Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 294(21): 8351-8360, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30962282

RESUMEN

Phosphodiesterase-6 (PDE6) plays a central role in both rod and cone phototransduction pathways. In the dark, PDE6 activity is suppressed by its inhibitory γ-subunit (Pγ). Rhodopsin-catalyzed activation of the G protein transducin relieves this inhibition and enhances PDE6 catalysis. We hypothesized that amino acid sequence differences between rod- and cone-specific Pγs underlie transducin's ability to more effectively activate cone-specific PDE6 than rod PDE6. To test this, we analyzed rod and cone Pγ sequences from all major vertebrate and cyclostome lineages and found that rod Pγ loci are far more conserved than cone Pγ sequences and that most of the sequence differences are located in the N-terminal region. Next we reconstituted rod PDE6 catalytic dimer (Pαß) with various rod or cone Pγ variants and analyzed PDE6 activation upon addition of the activated transducin α-subunit (Gtα*-GTPγS). This analysis revealed a rod-specific Pγ motif (amino acids 9-18) that reduces the ability of Gtα*-GTPγS to activate the reconstituted PDE6. In cone Pγ, Asn-13 and Gln-14 significantly enhanced Gtα*-GTPγS activation of cone Pγ truncation variants. Moreover, we observed that the first four amino acids of either rod or cone Pγ contribute to Gtα*-GTPγS-mediated activation of PDE6. We conclude that physiological differences between rod and cone photoreceptor light responsiveness can be partially ascribed to ancient, highly conserved amino acid differences in the N-terminal regions of Pγ isoforms, demonstrating for the first time a functional role for this region of Pγ in the differential activation of rod and cone PDE6 by transducin.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/química , Guanosina 5'-O-(3-Tiotrifosfato)/química , Células Fotorreceptoras Retinianas Conos/enzimología , Células Fotorreceptoras Retinianas Bastones/enzimología , Animales , Catálisis , Bovinos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo
2.
Bioorg Med Chem Lett ; 30(18): 127443, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32730942

RESUMEN

Positive allosteric modulators (PAMs) of GABAB receptor represent an interesting alternative to receptor agonists such as baclofen, as they act on the receptor in a more physiological way and thus are devoid of the side effects typically exerted by the agonists. Based on our interest in the identification of new GABAB receptor PAMs, we followed a merging approach to design new chemotypes starting from selected active compounds, such as GS39783, rac-BHFF, and BHF177, and we ended up with the synthesis of four different classes of compounds. The new compounds were tested alone or in the presence of 10 µM GABA using [35S]GTPγS binding assay to assess their functionality at the receptor. Unexpectedly, a number of them significantly inhibited GABA-stimulated GTPγS binding thus revealing a functional switch with respect to the prototype molecules. Further studies on selected compounds will clarify if they act as negative modulators of the receptor or, instead, as antagonists at the orthosteric binding site.


Asunto(s)
Baclofeno/síntesis química , Agonistas de Receptores GABA-B/síntesis química , Guanosina 5'-O-(3-Tiotrifosfato)/química , Receptores de GABA-B/metabolismo , Regulación Alostérica , Baclofeno/metabolismo , Benzofuranos/farmacología , Sitios de Unión , Ciclización , Ciclopentanos/farmacología , Evaluación Preclínica de Medicamentos , Moduladores del GABA/metabolismo , Agonistas de Receptores GABA-B/metabolismo , Humanos , Norbornanos/farmacología , Unión Proteica , Pirimidinas/farmacología , Relación Estructura-Actividad
3.
J Struct Biol ; 202(1): 13-24, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29196061

RESUMEN

Rho family GTPases regulate a wide range of cellular processes. This includes cellular dynamics where three subfamilies, Rho, Rac, and Cdc42, are known to regulate cell shape and migration though coordinate action. Activation of Rho proteins largely depends on Rho Guanine nucleotide Exchange Factors (RhoGEFs) through a catalytic Dbl homology (DH) domain linked to a pleckstrin homology (PH) domain that subserves various functions. The PH domains from Lbc RhoGEFs, which specifically activate RhoA, have been shown to bind to activated RhoA. Here, p190RhoGEF is shown to also bind Rac1·GTP. Crystal structures reveal that activated Rac1 and RhoA use their effector-binding surfaces to associate with the same hydrophobic surface on the PH domain. Both activated RhoA and Rac1 can stimulate exchange of nucleotide on RhoA by localization of p190RhoGEF to its substrate, RhoA·GDP, in vitro. The binding of activated RhoA provides a mechanism for positive feedback regulation as previously proposed for the family of Lbc RhoGEFs. In contrast, the novel interaction between activated Rac1 and p190RhoGEF reveals a potential mechanism for cross-talk regulation where Rac can directly effect stimulation of RhoA. The greater capacity of Rac1 to stimulate p190RhoGEF among the Lbc RhoGEFs suggests functional specialization.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Dominios Proteicos , Proteínas de Unión al GTP rac/química , Proteína de Unión al GTP rhoA/química , Secuencia de Aminoácidos , Retroalimentación Fisiológica , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Modelos Moleculares , Unión Proteica , Homología de Secuencia de Aminoácido , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
4.
Biochemistry ; 56(1): 271-280, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27936598

RESUMEN

G proteins are part of the G-protein-coupled receptor (GPCR) signal transduction cascade in which they transfer a signal from the membrane-embedded GPCR to other proteins in the cell. In the case of the inhibitory G-protein heterotrimer, permanent N-terminal myristoylation can transiently localize the Gαi subunit at the membrane as well as crucially influence Gαi's function in the GTP-bound conformation. The attachment of lipids to proteins is known to be essential for membrane trafficking; however, our results suggest that lipidation is also important for protein-protein interactions during signal transduction. Here we investigate the effect of myristoylation on the structure and dynamics of soluble Gαi1 and its possible implication for signal transduction. A 2 µs classical molecular dynamics simulation of a myristoylated Gαi1-GTP complex suggests that the myristoyl-induced conformational changes of the switch II and alpha helical domains create new possibilities for protein-protein interactions and emphasize the importance of permanent lipid attachment for the conformation and functional tunability of signaling proteins.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Guanosina Trifosfato/química , Ácido Mirístico/química , Conformación Proteica , Factor 1 de Ribosilacion-ADP/química , Factor 1 de Ribosilacion-ADP/genética , Factor 1 de Ribosilacion-ADP/metabolismo , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Conformación Molecular , Simulación de Dinámica Molecular , Ácido Mirístico/metabolismo , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Ratas , Homología de Secuencia de Aminoácido , Electricidad Estática
5.
Proteins ; 84(4): 532-43, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26994549

RESUMEN

Despite the important role of the carboxyl-terminus (Ct) of the activated brain cannabinoid receptor one (CB1) in the regulation of G protein signaling, a structural understanding of interactions with G proteins is lacking. This is largely due to the highly flexible nature of the CB1 Ct that dynamically adapts its conformation to the presence of G proteins. In the present study, we explored how the CB1 Ct can interact with the G protein by building on our prior modeling of the CB1-Gi complex (Shim, Ahn, and Kendall, The Journal of Biological Chemistry 2013;288:32449-32465) to incorporate a complete CB1 Ct (Glu416(Ct)-Leu472(Ct)). Based on the structural constraints from NMR studies, we employed ROSETTA to predict tertiary folds, ZDOCK to predict docking orientation, and molecular dynamics (MD) simulations to obtain two distinct plausible models of CB1 Ct in the CB1-Gi complex. The resulting models were consistent with the NMR-determined helical structure (H9) in the middle region of the CB1 Ct. The CB1 Ct directly interacted with both Gα and Gß and stabilized the receptor at the Gi interface. The results of site-directed mutagenesis studies of Glu416(Ct), Asp423(Ct), Asp428(Ct), and Arg444(Ct) of CB1 Ct suggested that the CB1 Ct can influence receptor-G protein coupling by stabilizing the receptor at the Gi interface. This research provided, for the first time, models of the CB1 Ct in contact with the G protein.


Asunto(s)
Proteínas de Unión al GTP/química , Subunidades de Proteína/química , Receptor Cannabinoide CB1/química , Secuencia de Aminoácidos , Analgésicos/química , Sitios de Unión , Membrana Celular/química , Membrana Celular/metabolismo , Ciclohexanoles/química , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Expresión Génica , Guanosina 5'-O-(3-Tiotrifosfato)/química , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
6.
J Recept Signal Transduct Res ; 36(2): 158-66, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26466637

RESUMEN

CONTEXT: The appearance of "bell"- (or "inverted U"-) shaped agonist concentration-response curves (CRCs) in in vitro pharmacological experiments is a frequently observed but poorly communicated phenomenon. In the context of G protein coupled receptor research, it is commonly attributed to the recruitment of secondary targets or to desensitization or feedback processes, but the concrete background of these observations often remains intriguing. OBJECTIVE: Here, we addressed the subject of bell-shaped agonist CRCs at the µ opioid receptor (µOR) by testing the impact of experimental conditions favoring G protein coupling. METHODS: G protein activation by recombinant human µORs heterologously expressed in CHO cells was assessed in [(35)S]GTPγS binding assays using the opioid ligands DAMGO, morphine, fentanyl and naloxone. Experimental conditions were varied by changing the NaCl (10-300 mM) and the GDP concentration (0.3-30 µM). RESULTS: Both the sodium and the GDP concentration were inversely related to G protein coupling, as evident by an increase in basal [(35)S]GTPγS incorporation at low sodium and low GDP levels and by the concomitant appearance of the partial agonist activity of the µOR antagonist, naloxone. Bell-shaped CRCs were observed for the efficacious agonists DAMGO, fentanyl and morphine, and this phenomenon was promoted by low sodium as well as by low GDP concentrations. CONCLUSION: µOR agonist CRCs show a non-monotonic behavior with a decline of maximal stimulation under conditions of strong receptor-G protein coupling, and this behavior is visible at the level of G protein activation itself.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Receptores Opioides mu/metabolismo , Animales , Cricetinae , Cricetulus , Encefalina Ala(2)-MeFe(4)-Gli(5)/química , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Fentanilo/química , Fentanilo/farmacología , Proteínas de Unión al GTP/química , Regulación de la Expresión Génica/efectos de los fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina Difosfato/química , Humanos , Morfina/química , Morfina/farmacología , Naloxona/química , Naloxona/farmacología , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Sodio/química
7.
J Biol Chem ; 289(16): 11331-11341, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24596087

RESUMEN

Receptor-mediated activation of the Gα subunit of heterotrimeric G proteins requires allosteric communication between the receptor binding site and the guanine nucleotide binding site, which are separated by >30 Å. Structural changes in the allosteric network connecting these sites are predicted to be transient in the wild-type Gα subunit, making studies of these connections challenging. In the current work, site-directed mutants that alter the energy barriers between the activation states are used as tools to better understand the transient features of allosteric signaling in the Gα subunit. The observed differences in relative receptor affinity for intact Gαi1 subunits versus C-terminal Gαi1 peptides harboring the K345L mutation are consistent with this mutation modulating the allosteric network in the protein subunit. Measurement of nucleotide exchange rates, affinity for metarhodopsin II, and thermostability suggest that the K345L Gαi1 variant has reduced stability in both the GDP-bound and nucleotide-free states as compared with wild type but similar stability in the GTPγS-bound state. High resolution x-ray crystal structures reveal conformational changes accompanying the destabilization of the GDP-bound state. Of these, the conformation for Switch I was stabilized by an ionic interaction with the phosphate binding loop. Further site-directed mutagenesis suggests that this interaction between Switch I and the phosphate binding loop is important for receptor-mediated nucleotide exchange in the wild-type Gαi1 subunit.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina Difosfato/química , Regulación Alostérica/fisiología , Sustitución de Aminoácidos , Animales , Cristalografía por Rayos X , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/genética , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Difosfato/genética , Guanosina Difosfato/metabolismo , Mutación Missense , Estructura Secundaria de Proteína , Ratas
8.
Proc Natl Acad Sci U S A ; 109(19): 7275-9, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22529365

RESUMEN

Proteins with similar crystal structures can have dissimilar rates of substrate binding and catalysis. Here we used molecular dynamics simulations and biochemical analysis to determine the role of intradomain and interdomain motions in conferring distinct activation rates to two Gα proteins, Gα(i1) and GPA1. Despite high structural similarity, GPA1 can activate itself without a receptor, whereas Gα(i1) cannot. We found that motions in these proteins vary greatly in type and frequency. Whereas motion is greatest in the Ras domain of Gα(i1), it is greatest in helices αA and αB from the helical domain of GPA1. Using protein chimeras, we show that helix αA from GPA1 is sufficient to confer rapid activation to Gα(i1). Gα(i1) has less intradomain motion than GPA1 and instead displays interdomain displacement resembling that observed in a receptor-heterotrimer crystal complex. Thus, structurally similar proteins can have distinct atomic motions that confer distinct activation mechanisms.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica
9.
Proc Natl Acad Sci U S A ; 109(34): 13596-601, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869700

RESUMEN

Physical biochemical techniques are used to establish the structure, subunit stoichiometry, and assembly pathway of the primosome complex of the bacteriophage T4 DNA replication system. Analytical ultracentrifugation and fluorescence anisotropy methods show that the functional T4 primosome consists of six gp41 helicase subunits that assemble into a hexagon, driven by the binding of six NTPs (or six nonhydrolyzable GTPγS analogues) that are located at and stabilize the intersubunit interfaces, together with a single tightly bound gp61 primase subunit. Assembling the components of the primosome onto a model DNA replication fork is a multistep process, but equilibrium cannot be reached along all mixing pathways. Producing a functional complex requires that the helicase hexamer be assembled in the presence of the DNA replication fork construct prior to the addition of the primase to avoid the formation of metastable DNA-protein aggregates. The gp41 helicase hexamer binds weakly to fork DNA in the absence of primase, but forms a much more stable primosome complex that expresses full and functional helicase (and primase) activities when bound to a gp61 primase subunit at a helicase:primase subunit ratio of 61. The presence of additional primase subunits does not change the molecular mass or helicase activity of the primosome, but significantly inhibits its primase activity. We develop both an assembly pathway and a minimal mechanistic model for the structure and function of the T4 primosome that are likely to be relevant to the assembly and function of the replication primosome subassemblies of higher organisms as well.


Asunto(s)
Bacteriófago T4/química , ADN Helicasas/química , ADN Primasa/química , Anisotropía , Proteínas Bacterianas/química , ADN/química , Replicación del ADN , Relación Dosis-Respuesta a Droga , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina Trifosfato/química , Hidrólisis , Sustancias Macromoleculares/química , Microscopía Fluorescente/métodos , Modelos Químicos , Desnaturalización Proteica , Termodinámica , Ultracentrifugación/métodos , Proteínas del Envoltorio Viral/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(38): 16086-91, 2011 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-21914848

RESUMEN

The active-state complex between an agonist-bound receptor and a guanine nucleotide-free G protein represents the fundamental signaling assembly for the majority of hormone and neurotransmitter signaling. We applied single-particle electron microscopy (EM) analysis to examine the architecture of agonist-occupied ß(2)-adrenoceptor (ß(2)AR) in complex with the heterotrimeric G protein Gs (Gαsßγ). EM 2D averages and 3D reconstructions of the detergent-solubilized complex reveal an overall architecture that is in very good agreement with the crystal structure of the active-state ternary complex. Strikingly however, the α-helical domain of Gαs appears highly flexible in the absence of nucleotide. In contrast, the presence of the pyrophosphate mimic foscarnet (phosphonoformate), and also the presence of GDP, favor the stabilization of the α-helical domain on the Ras-like domain of Gαs. Molecular modeling of the α-helical domain in the 3D EM maps suggests that in its stabilized form it assumes a conformation reminiscent to the one observed in the crystal structure of Gαs-GTPγS. These data argue that the α-helical domain undergoes a nucleotide-dependent transition from a flexible to a conformationally stabilized state.


Asunto(s)
Cristalografía por Rayos X , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores Adrenérgicos beta 2/química , Animales , Cristalización , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/ultraestructura , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Microscopía Electrónica , Modelos Moleculares , Unión Proteica , Conformación Proteica , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 2/ultraestructura
11.
Biochemistry ; 52(15): 2518-25, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23560983

RESUMEN

RtcB is an atypical RNA ligase that joins either 2',3'-cyclic phosphate or 3'-phosphate termini to 5'-hydroxyl termini. In contrast to typical RNA ligases, which rely on ATP and Mg(II), catalysis by RtcB is dependent on GTP and Mn(II) with ligation proceeding through a covalent RtcB-histidine-GMP intermediate. Here, we present three structures of Pyrococcus horikoshii RtcB complexes that capture snapshots along the entire guanylylation pathway. These structures show that prior to binding GTP, a single manganese ion (Mn1) is bound to RtcB. To capture the step immediately preceding RtcB guanylylation, we determined a structure of RtcB in complex with Mn(II) and the unreactive GTP analogue guanosine 5'-(α-thio)triphosphate (GTPαS). This structure shows that Mn1 is poised to stabilize the pentavalent transition state of guanylylation while a second manganese ion (Mn2) is coordinated to a nonbridging oxygen of the γ-phosphoryl group. The pyrophosphate leaving group of GTPαS is oriented apically to His404 with the ε-nitrogen poised for in-line attack on the α-phosphorus atom. The structure of RtcB in complex with GTPαS also reveals the network of hydrogen bonds that recognize GTP and illuminates the significant conformational changes that accompany the binding of this cofactor. Finally, a structure of the enzymic histidine-GMP intermediate depicts the end of the guanylylation pathway. The ensuing molecular description of the RtcB guanylylation pathway shows that RtcB and classical ATP- and Mg(II)-dependent nucleic acid ligases have converged upon a similar two-metal mechanism for formation of the nucleotidylated enzyme intermediate.


Asunto(s)
Histidina/metabolismo , Pyrococcus horikoshii/enzimología , ARN Ligasa (ATP)/química , ARN Ligasa (ATP)/metabolismo , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Cristalografía por Rayos X , Guanosina 5'-O-(3-Tiotrifosfato)/análogos & derivados , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Trifosfato/metabolismo , Enlace de Hidrógeno , Manganeso/metabolismo , Modelos Moleculares , Conformación Proteica
12.
J Biol Chem ; 287(6): 3630-41, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22117076

RESUMEN

Despite its central role in signaling and the potential therapeutic applications of inverse agonists, the molecular mechanisms underlying G protein-coupled receptor (GPCR) constitutive activity remain largely to be explored. In this context, ghrelin receptor GHS-R1a is a peculiar receptor in the sense that it displays a strikingly high, physiologically relevant, constitutive activity. To identify the molecular mechanisms responsible for this high constitutive activity, we have reconstituted a purified GHS-R1a monomer in a lipid disc. Using this reconstituted system, we show that the isolated ghrelin receptor per se activates G(q) in the absence of agonist, as assessed through guanosine 5'-O-(thiotriphosphate) binding experiments. The measured constitutive activity is similar in its extent to that observed in heterologous systems and in vivo. This is the first direct evidence for the high constitutive activity of the ghrelin receptor being an intrinsic property of the protein rather than the result of influence of its cellular environment. Moreover, we show that the isolated receptor in lipid discs recruits arrestin-2 in an agonist-dependent manner, whereas it interacts with µ-AP2 in the absence of ligand or in the presence of ghrelin. Of importance, these differences are linked to ligand-specific GHS-R1a conformations, as assessed by intrinsic fluorescence measurements. The distinct ligand requirements for the interaction of purified GHS-R1a with arrestin and AP2 provide a new rationale to the differences in basal and agonist-induced internalization observed in cells.


Asunto(s)
Lípidos/química , Membranas Artificiales , Receptores de Ghrelina/química , Animales , Arrestinas/química , Arrestinas/genética , Arrestinas/metabolismo , Activación Enzimática , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Estructura Terciaria de Proteína , Receptores de Ghrelina/genética , Receptores de Ghrelina/metabolismo , Sepia
13.
J Chem Inf Model ; 53(12): 3202-11, 2013 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-24245825

RESUMEN

The serotonin receptor subtype 1A (5-HT(1A)R) has been implicated in several neurological conditions, and potent 5-HT(1A)R agonists have therapeutic potential for the treatment of depression, anxiety, schizophrenia, and Parkinson's disease. In the present study, a homology model of 5-HT(1A)R was built based on the latest released high-resolution crystal structure of the ß2AR in its active state (PDB: 3SN6). A dynamic pharmacophore model, which takes the receptor flexibility into account, was constructed, validated, and applied to our dynamic pharmacophore-based virtual screening approach with the aim to identify potential 5-5-HT(1A)R agonists. The obtained hits were subjected to 55-HT(1A)R binding and functional assays, and 10 compounds with medium or high K(i) and EC50 values were identified. Among them, FW01 (K(i) = 51.9 nM, EC50 = 7 nM) was evaluated as the strongest agonist for 5-HT(1A)R. The active 5-HT(1A)R model and dynamic pharmacophore model obtained from this study can be used for future discovery and design of novel 5-HT(1A)R agonists. Also, by integrating all computational and available experimental data, a stepwise 5-HT(1A)R signal transduction model induced by agonist FW01 was proposed.


Asunto(s)
Indoles/química , Simulación de Dinámica Molecular , Piperazinas/química , Receptor de Serotonina 5-HT1A/química , Agonistas del Receptor de Serotonina 5-HT1/química , Bibliotecas de Moléculas Pequeñas/química , Interfaz Usuario-Computador , Dominio Catalítico , Bases de Datos de Proteínas , Descubrimiento de Drogas , Guanosina 5'-O-(3-Tiotrifosfato)/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Receptores Adrenérgicos beta 2/química , Homología Estructural de Proteína , Relación Estructura-Actividad
14.
Bioorg Med Chem ; 21(11): 3164-74, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23623679

RESUMEN

The goal of the present study was to explore, in our previously developed hybrid template, the effect of introduction of additional heterocyclic rings (mimicking catechol hydroxyl groups as bioisosteric replacement) on selectivity and affinity for the D3 versus D2 receptor. In addition, we wanted to explore the effect of derivatization of functional groups of the agonist binding moiety in compounds developed by us earlier from the hybrid template. Binding affinity (K(i)) of the new compounds was measured with tritiated spiperone as the radioligand and HEK-293 cells expressing either D2 or D3 receptors. Functional activity of selected compounds was assessed in the GTPγS binding assay. In the imidazole series, compound 10a exhibited the highest D3 affinity whereas the indole derivative 13 exhibited similar high D3 affinity. Functionalization of the amino group in agonist (+)-9d with different sulfonamides derivatives improved the D3 affinity significantly with (+)-14f exhibiting the highest affinity. However, functionalization of the hydroxyl and amino groups of 15 and (+)-9d, known agonist and partial agonist, to sulfonate ester and amide in general modulated the affinity. In both cases loss of agonist potency resulted from such derivatization.


Asunto(s)
Agonistas de Dopamina/síntesis química , Naftalenos/síntesis química , Piperazinas/síntesis química , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D3/agonistas , Animales , Células CHO , Cricetulus , Agonistas de Dopamina/química , Guanosina 5'-O-(3-Tiotrifosfato)/química , Células HEK293 , Humanos , Cinética , Naftalenos/química , Piperazinas/química , Unión Proteica , Ensayo de Unión Radioligante , Receptores de Dopamina D2/química , Receptores de Dopamina D3/química , Espiperona/química , Relación Estructura-Actividad
15.
J Struct Funct Genomics ; 13(4): 241-5, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23001747

RESUMEN

RhoA, a member of the Rho sub-family of small GTPases, plays a significant signaling role in cell morphogenesis, migration, neuronal development, cell division and adhesion. So far, 4 structures of RhoA:GDP/GTP analogs and 14 structures of RhoA in complex with other proteins have been reported. All RhoA:GDP/GTP analog complexes have been crystallized in primitive lattices and RhoA is monomeric. This is the first time a RhoA:GTP analog complex has been crystallized as a dimer in a centered lattice. The present structure reveals structural differences in the switch-I (residues 28-42) and switch-II (residues 61-66) regions, which play important roles in interactions with downstream targets to transduce signals, when compared to the previously reported structures.


Asunto(s)
Cristalografía por Rayos X/métodos , Guanosina 5'-O-(3-Tiotrifosfato)/química , Complejos Multiproteicos/química , Proteínas de Unión al GTP rho/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Activación Enzimática , GTP Fosfohidrolasas/química , Guanosina Difosfato/química , Ratones , Datos de Secuencia Molecular , Complejos Multiproteicos/genética , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Alineación de Secuencia , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/aislamiento & purificación , Proteína de Unión al GTP rhoA
16.
J Biol Chem ; 285(12): 9008-17, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20097748

RESUMEN

The protein G18 (also known as AGS4 or GPSM3) contains three conserved GoLoco/GPR domains in its central and C-terminal regions that bind to inactive Galpha(i), whereas the N-terminal region has not been previously characterized. We investigated whether this domain might itself regulate G protein activity by assessing the abilities of G18 and mutants thereof to modulate the nucleotide binding and hydrolytic properties of Galpha(i1) and Galpha(o). Surprisingly, in the presence of fluoroaluminate (AlF(4)(-)) both G proteins bound strongly to full-length G18 (G18wt) and to its isolated N-terminal domain (G18DeltaC) but not to its GoLoco region (DeltaNG18). Thus, it appears that its N-terminal domain promotes G18 binding to fluoroaluminate-activated Galpha(i/o). Neither G18wt nor any G18 mutant affected the GTPase activity of Galpha(i1) or Galpha(o). In contrast, complex effects were noted with respect to nucleotide binding. As inferred by the binding of [(35)S]GTPgammaS (guanosine 5'-O-[gamma-thio]triphosphate) to Galpha(i1), the isolated GoLoco region as expected acted as a guanine nucleotide dissociation inhibitor, whereas the N-terminal region exhibited a previously unknown guanine nucleotide exchange factor effect on this G protein. On the other hand, the N terminus inhibited [(35)S]GTPgammaS binding to Galpha(o), albeit to a lesser extent than the effect of the GoLoco region on Galpha(i1). Taken together, our results identify the N-terminal region of G18 as a novel G protein-interacting domain that may have distinct regulatory effects within the G(i/o) subfamily, and thus, it could potentially play a role in differentiating signals between these related G proteins.


Asunto(s)
Proteínas de Unión al GTP/química , Inhibidores de Disociación de Guanina Nucleótido/química , Prolina/química , Secuencia de Aminoácidos , Animales , Epítopos/química , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina Trifosfato/química , Hidrólisis , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Factores de Tiempo , Distribución Tisular
17.
Methods Mol Biol ; 2201: 109-116, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32975793

RESUMEN

The opioid receptors have been an interesting target for the drug industry for decades. These receptors were pharmacologically characterized in the 1970s and several drugs and peptides have emerged over the years. In 2012, the crystal structures were also demonstrated, with new data on the receptor sites, and thus new possibilities will appear. The role of opioids in the brain has attracted considerable interest in several diseases, especially pain and drug dependence. The opioid receptors are G-protein-coupled receptors (GPCR ) that are Gi coupled which make them suitable for studying the receptor functionality. The [35S]GTP γS autoradiography assay is a good option that has the benefit of generating both anatomical and functional data in the area of interest. It is based on the first step of the signaling mechanism of GPCRs. When a ligand binds to the receptor GTP will replace GDP on the a-subunit of the G-protein, leading to a dissociation of the ßγ-subunit. These subunits will start a cascade of second messengers and subsequently a physiological response.


Asunto(s)
Autorradiografía/métodos , Guanosina 5'-O-(3-Tiotrifosfato)/química , Receptores Opioides mu/metabolismo , Analgésicos Opioides/metabolismo , Animales , Proteínas de Unión al GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Receptores Opioides/metabolismo , Transducción de Señal , Radioisótopos de Azufre/química , Radioisótopos de Azufre/metabolismo
18.
FEBS J ; 288(2): 582-599, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32352209

RESUMEN

Human guanylate-binding protein 1 (hGBP-1) shows a dimer-induced acceleration of the GTPase activity yielding GDP as well as GMP. While the head-to-head dimerization of the large GTPase (LG) domain is well understood, the role of the rest of the protein, particularly of the GTPase effector domain (GED), in dimerization and GTP hydrolysis is still obscure. In this study, with truncations and point mutations on hGBP-1 and by means of biochemical and biophysical methods, we demonstrate that the intramolecular communication between the LG domain and the GED (LG:GED) is crucial for protein dimerization and dimer-stimulated GTP hydrolysis. In the course of GTP binding and γ-phosphate cleavage, conformational changes within hGBP-1 are controlled by a chain of amino acids ranging from the region near the nucleotide-binding pocket to the distant LG:GED interface and lead to the release of the GED from the LG domain. This opening of the structure allows the protein to form GED:GED contacts within the dimer, in addition to the established LG:LG interface. After releasing the cleaved γ-phosphate, the dimer either dissociates yielding GDP as the final product or it stays dimeric to further cleave the ß-phosphate yielding GMP. The second phosphate cleavage step, that is, the formation of GMP, is even more strongly coupled to structural changes and thus more sensitive to structural restraints imposed by the GED. Altogether, we depict a comprehensive mechanism of GTP hydrolysis catalyzed by hGBP-1, which provides a detailed molecular understanding of the enzymatic activity connected to large structural rearrangements of the protein. DATABASE: Structural data are available in RCSB Protein Data Bank under the accession numbers: 1F5N, 1DG3, 2B92.


Asunto(s)
Proteínas de Unión al GTP/química , Guanosina Difosfato/química , Guanosina Trifosfato/química , Dominios y Motivos de Interacción de Proteínas , Sitios de Unión , Biocatálisis , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Cinética , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
19.
Nat Struct Mol Biol ; 12(12): 1094-100, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16249794

RESUMEN

The exocyst is a large complex that is required for tethering vesicles at the final stages of the exocytic pathway in all eukaryotes. Here we present the structures of the Exo70p subunit of this complex and of the C-terminal domains of Exo84p, at 2.0-A and 2.85-A resolution, respectively. Exo70p forms a 160-A-long rod with a novel fold composed of contiguous alpha-helical bundles. The Exo84p C terminus also forms a long rod (80 A), which unexpectedly has the same fold as the Exo70p N terminus. Our structural results and our experimental observations concerning the interaction between Exo70p and other exocyst subunits or Rho3p GTPase are consistent with an architecture wherein exocyst subunits are composed of mostly helical modules strung together into long rods.


Asunto(s)
Exocitosis , Proteínas de la Membrana/química , Proteínas de Saccharomyces cerevisiae/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Cristalografía , Guanosina 5'-O-(3-Tiotrifosfato)/química , Datos de Secuencia Molecular , Pliegue de Proteína , Estructura Terciaria de Proteína , Vesículas Secretoras/química , Proteínas de Transporte Vesicular , Proteínas de Unión al GTP rho/química
20.
Science ; 278(5345): 1907-16, 1997 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-9417641

RESUMEN

The crystal structure of a soluble, catalytically active form of adenylyl cyclase in a complex with its stimulatory heterotrimeric G protein alpha subunit (Gsalpha) and forskolin was determined to a resolution of 2.3 angstroms. When P-site inhibitors were soaked into native crystals of the complex, the active site of adenylyl cyclase was located and structural elements important for substrate recognition and catalysis were identified. On the basis of these and other structures, a molecular mechanism is proposed for the activation of adenylyl cyclase by Gsalpha.


Asunto(s)
Adenilil Ciclasas/química , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Guanosina 5'-O-(3-Tiotrifosfato)/química , Adenosina Trifosfato/metabolismo , Inhibidores de Adenilato Ciclasa , Adenilil Ciclasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Colforsina/metabolismo , Cristalización , Cristalografía por Rayos X , Dimerización , Activación Enzimática , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Fosforilación , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA