends a homologous series of compounds (cutoff effect) is often used to map the dimensions of binding sites on a protein target. The implicit assumption of steric hindrance is rarely confirmed with direct binding measurements, yet other mechanisms for cutoff exist. We studied the binding and effect of a series of n-alkanols up to hexadecanol (C16) on two model proteins, BSA and myoglobin (MGB), using hydrogen-tritium exchange and light scattering. BSA binds the n-alkanols specifically and, at 1 mM total concentration, is stabilized with increasing potency up to decanol (C10), where a loss in stabilizing potency occurs. Cutoff in stabilizing potency is concentration-dependent and occurs at progressively longer n-alkanols at progressively lower total n-alkanol concentrations. Light scattering measurements of n-alkanol/BSA solutions show a smooth decline in binding stoichiometry with increasing chain length until C14-16, where it levels off at approximately 2:1 (alkanol:BSA). MGB does not bind the n-alkanols specifically and is destabilized by them with increasing potency until C10, where a loss in destabilizing potency occurs. Like BSA, MGB demonstrates a concentration-dependent cutoff point for the n-alkanols. Derivation of the number of methylenes bound at K(D) and the free energy contribution per bound methylene showed that no discontinuity existed to explain cutoff, rendering steric hindrance unlikely. The data also allow an energetic explanation for the variance of the cutoff point in various reductionist systems. Finally, these results render cutoff an untenable approach for mapping binding site sterics in the absence of complementary binding measurements, and a poor discriminator of target relevance to general anesthesia.


Subject(s)
1-Propanol/chemistry , Fatty Alcohols/chemistry , Myoglobin/chemistry , Serum Albumin, Bovine/chemistry , Animals , Binding Sites , Cattle , Horses , Protein Binding , Structure-Activity Relationship
10.
Biophys Chem ; 79(2): 107-16, 1999 Jun 07.
Article in English | MEDLINE | ID: mdl-10389237

ABSTRACT

Partitioning of four modern inhalational anesthetics (halothane, isoflurane, enflurane, and sevoflurane) between the gas phase and nine organic solvents that model different amino acid side-chains and lipid membrane domains was performed in an effort to define which microenvironments present in proteins and lipid bilayers might be favored. Compared to a purely aliphatic environment (hexane), the presence of an aromatic-, alcohol-, thiol- or sulfide group on the solvent improved anesthetic partitioning, by factors of 1.3-5.2 for halothane, 1.7-5.6 for isoflurane, 1.7-7.6 for enflurane, and 1.5-7.3 for sevoflurane. The most favorable solvent for halothane partitioning was ethyl methyl sulfide, a model for methionine. Enflurane and isoflurane partitioned most extensively into methanol, a model for serine, and sevoflurane into ethanol, a model for threonine. Isoflurane also partitioned favorably into ethyl methyl sulfide. The results suggest that volatile general anesthetics interact better with partly polar groups, which are present on amino acids frequently found buried in the hydrophobic core of proteins, compared to purely aliphatic side-chains. Furthermore, if an anesthetic molecule was located in a saturated region of a phospholipid bilayer membrane, there would be an energetically favorable driving force for it to move into several higher dielectric microenvironments present on membrane proteins. The results provide evidence that proteins rather than lipids are the likely targets of volatile general anesthetics in biological membranes.


Subject(s)
Amino Acids/chemistry , Anesthetics, General/chemistry , Anesthetics, General/pharmacology , Models, Chemical , Animals , Binding Sites , Humans , Membrane Lipids/chemistry , Membrane Proteins/chemistry , Solvents , Thermodynamics
11.
Anesthesiology ; 90(1): 235-45, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9915333

ABSTRACT

BACKGROUND: Recent studies have demonstrated that volatile general anesthetic agents such as halothane and isoflurane may bind to discrete sites on protein targets. In the case of bovine serum albumin, the sites of halothane and chloroform binding have been identified as being located in the IB and IIA subdomains. This structural information provides a foundation for more detailed studies into the potential mechanisms of anesthetic action. METHODS: The effect of halothane and isoflurane and the nonimmobilizer 1,2-dichlorohexafluorocyclobutane on the mobility of the indole ring in the tryptophan residues of albumin was investigated using measurements of fluorescence anisotropy. Myoglobin served as a negative control. In addition, the effect of bound anesthetic agents on global protein stability was determined by thermal denaturation experiments using near-ultraviolet circular dichroism spectroscopy. RESULTS: The fluorescence anisotropy measurements showed that halothane and isoflurane decreased the mobility of the indole rings in a concentration-dependent manner. The calculated dissociation constants were 1.6+/-0.4 and 1.3+/-0.3 mM for isoflurane and halothane, respectively. In contrast, both agents failed to increase the fluorescence anisotropy of the tryptophan residues in myoglobin, compatible with lack of binding. The nonimmobilizer 1,2-dichlorohexafluorocyclobutane caused no change in the fluorescence anisotropy of albumin. Binding of the anesthetic agents stabilized the native folded form of albumin to thermal denaturation. Analysis of the thermal denaturation data yielded dissociation constant values of 0.98+/-0.10 mM for isoflurane and 1.0+/-0.1 mM for halothane. CONCLUSIONS: Attenuation of local side-chain dynamics and stabilization of folded protein conformations may represent fundamental modes of action of volatile general anesthetic agents. Because protein activity is crucially dependent on inherent flexibility, anesthetic-induced stabilization of certain protein conformations may explain how these important clinical agents change protein function.


Subject(s)
Anesthetics, Inhalation/chemistry , Serum Albumin, Bovine/chemistry , Algorithms , Anesthetics, Inhalation/metabolism , Animals , Cattle , Chlorofluorocarbons/chemistry , Circular Dichroism , Cyclobutanes/chemistry , Fluorescence Polarization , Halothane/chemistry , Halothane/metabolism , Humans , Isoflurane/chemistry , Isoflurane/metabolism , Myoglobin/chemistry , Myoglobin/metabolism , Protein Binding , Protein Conformation , Protein Denaturation , Serum Albumin, Bovine/metabolism , Temperature , Tryptophan/chemistry , Tryptophan/metabolism
12.
Biochemistry ; 37(5): 1421-9, 1998 Feb 03.
Article in English | MEDLINE | ID: mdl-9477971

ABSTRACT

The structural features of protein binding sites for volatile anesthetics are being explored using a defined model system consisting of a four-alpha-helix bundle scaffold with a hydrophobic core. Earlier work has demonstrated that a prototype hydrophobic core is capable of binding the volatile anesthetic halothane. Exploratory work on the design of an improved affinity anesthetic binding site is presented, based upon the introduction of a simple cavity into a prototype (alpha 2)2 four-alpha-helix bundle by replacing six core leucines with smaller alanines. The presence of such a cavity increases the affinity (Kd = 0.71 +/- 0.04 mM) of volatile anesthetic binding to the designed bundle core by a factor of 4.4 as compared to an analogous bundle core lacking such a cavity (Kd = 3.1 +/- 0.4 mM). This suggests that such packing defects present on natural proteins are likely to be occupied by volatile general anesthetics in vivo. Replacing six hydrophobic core leucine residues with alanines results in a destabilization of the folded bundle by 1.7-2.7 kcal/mol alanine, although the alanine-substituted bundle still exhibits a high degree of thermodynamic stability with an overall folded conformational delta GH2O = 14.3 +/- 0.8 kcal/mol. Covalent attachment of the spin label MTSSL to cysteine residues in the alanine-substituted four-alpha-helix bundle indicates that the di-alpha-helical peptides dimerize in an anti orientation. The rotational correlation time of the four-alpha-helix bundle is 8.1 +/- 0.5 ns, in line with earlier work on similar peptides. Fluorescence, far-UV circular dichroism, and Fourier transform infrared spectroscopies verified the hydrophobic core location of the tryptophan and cysteine residues, showing good agreement between experiment and design. These small synthetic proteins may prove useful for the study of the structural features of small molecule binding sites.


Subject(s)
Anesthetics, Inhalation/metabolism , Drug Design , Halothane/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Protein Structure, Secondary , Amino Acid Sequence , Binding Sites , Cysteine , Electron Spin Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Solubility , Spectroscopy, Fourier Transform Infrared , Tryptophan , Water
14.
Toxicol Lett ; 100-101: 369-75, 1998 Nov 23.
Article in English | MEDLINE | ID: mdl-10049167

ABSTRACT

The structural features of volatile anesthetic binding sites on proteins were explored using a model system consisting of a four-alpha-helix bundle scaffold with a hydrophobic core. This system serves as a model for the lipid-spanning portions of several membrane proteins. Two hydrophobic core designs were compared: H10A24 consisting mainly of leucine residues, and (Aalpha2)2 which has four leucine and two histidine residues replaced by smaller alanines with the intent of forming a cavity. Halothane binds to (Aalpha2)2 with a Kd of 0.71 +/- 0.04 mM as monitored by the quenching of tryptophan fluorescence. This is a 3.2-fold higher affinity compared with binding to H10A24 (Kd = 2.3 +/- 0.4 mM). The presence of a preexisting protein hydrophobic cavity may favor volatile anesthetic binding. Guanidinium chloride denaturation studies reveal that bound anesthetic favors the native folded form of (Aalpha2)2 by 1.8 kcal/mol. The use of synthetic peptides should allow predictions to be made concerning the structural composition of in vivo anesthetic binding sites and may provide clues to how anesthetics alter protein function.


Subject(s)
Anesthetics, Inhalation/chemistry , Peptides/chemistry , Amino Acid Sequence , Binding Sites , Halothane/chemistry , Molecular Sequence Data , Protein Conformation , Protein Denaturation , Spectrometry, Fluorescence
15.
J Biol Chem ; 272(29): 17961-5, 1997 Jul 18.
Article in English | MEDLINE | ID: mdl-9218421

ABSTRACT

The site(s) of action of the volatile general anesthetics remain(s) controversial, but evidence in favor of specific protein targets is accumulating. The techniques to measure directly volatile anesthetic binding to proteins are still under development. Further experience with the intrinsic protein fluorescence quenching approach to monitor anesthetic-protein complexation is reported using chloroform. Chloroform quenches the steady-state tryptophan fluorescence of bovine serum albumin (BSA) in a concentration-dependent, saturable manner with a Kd = 2.7 +/- 0.2 mM. Tryptophan fluorescence lifetime analysis reveals that the majority of the quenching is due to a static mechanism, indicative of anesthetic binding. The ability of chloroform to quench BSA tryptophan fluorescence was decreased markedly in the presence of 50% 2,2,2-trifluoroethanol, which causes loss of tertiary structural contacts in BSA, indicating that protein conformation is crucial for anesthetic binding. Circular dichroism spectroscopy revealed no measurable effect of chloroform on the secondary structure of BSA. The results suggest that chloroform binds to subdomains IB and IIA in BSA, each of which contains a single tryptophan. Earlier work has shown that these sites are also occupied by halothane. The present study therefore provides experimental support for the theory that structurally distinct general anesthetics may occupy the same domains on protein targets.


Subject(s)
Chloroform/blood , Serum Albumin, Bovine/metabolism , Serum Albumin/metabolism , Anesthetics, Inhalation , Animals , Binding Sites , Cattle , Circular Dichroism , Humans , Kinetics , Protein Binding , Protein Conformation , Serum Albumin/chemistry , Serum Albumin, Bovine/chemistry , Spectrometry, Fluorescence , Tryptophan
16.
Biochemistry ; 36(10): 2798-806, 1997 Mar 11.
Article in English | MEDLINE | ID: mdl-9062107

ABSTRACT

A maleimide nitroxide spin-label (MAL-6) linked to a cysteine in the hydrophobic core and a coproporphyrin I (CP) appended on the N-terminus of a synthetic helix-loop-helix peptide ([alpha2]) have been used to examine the designed self-association of a four-helix bundle ([alpha2]2), focusing on the bundle topology and stability and the rotational dynamics of the spin-label. Gel-permeation chromatography demonstrated that the [alpha2] peptide and the peptide modified with a spin-label ([MAL-6-alpha2]), a coproporphyrin ([CP-alpha2]) and a coproporphyrin plus a spin-label ([CP-MAL-6-alpha2]) self-associate into four helix bundles in solution as designed. Circular dichroism (CD) spectra prove that all these peptides are highly alpha-helical, confirmed for [alpha2]2 by Fourier transform infrared (FTIR) spectroscopic analysis. Electron spin resonance (ESR) spectra of the two attached maleimide spin-labels in [MAL-6-alpha2]2 shows their effective rotational correlation time (tau(c)) is 7.3 +/- 0.5 ns, consistent with that expected for the tumbling of the four helix bundle itself, indicating the labels are immobilized. The ESR spectra were also unaltered by aqueous-phase paramagnetic ions, Ni(II), demonstrating all of the spin-labels are buried within the hydrophobic core. The lack of spin-spin interaction between the buried, immobilized spin-labels indicates they are remote (> 15 A) from each other, indicating an antiparallel topology of the monomers in [MAL-6-alpha2]2. The parent [alpha2]2 and the modified [MAL-6-alpha2]2 and [CP-alpha2]2 peptides are highly stable (deltaG(H2O) approximately 25 kcal/mol) as investigated by guanidine hydrochloride denaturation curves monitored by ESR and CD spectroscopies. Guanidine hydrochloride denaturation leads to a shorter correlation time of the spin-label, tau(c) < 1 ns, approaching that of an unrestricted spin-label in solution. In contrast, trifluoroethanol caused dissociation of [MAL-6-alpha2]2 to yield two [MAL-6-alpha2] monomers with retention of secondary structure and changed the tau(c) to 2.5 +/- 0.5 ns, indicating that a significant degree of motional restriction is imposed on the spin-label by the secondary structure. The coproporphyrin probes covalently attached to the N-termini of [CP-alpha2]2 and [CP-MAL-6-alpha2]2 provided evidence that the helical monomers of both were in a parallel orientation, in contrast to the antiparallel orientation determined for [MAL-6-alpha2]2. Consequently, the ESR spectra of [MAL-6-alpha2]2 and [CP-MAL-6-alpha2]2 reveal major structural differences in the local vicinity of the spin-labels due to the topological difference between these two bundles. The ESR spectra of [CP-MAL-6-alpha2]2 contains two distinct nitroxide populations, indicating that one spin-label remains buried in the hydrophobic core and the other is excluded to solvent in this parallel topology. Alleviation of the steric interactions causing one spin-label in [CP-MAL-6-alpha2]2 to be solvent-exposed by addition of [CP-alpha2]2 results in formation of the heterodimeric [CP-alpha2]/[CP-MAL-6-alpha2], as evidenced by insertion of all the spin-labels into hydrophobic cores. The changes in global topology and local structure as evidenced by this pair of spectral probes have relatively minor effects on the course of guanidine denaturation of these bundles.


Subject(s)
Helix-Loop-Helix Motifs , Peptides/chemistry , Protein Conformation , Protein Structure, Secondary , Amino Acid Sequence , Circular Dichroism , Coproporphyrins , Cyclic N-Oxides , Electron Spin Resonance Spectroscopy , Guanidine , Guanidines , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Molecular Structure , Peptides/chemical synthesis , Protein Denaturation , Spectrophotometry , Spin Labels , Thermodynamics
17.
Pharmacol Rev ; 49(4): 343-67, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9443162

ABSTRACT

The fundamental interactions of inhalational anesthetics with proteins have been considered in some detail, using specific examples where appropriate to illustrate these interactions and demonstrate progress. It is now clear that these low-affinity volatile molecules with rapid kinetics can specifically bind to discrete sites in some proteins at reasonable pharmacological concentrations, and some general features of these sites are beginning to emerge. The structural or dynamic consequences of anesthetic binding, however, are still vague at best. The remaining challenge is to define which interactions produce anesthetic binding to relevant targets and what the features of this relevant anesthetic binding site are. Finally, and most importantly, how does the occupancy of these pockets, patches, or cavities result in the subtle alterations in protein conformation and dynamics that confound their function and ultimately produce the behavioral response that we term "anesthesia"?


Subject(s)
Anesthetics, Inhalation/metabolism , Protein Conformation , Proteins/metabolism , Anesthetics, Inhalation/chemistry , Binding Sites , Kinetics , Protein Binding , Proteins/chemistry , Thermodynamics
18.
J Pharmacol Exp Ther ; 279(1): 56-61, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8858975

ABSTRACT

Although volatile general anesthetics interact with several proteins, little is known about the location or characteristics of the binding sites at the molecular level. A detailed structural description of how anesthetics associate with macromolecules is necessary for understanding anesthetic mechanisms of action. The recent introduction of designed synthetic proteins provides new opportunities for obtaining structural and functional information on anesthetic-protein interactions. A synthetic tetra-alpha-helix-bundle protein was used to examine the interaction of halothane with a designed protein interior. The tetra-alpha-helix-bundle comprises 124 residues in the form of two identical 62-residue di-alpha-helical peptides, held together in an all-parallel bundle by hydrophobic forces. Steady-state and time-resolved tryptophan fluorescence and circular dichroism spectroscopy were used to study the anesthetic-protein interaction. Halothane quenches bundle tryptophan fluorescence with a dissociation constant of 2.3 +/- 0.4 mM and a Hill number of 0.9 +/- 0.1. Tryptophan fluorescence decay analysis indicates that halothane quenches the protein fluorescence by a static mechanism. Circular dichroism spectroscopy revealed no change in protein secondary structure on exposure to halothane. Dissociation of the tetra-alpha-helix-bundle into 62-residue di-alpha-helical peptides by trifluoroethanol eliminated the halothane-protein interaction. The results suggest that halothane binds to the hydrophobic interior of the tetra-alpha-helix-bundle, close to the tryptophan residues. The protein tertiary and quaternary structures are required for anesthetic binding. This study demonstrates the feasibility of using synthetic tetra-alpha-helix-bundles as model anesthetic-binding proteins. The use of de novo designed bundle proteins should allow structural, energetic and functional descriptions of anesthetic-protein interactions.


Subject(s)
Anesthetics, Inhalation/metabolism , Halothane/metabolism , Amino Acid Sequence , Binding Sites , Fluorescence , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Structure-Activity Relationship
20.
Biochim Biophys Acta ; 1290(1): 63-8, 1996 May 21.
Article in English | MEDLINE | ID: mdl-8645708

ABSTRACT

The present study makes use of direct photoaffinity labeling and fluorescence and circular dichroism spectroscopy to examine the interaction of the inhalational anesthetic halothane with the uncharged alpha-helical form of poly(L-lysine) over a range of chain lengths. Halothane bound specifically to long chain homopolymers (190 to 1060 residues), reaching a stable stoichiometry of 1 halothane to 160 lysine residues in polymers longer than 300 residues. Halothane bound only non-specifically to an alpha-helical 30 residue polymer and to all of the polymers in their charged, random coil form. The data suggest that halothane binding is a function of supersecondary structure whereby intramolecular helix-helix clusters form in the longer polymers, resulting in the creation of confined hydrophobic domains. Circular dichroism spectroscopy cannot demonstrate changes in poly(L-lysine) secondary structure at any chain length with up to 12 mM halothane, suggesting that extensive hydrogen bond disruption by the anesthetic does not occur.


Subject(s)
Anesthetics, Inhalation/metabolism , Affinity Labels , Anesthetics, Inhalation/chemistry , Anilino Naphthalenesulfonates , Binding Sites , Circular Dichroism , Fluorescent Dyes , Protein Binding , Protein Structure, Secondary , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...