Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Top Membr ; 78: 117-44, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27586283

RESUMO

Voltage-gated sodium channels are targets for many toxins and medically important drugs. Despite decades of intensive studies in industry and academia, atomic mechanisms of action are still not completely understood. The major cause is a lack of high-resolution structures of eukaryotic channels and their complexes with ligands. In these circumstances a useful approach is homology modeling that employs as templates X-ray structures of potassium channels and prokaryotic sodium channels. On one hand, due to inherent limitations of this approach, results should be treated with caution. In particular, models should be tested against relevant experimental data. On the other hand, docking of drugs and toxins in homology models provides a unique possibility to integrate diverse experimental data provided by mutational analysis, electrophysiology, and studies of structure-activity relations. Here we describe how homology modeling advanced our understanding of mechanisms of several classes of ligands. These include tetrodotoxins and mu-conotoxins that block the outer pore, local anesthetics that block of the inner pore, batrachotoxin that binds in the inner pore but, paradoxically, activates the channel, pyrethroid insecticides that activate the channel by binding at lipid-exposed repeat interfaces, and scorpion alpha and beta-toxins, which bind between the pore and voltage-sensing domains and modify the channel gating. We emphasize importance of experimental data for elaborating the models.


Assuntos
Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Batraquiotoxinas/química , Batraquiotoxinas/metabolismo , Batraquiotoxinas/farmacologia , Sítios de Ligação , Conotoxinas/química , Conotoxinas/metabolismo , Conotoxinas/toxicidade , Inseticidas/química , Inseticidas/metabolismo , Inseticidas/toxicidade , Ativação do Canal Iônico/efeitos dos fármacos , Ligantes , Simulação de Dinâmica Molecular , Método de Monte Carlo , Estrutura Terciária de Proteína , Piretrinas/química , Piretrinas/metabolismo , Piretrinas/toxicidade , Esteroides/química , Esteroides/metabolismo , Tetrodotoxina/química , Tetrodotoxina/metabolismo , Tetrodotoxina/toxicidade , Agonistas do Canal de Sódio Disparado por Voltagem/química , Agonistas do Canal de Sódio Disparado por Voltagem/metabolismo , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/metabolismo , Canais de Sódio Disparados por Voltagem/química
2.
Zh Evol Biokhim Fiziol ; 52(1): 26-33, 2016.
Artigo em Russo | MEDLINE | ID: mdl-27220237

RESUMO

An integral part of modern evolutionary biology is comparative analysis of structure and function of macromolecules such as proteins. The first and critical step to understand evolution of homologous proteins is their amino acid sequence alignment. However, standard algorithms fop not provide unambiguous sequence alignments for proteins of poor homology. More reliable results can be obtained by comparing experimental 3D structures obtained at atomic resolution, for instance, with the aid of X-ray structural analysis. If such structures are lacking, homology modeling is used, which may take into account indirect experimental data on functional roles of individual amino-acid residues. An important problem is that the sequence alignment, which reflects genetic modifications, does not necessarily correspond to the functional homology. The latter depends on three-dimensional structures which are critical for natural selection. Since alignment techniques relying only on the analysis of primary structures carry no information on the functional properties of proteins, including 3D structures into consideration is very important. Here we consider several examples involving ion channels and demonstrate that alignment of their three-dimensional structures can significantly improve sequence alignments obtained by traditional methods.


Assuntos
Evolução Molecular , Canais Iônicos/química , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Conformação Proteica , Alinhamento de Sequência/métodos
3.
Eur Biophys J ; 45(4): 321-9, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26646260

RESUMO

The family of P-loop channels includes potassium, sodium, calcium, cyclic nucleotide-gated and TRPV channels, as well as ionotropic glutamate receptors. Despite vastly different physiological and pharmacological properties, the channels have structurally conserved folding of the pore domain. Furthermore, crystallographic data demonstrate surprisingly similar mutual disposition of transmembrane and membrane-diving helices. To understand determinants of this conservation, here we have compared available high-resolution structures of sodium, potassium, and TRPV1 channels. We found that some residues, which are in matching positions of the sequence alignment, occur in different positions in the 3D alignment. Surprisingly, we found 3D mismatches in well-packed P-helices. Analysis of energetics of individual residues in Monte Carlo minimized structures revealed cyclic patterns of energetically favorable inter- and intra-subunit contacts of P-helices with S6 helices. The inter-subunit contacts are rather conserved in all the channels, whereas the intra-subunit contacts are specific for particular types of the channels. Our results suggest that these residue-residue contacts contribute to the folding stabilization. Analysis of such contacts is important for structural and phylogenetic studies of homologous proteins.


Assuntos
Canais de Potássio/química , Dobramento de Proteína , Canais de Sódio/química , Canais de Cátion TRPV/química , Sequência de Aminoácidos , Modelos Moleculares , Método de Monte Carlo , Conformação Proteica , Estabilidade Proteica
4.
Curr Med Chem ; 19(25): 4306-23, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22709009

RESUMO

1,4-Dihydropyridines were introduced in the last century for the treatment of coronary diseases. Then medicinal chemists decorated the 1,4-DHP nucleus, the most studied scaffold among L-type calcium channel blockers, achieving diverse activities at several receptors, channels and enzymes. We already described (Ioan et al. Curr. Med. Chem. 2011, 18, 4901-4922) the effects of 1,4-DHPs at ion channels and G-protein coupled receptors. In this paper we continue the analysis of the wide range of biological effects exerted by compounds belonging to this chemical class. In particular, focus is given to the ability of 1,4-DHPs to revert multi drug resistance that, after over 20 years of research, continues to be of great interest. We also describe activities on other targets and the action of 1,4-DHPs against several diseases. Finally, we report and review the interaction of 1,4-DHPs with the hERG channel, transporters and phase I metabolizing enzymes. This work is a starting point for further exploration of the 1,4-DHP core activities on targets, off-targets and antitargets.


Assuntos
Di-Hidropiridinas/química , Di-Hidropiridinas/farmacologia , Doença de Alzheimer/tratamento farmacológico , Animais , Antituberculosos/química , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Aterosclerose/tratamento farmacológico , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Di-Hidropiridinas/uso terapêutico , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Humanos , Fator de Ativação de Plaquetas/antagonistas & inibidores , Tuberculose/tratamento farmacológico
5.
Curr Med Chem ; 18(32): 4901-22, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22050742

RESUMO

Since the pioneering studies of Fleckenstein and co-workers, L-Type Calcium Channel (LTCC) blockers have attracted large interest due to their effectiveness in treating several cardiovascular diseases. Medicinal chemists achieved high potency and tissue selectivity by decorating the 1-4-DHP nucleus, the most studied scaffold among LTCC blockers. Nowadays it is clear that the 1,4-DHP nucleus is a privileged scaffold since, when appropriately substituted, it can selectively modulate diverse receptors, channels and enzymes. Therefore, the 1,4-DHP scaffold could be used to treat various diseases by a single-ligand multi-target approach. In this review, we describe the structure-activity relationships of 1,4-DHPs at ion channels, G-protein coupled receptors, and outline the potential for future therapeutic applications.


Assuntos
Di-Hidropiridinas/química , Canais Iônicos/química , Receptores Acoplados a Proteínas G/química , Animais , Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Química Farmacêutica , Di-Hidropiridinas/farmacologia , Humanos , Canais Iônicos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Relação Estrutura-Atividade
6.
Ross Fiziol Zh Im I M Sechenova ; 97(7): 661-77, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21961291

RESUMO

Ion channels in cell membranes are targets for a multitude of ligands including naturally occurring toxins, illicit drugs, and medications used to manage pain and treat cardiovascular, neurological, autoimmune, and other health disorders. In the past decade, the x-ray crystallography revealed 3D structures of several ion channels in their open, closed, and inactivated states, shedding light on mechanisms of channel gating, ion permeation and selectivity. However, atomistic mechanisms of the channel modulation by ligands are poorly understood. Increasing evidence suggest that cationophilic groups in ion channels and in some ligands may simultaneously coordinate permeant cations, which form indispensible (but underappreciated) components of respective receptors. This review describes ternary ligand-metal-channel complexes predicted by means of computer-based molecular modeling. The models rationalize a large body of experimental data including paradoxes in structure-activity relationships, effects of mutations on the ligand action, sensitivity of the ligand action to the nature of current-carrying cations, and action of ligands that bind in the ion-permeation pathway but increase rather than decrease the current. Recent mutational and ligand-binding experiments designed to test the models have confirmed the ternary-complex concept providing new knowledge on physiological roles of metal ions and atomistic mechanisms of action of ion channel ligands.


Assuntos
Canais de Cálcio , Cálcio/metabolismo , Ativação do Canal Iônico/fisiologia , Canais de Potássio , Potássio/metabolismo , Canais de Sódio , Sódio/metabolismo , Canais de Cálcio/química , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Cátions/metabolismo , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cristalografia por Raios X , Medicamentos Essenciais/farmacologia , Humanos , Drogas Ilícitas/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Ligantes , Modelos Químicos , Modelos Moleculares , Canais de Potássio/química , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Ligação Proteica/efeitos dos fármacos , Canais de Sódio/química , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo , Relação Estrutura-Atividade , Toxinas Biológicas/farmacologia
7.
Mini Rev Med Chem ; 9(12): 1379-88, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19929811

RESUMO

The diltiazem binding site of L-type calcium channels is the least characterized to date. In this paper, we present some of the available chemotypes that bind to the benzothiazepine binding site: natural compounds, compounds synthesized by varying the benzothiazepine scaffold, and compounds discovered by means of computational approaches.


Assuntos
Bloqueadores dos Canais de Cálcio/química , Diltiazem/análogos & derivados , Ligantes , Sítios de Ligação , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Simulação por Computador , Diltiazem/química
9.
J Biol Chem ; 276(45): 42091-8, 2001 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-11514561

RESUMO

The first crystallographic structure of human type 3 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD3, AKR1C2), an enzyme playing a critical role in steroid hormone metabolism, has been determined in complex with testosterone and NADP at 1.25-A resolution. The enzyme's 17beta-HSD activity was studied in comparison with its 3alpha-HSD activity. The enzyme catalyzes the inactivation of dihydrotestosterone into 5alpha-androstane-3alpha,17beta-diol (3alpha-diol) as well as the transformation of androstenedione into testosterone. Using our homogeneous and highly active enzyme preparation, we have obtained 150-fold higher 3alpha-HSD specificity as compared with the former reports in the literature. Although the rat and the human 3alpha-HSDs share 81% sequence homology, our structure reveals significantly different geometries of the active sites. Substitution of the Ser(222) by a histidine in the human enzyme may compel the steroid to adopt a different binding to that previously described for the rat (Bennett, M. J., Albert, R. H., Jez, J. M., Ma, H., Penning, T. M., and Lewis, M. (1997) Structure 5, 799-T812). Furthermore, we showed that the affinity for the cofactor is higher in the human 3alpha-HSD3 than the rat enzyme due to the presence of additional hydrogen bonds on the adenine moiety and that the cofactor is present under its reduced form in the active site in our preparation.


Assuntos
3-Hidroxiesteroide Desidrogenases/química , Isoenzimas/química , NADP/química , Testosterona/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , 3-Hidroxiesteroide Desidrogenases/metabolismo , 3-alfa-Hidroxiesteroide Desidrogenase (B-Específica) , Sítios de Ligação , Humanos
10.
Arch Biochem Biophys ; 393(1): 22-41, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11516158

RESUMO

L-type calcium channels (LCCs) are transmembrane (TM) proteins that respond to membrane depolarization by selectively permeating Ca(2+) ions. Dihydropyridine (DHP) agonists and antagonist modulate Ca(2+) permeation by stabilizing, respectively, the open and closed states of the channel. The mechanism of action of these drugs remains unclear. Using, as a template, the crystal structure of the KcsA K(+) channel (Doyle et al. (1998) Science 280, 69-77), we have built several homology models of LCC with alternative alignments of TM segments between the proteins. In each model, nifedipine was docked in the pore region and in the interface between repeats III and IV. Several starting structures were generated by constraining the ligand to residues whose mutations reportedly affect DHP binding (DHP-sensing residues). These structures were Monte Carlo-minimized with and without constraints. In the complex with the maximum number of contacts between the ligand and DHP-sensing residues and the lowest ligand-receptor energy, the drug fits snugly in the "water-lake" cavity between segments S6s, which were aligned with M2 segment of KcsA as proposed for Na(+) channel (Lipkind and Fozzard (2000) Biochemistry 39, 8161-8170). In the flattened-boat conformation of DHP ring, the NH group at the stern approaches the DHP-sensing tyrosines in segments IIIS6 and IVS6. Stacking interactions of IVS6 Tyr with the bowsprit aromatic ring stabilize the ligand's orientation in which the starboard COOMe group coordinates Ca(2+) ion chelated by two conserved glutamates in the selectivity filter. In the inverted teepee structure of LCC, the portside COOMe group approaches a bracelet of conserved hydrophobic residues at the helical-bundle crossing, which may function as the activation gate. The dimensions of the gate may readily change upon small rotation of the pore-forming TM segments. The end of the portside group is hydrophobic in nifedipine, (R)-Bay K 8644, and other antagonists. Favorable interactions of this group with the hydrophobic bracelet would stabilize its closed conformation. In contrast, (S)-Bay K 8644 and several other agonists have hydrophilic groups at the portside. Unfavorable interactions of the hydrophilic group with the hydrophobic bracelet would destabilize its closed conformation thereby stabilizing the open conformation. In the agonist-bound channel, Ca(2+) ions would permeate between the hydrophilic face of the ligand and conserved hydrophilic residues in segments IS6 and IIS6. Our model suggests mutational experiments that could further our understanding of the pharmacological modulation of voltage-gated ion channels.


Assuntos
Proteínas de Bactérias , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Fenômenos Biofísicos , Biofísica , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Técnicas In Vitro , Ativação do Canal Iônico , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Nifedipino/metabolismo , Canais de Potássio/química , Canais de Potássio/genética , Conformação Proteica , Coelhos , Homologia de Sequência de Aminoácidos
11.
Biophys J ; 78(4): 1786-803, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10733960

RESUMO

GABA and glycine receptors (GlyRs) are pentameric ligand-gated ion channels that respond to the inhibitory neurotransmitters by opening a chloride-selective central pore lined with five M2 segments homologous to those of alpha(1) GlyR/ ARVG(2')LGIT(6')TVLTMTTQSSGSR. The activity of cyanotriphenylborate (CTB) and picrotoxinin (PTX), the best-studied blockers of the Cl(-) pores, depends essentially on the subunit composition of the receptors, in particular, on residues in positions 2' and 6' that form the pore-facing rings R(2') and R(6'). Thus, CTB blocks alpha(1) and alpha(1)/beta, but not alpha(2) GlyRs (Rundström, N., V. Schmieden, H. Betz, J. Bormann, and D. Langosch. 1994. Proc. Natl. Acad. Sci. U.S.A. 91:8950-8954). PTX blocks homomeric receptors (alpha(1) GlyR and rat rho(1) GABAR), but weakly antagonizes heteromeric receptors (alpha(1)/beta GlyR and rho(1)/rho(2) GABAR) (Pribilla, I., T. Takagi, D. Langosch, J. Bormann, and H. Betz. 1992. EMBO J. 11:4305-4311; Zhang D., Z. H. Pan, X. Zhang, A. D. Brideau, and S. A. Lipton. 1995. Proc. Natl. Acad. Sci. U.S.A. 92:11756-11760). Using as a template the kinked-helices model of the nicotinic acetylcholine receptor in the open state (Tikhonov, D. B., and B. S. Zhorov. 1998. Biophys. J. 74:242-255), we have built homology models of GlyRs and GABARs and calculated Monte Carlo-minimized energy profiles for the blockers pulled through the pore. The profiles have shallow minima at the wide extracellular half of the pore, a barrier at ring R(6'), and a deep minimum between rings R(6') and R(2') where the blockers interact with five M2s simultaneously. The star-like CTB swings necessarily on its way through ring R(6') and its activity inversely correlates with the barrier at R(6'): Thr(6')s and Ala(2')s in alpha(2) GlyR confine the swinging by increasing the barrier, while Gly(2')s in alpha(1) GlyR and Phe(6')s in beta GlyR shrink the barrier. PTX has an egg-like shape with an isopropenyl group at the elongated end and the rounded end trimmed by ether and carbonyl oxygens. In the optimal binding mode to alpha(1) GlyR and rho(1) GABAR, the rounded end of PTX accepts several H-bonds from Thr(6')s, while the elongated end enters ring R(2'). The lack of H-bond donors on the side chains of Phe(6')s (beta GlyR) and Met(6')s (rho(2) GABAR) deteriorates the binding. The hydrophilic elongated end of picrotin does not fit the hydrophobic ring of Pro(2')s/Ala(2')s in GABARs, but fit a more hydrophilic ring with Gly(2')s in GlyRs. This analysis provides explanations for structure-activity relationships of noncompetitive agonists and predicts a narrow pore of LGICs in agreement with experimental data on the permeation of organic cations.


Assuntos
Canais de Cloreto/metabolismo , Receptores de GABA/metabolismo , Receptores de Glicina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Fenômenos Biofísicos , Biofísica , Boratos/química , Boratos/farmacologia , Canais de Cloreto/química , Canais de Cloreto/genética , Humanos , Substâncias Macromoleculares , Modelos Moleculares , Dados de Sequência Molecular , Método de Monte Carlo , Picrotoxina/análogos & derivados , Picrotoxina/química , Picrotoxina/farmacologia , Ratos , Receptores de GABA/química , Receptores de GABA/genética , Receptores de Glicina/química , Receptores de Glicina/genética , Sesterterpenos , Relação Estrutura-Atividade , Termodinâmica
12.
Proteins ; 38(4): 414-27, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10707028

RESUMO

17 beta-Estradiol (E2) is a potent stimulator of certain forms of breast cancer. The final step of E2 biosynthesis is catalyzed by the estrogenic 17 beta-hydroxysteroid dehydrogenase (17 beta-HSD1), which is an important target for anti-cancer drugs. X-ray crystallography indicated that the binding site for the steroids has a tunnel-like shape. We have used a Monte Carlo-Minimization (MCM) protocol to explore possibilities of interactions of E2 with the binding site tunnel of 17 beta-HSD1. The enzyme was represented by flexible residues having at least one atom within 6 A from either E2 or NADP (as seen in a crystal ternary complex) and by rigid residues having at least one atom within 10 A from E2 or NADP. Special constraints were used to pull the substrate 10 A along the tunnel with 1 A step; the complex was MCM-optimized at each position of the steroid. The optimal binding mode of E2 in 17 beta-HSD agrees with the crystallographic data; however, wide and flat minima of the MCM profile suggest alternative modes of the steroid binding. The advance of the steroid along the tunnel is accompanied by essential conformational rearrangements of the enzyme side chains, noticeable rotation of the substrate along its longitudinal axis, and certain conformational deformations of the substrate. The contributions of the enzyme residues and of the steroid atoms to the intermolecular energy were estimated.


Assuntos
17-Hidroxiesteroide Desidrogenases/química , Estradiol/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Estradiol/metabolismo , Ligantes , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Método de Monte Carlo , Ligação Proteica , Conformação Proteica
13.
J Pept Sci ; 6(2): 57-83, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10718128

RESUMO

The biologically relevant conformation of substance P is likely to be dictated by the lipid milieu wherein the hormone would interact with its receptor. Assuming that specific constraints to the hormone structure may be imparted by its interaction with Ca2+ ions in the low dielectric lipid medium, the interaction of substance P and its inactive analog, Ala7-substance P, has been characterized in a lipid-mimetic solvent. Circular dichroism (CD) and NMR spectral methods were employed to study the conformation of the free and Ca2+-bound forms of the peptides and the conformational changes that occur on Ca2+ binding. The results show that both peptides assume a helical structure in the non-polar solvent used, a mixture of acetonitrile and trifluoroethanol. The N-terminal region is, however, less ordered in the analog peptide compared with the native hormone. Ca2+ addition causes significant conformational changes in both the peptides. However, while substance P binds two Ca2+ ions in a cooperative manner, Ala7-substance P binds only one Ca2+ ion with a relatively weaker affinity. Computations of the minimum-energy conformations of the free and Ca2+-bound peptides were performed using interproton distances derived from nuclear Overhauser enhancement spectra of the two peptides, as well as the information provided by changes in proton chemical shifts caused by Ca2+ addition. Taken together, the results of this study suggest that differences in the interaction of substance P and Ala7-substance P with Ca2+ in the non-polar milieu, which in turn leads to differences in their Ca2+-bound conformations, may be the basis for the differences in their biological potencies.


Assuntos
Cálcio/química , Substância P/química , Dicroísmo Circular , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Oxigênio/química , Ligação Proteica , Conformação Proteica , Solventes/química , Substância P/análogos & derivados
14.
Arch Biochem Biophys ; 375(1): 31-49, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10683246

RESUMO

Metal ions affect ligand binding to G-protein-coupled receptors by as yet unknown mechanisms. In particular, Na(+) increases the affinity for antagonists but decreases it for agonists. We had modeled the mu-opioid receptor (muR) based on the low-resolution structure of rhodopsin by G. F. X. Schertler, C. Villa, and R. Henderson (1993, Nature 362, 770-772) and proposed that metal ions may be directly involved in the binding of ligands and receptor activation (B. S. Zhorov and V. S. Ananthanarayanan, 1998, J. Biomol. Struct. Dyn. 15, 631-637). Developing this concept further, we present here homology models of muR using as templates the structure of rhodopsin elaborated by I. D. Pogozheva, A. L. Lomize, and H. I. Mosberg (1997, Biophys. J. 70, 1963-1985) and J. M. Baldwin, G. F. X. Schertler, and V. M. Unger (1997, J. Mol. Biol., 272, 144-164). Using the Monte Carlo minimization (MCM) method, we docked the Na(+)-bound forms of muR ligands: naloxone, bremazocine, and carfentanyl. The resultant low-energy complexes showed that the two positive charges in the protonated metal-bound ligands interact with the two negative charges at Asp(3.32) and Asp(2.50) (for notations, see J. A. Ballesteros and H. Weinstein, 1995, Methods Neurosci. 25, 366-426). MCM computation on morphine docked inside the model of muR by I. D. Pogozheva, A. L. Lomize, and H. I. Mosberg (1998, Biophys. J. 75, 612-634) yielded two binding modes with the ligand's ammonium group salt-bridged either to Asp(3.32) (generally regarded as the ligand recognition site) or to Asp(2.50). The latter is the presumed site for Na(+) ion, which is known to modulate ligand binding. Assuming that in the low-dielectric transmembrane region of muR, organic and inorganic cations would compete for Asp(3.32) and Asp(2.50), we propose that ligand binding, as visualized in the above models, would first displace Na(+) from Asp(3.32). A subsequent progress of the ligand toward Asp(2.50) would result in either the retention of Na(+) at Asp(2.50) in the case of antagonists or the displacement of Na(+) from Asp(2.50) in the case of agonists. The displaced Na(+) would move toward the salt-bridged Asp(3.49)-Arg(3.50) and disengage the salt bridge. This, in turn, would result in conformational changes at the cytoplasmic face of the receptor that facilitate the interaction with the G-protein.


Assuntos
Ácido Aspártico/metabolismo , Modelos Moleculares , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Sequência de Aminoácidos , Analgésicos Opioides/metabolismo , Animais , Benzomorfanos/metabolismo , Sítios de Ligação/genética , Sítios de Ligação/fisiologia , Cátions/metabolismo , Bovinos , Simulação por Computador , Sequência Conservada , Fentanila/análogos & derivados , Fentanila/metabolismo , Humanos , Ligantes , Metais/metabolismo , Dados de Sequência Molecular , Morfina/metabolismo , Naloxona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/genética , Ratos , Receptores Opioides mu/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
15.
Biophys J ; 77(4): 1914-26, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10512812

RESUMO

Specific electrophysiological and pharmacological properties of ionic channels in NMDA, AMPA, and kainate subtypes of ionotropic glutamate receptors (GluRs) are determined by the Asn (N), Gln (Q), and Arg (R) residues located at homologous positions of the pore-lining M2 segments (the N/Q/R site). Presumably, the N/Q/R site is located at the apex of the reentrant membrane loop and forms the narrowest constriction of the pore. Although the shorter Asn residues are expected to protrude in the pore to a lesser extent than the longer Gln residues, the effective dimension of the NMDA channel (corresponding to the size of the largest permeant organic cation) is, surprisingly, smaller than that of the AMPA channel. To explain this paradox, we propose that the N/Q/R residues form macrocyclic structures (rings) stabilized by H-bonds between a NH(2) group in the side chain of a given M2 segment and a C==O group of the main chain in the adjacent M2 segment. Using Monte Carlo minimization, we have explored conformational properties of the rings. In the Asn, but not in the Gln ring, the side-chain oxygens protruding into the pore may facilitate ion permeation and accept H-bonds from the blocking drugs. In this way, the model explains different electrophysiological and pharmacological properties of NMDA and non-NMDA GluR channels. The ring of H-bonded polar residues at the pore narrowing resembles the ring of four Thr(75) residues observed in the crystallographic structure of the KcsA K(+) channel.


Assuntos
Arginina/metabolismo , Asparagina/metabolismo , Glutamina/metabolismo , Modelos Moleculares , Receptores de Glutamato/química , Receptores de Glutamato/metabolismo , Arginina/química , Asparagina/química , Ácido Aspártico/metabolismo , Sítios de Ligação , Cálcio/metabolismo , Antagonistas de Aminoácidos Excitatórios/química , Antagonistas de Aminoácidos Excitatórios/metabolismo , Glutamina/química , Ligação de Hidrogênio , Cinética , Método de Monte Carlo , Oxigênio/metabolismo , Conformação Proteica , Receptores de AMPA/química , Receptores de AMPA/metabolismo , Receptores de Ácido Caínico/química , Receptores de Ácido Caínico/metabolismo , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo , Eletricidade Estática
16.
Bioorg Khim ; 25(5): 348-57, 1999 May.
Artigo em Russo | MEDLINE | ID: mdl-10495892

RESUMO

A conformational variability of the collagen triple helix was studied with the methods of molecular mechanics. The Rich-Crick model with one hydrogen bond per tripeptide fragment or the model with two hydrogen bonds per tripeptide fragment were used for tripeptides forming the primary structure of the protein. Imino acid and amino acid residues were located in the second position of the tripeptide fragments in the first and second cases, respectively. Conformations on domain boundaries, which had alternating structures with one and two hydrogen bonds per tripeptide, were particularly studied. Essentially all types of collagen backbone composed of amino acid residues most frequently occurring in this protein were considered. A new model was suggested that combined elements of the Rich-Crick model and our new approach. This was shown to be stereochemically valid, energetically advantageous, and consistent with the experimental data. It was conclusively demonstrated that the primary structure of collagen determines its tertiary structure.


Assuntos
Colágeno/química , Ligação de Hidrogênio , Modelos Moleculares , Conformação Proteica , Estereoisomerismo
17.
J Biomol Struct Dyn ; 16(4): 977-88, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10217463

RESUMO

This paper concerns the conformational variability of collagen as related to the concrete tripeptides (GXY)n constituting its primary structure. The previously elaborated model (V.G.Tumanyan, N.G.Esipova, Biophysics 28, 1021-1025, 1983) with two nets of hydrogen bonds is useful for tripeptides where X is an amino acid. If X is an imino acid, the common one-bonded Rich & Crick model is valid. In this work, compound sequences including tripeptides of different types are considered. Molecular mechanics is used to assess the conformations of the junction regions when a structure with two nets of hydrogen bonds precedes the structure with one net, and vice versa. Thus, all types of sequences typical for natural collagen are covered. It is shown that the combined model representing an alternation of the two-H-bonded model and the one-H-bonded Rich & Crick model is satisfactory stereochemically, and provides more favorable energy in comparison with the continuous one-H-bonded model. Besides, a more favorable hydration of the molecule occures in this case. Some conclusions are made about interchain and intrachain ionic bonds. Thus, it is deduced for the concrete fibrillar protein how a one-dimensional structure determines three-dimensional structure. The macromolecular structure thus suggested is in accord with the experimental data on hydrogen exchange.


Assuntos
Colágeno/química , Ligação de Hidrogênio , Simulação por Computador , DNA/química , Modelos Moleculares
18.
J Biomol Struct Dyn ; 15(4): 631-7, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9514241

RESUMO

Based on molecular modeling of the complexes between the mu-opioid receptor and its ligands, we present a hypothesis that accounts for several of the experimental data including the importance of conserved polar residues in rhodopsin-like G-protein-coupled receptors and the effect of Na+ on the binding of ligands to these receptors. We propose that agonists, but not antagonists, would displace Na+ from its initial binding site at the conserved D2.50 residue in the second transmembrane alpha-helical segment, H2. The displaced Na+ would pass through a "gate" of conserved hydrophobic residues and move along a tunnel-like interface (formed of H2, H3 and H7) enriched with several conserved hydrophilic residues including D3.49. Interaction of Na+ with D3.49 would result in the breaking of a salt-bridge between D3.49 and the conserved R3.50 residue thus exposing the latter for interaction with the G-protein.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Modelos Moleculares , Receptores Opioides mu/química , Transdução de Sinais/fisiologia , Sódio/química , Sequência de Aminoácidos , Animais , Membrana Celular/química , Íons , Ligantes , Dados de Sequência Molecular , Morfina/agonistas , Morfina/química , Antagonistas de Entorpecentes , Ratos , Rodopsina/química , Relação Estrutura-Atividade
19.
Biophys J ; 74(1): 242-55, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9449326

RESUMO

A model of the nicotinic acetylcholine receptor ion channel was elaborated based on the data from electron microscopy, affinity labeling, cysteine scanning, mutagenesis studies, and channel blockade. A restrained Monte Carlo minimization method was used for the calculations. Five identical M2 segments (the sequence EKMTLSISVL10LALTVFLLVI20V) were arranged in five-helix bundles with various geometrical profiles of the pore. For each bundle, energy profiles for chlorpromazine, QX-222, pentamethonium, and other blocking drugs pulled through the pore were calculated. An optimal model obtained allows all of the blockers free access to the pore, but retards them at the rings of residues known to contribute to the corresponding binding sites. In this model, M2 helices are necessarily kinked. They come into contact with each other at the cytoplasmic end but diverge at the synaptic end, where N-termini of M1 segments may contribute to the pore. The kinks disengage alpha-helical H-bonds between Ala12 and Ser8. The uncoupled lone electron pairs of Ser8 carbonyl oxygens protrude into the pore, forming a hydrophilic ring that may be important for the permeation of cations. A split network of H-bonds provides a flexibility to the chains Val9-Ala12, the numerous conformations of which form only two or three intrasegment H-bonds. The cross-ectional dimensions of the interface between the flexible chains vary essentially at the level of Leu11. We suggest that conformational transitions in the chains Val9-Ala12 are responsible for the channel gating, whereas rotations of more stable alpha-helical parts of M2 segments may be necessary to transfer the channel in the desensitized state.


Assuntos
Canais Iônicos/química , Antagonistas Nicotínicos/química , Estrutura Secundária de Proteína , Receptores Nicotínicos/química , Sequência de Aminoácidos , Animais , Calorimetria , Galinhas , Simulação por Computador , Sequência Consenso , Ligação de Hidrogênio , Ativação do Canal Iônico , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/fisiologia , Ligantes , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Método de Monte Carlo , Antagonistas Nicotínicos/farmacologia , Ratos , Receptores Nicotínicos/fisiologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Serina , Eletricidade Estática , Estresse Mecânico , Torpedo
20.
Arch Biochem Biophys ; 341(2): 238-44, 1997 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9169010

RESUMO

The mechanism by which diverse drugs modulate voltage-dependent Ca2+ channels is ill-understood. We have approached this problem by examining the interaction of verapamil with a 97-residue synthetic channel peptide (SCP) that exhibits functional similarities to authentic L-type Ca2+ channels in terms of cation selectivity and permeation as well as interaction with channel-activating and blocking drugs (Grove et al. (1991) Proc. Natl. Acad. Sci. USA 88, 6418). Different possibilities of binding of verapamil inside the Ca(2+)-bound SCP were simulated using the Monte Carlo-with-energy-minimization method. In the optimal mode of the binding, verapamil adopted a folded conformation and fit snugly in the pore. The dimethoxyphenyl groups of the drug interacted with two Ca2+ ions coordinated to the acidic residues of SCP, thus forming a ternary complex of the drug, Ca2+, and channel. The isopropyl group of verapamil abetted a ring of four Ile residues constituting the putative SCP gate. The occlusion of this gate by verapamil in this manner was strikingly similar to that accomplished by the methyl group of dihydropyridine drugs. In conjunction with an earlier study on SCP bound to dihydropyridine drugs (Zhorov and Ananthanarayanan (1996) Biophys. J. 70, 22), our data suggest that, in general, drug modulation of SCP would involve the interaction of the ligands with the pore-bound Ca2+ and with the hydrophobic gate. In light of the functional similarity between SCP and L-type Ca2+ channel, it is likely that the latter would also interact with drugs in a similar fashion.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Verapamil/metabolismo , Sequência de Aminoácidos , Canais de Cálcio/síntese química , Canais de Cálcio Tipo L , Simulação por Computador , Transporte de Íons , Substâncias Macromoleculares , Modelos Moleculares , Conformação Molecular , Dados de Sequência Molecular , Método de Monte Carlo , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...