Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
IUBMB Life ; 72(6): 1233-1242, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32271995

RESUMEN

C─H⋯O hydrogen bonds constitute a unique class of cohesive interactions. Their properties are similar to those of canonical H-bonds, although their energy is significantly lower, typically in the 0.5-2.5 kcal/mol range. Polarised C─H groups, such as those adjacent to electronegative groups, or within aromatic moieties, are particularly strong donors. C─H⋯O bonds are ubiquitous in nucleic acids and in proteins, notably stabilizing the ß-sheet secondary structure. They have also been observed in numerous protein-ligand interactions. Here, we analysed crystal structures, deposited in the Protein Data Bank, of complexes of FDA-approved protein kinase inhibitors with cognate kinases, to assess the possible role of C─Hinhibitor ⋯Oprotein hydrogen bonds. The conserved hinge motif of protein kinases with two solvent-exposed carbonyl groups and one exposed backbone amide, is well known to be involved in canonical H-bonding with inhibitors. We now find that in virtually all complexes where the inhibitor interacts with the hinge backbone, at least one of the hinge carbonyl groups accepts an H-bond from a C─H inhibitor group, which is either aromatic or adjacent to an electronegative group. These observations are important for design of hinge-binding scaffolds of novel kinase inhibitors for therapeutic use.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Enlace de Hidrógeno , Modelos Moleculares , Estereoisomerismo
2.
J Biol Chem ; 291(39): 20353-71, 2016 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-27481945

RESUMEN

RHO GTPase-activating proteins (RHOGAPs) are one of the major classes of regulators of the RHO-related protein family that are crucial in many cellular processes, motility, contractility, growth, differentiation, and development. Using database searches, we extracted 66 distinct human RHOGAPs, from which 57 have a common catalytic domain capable of terminating RHO protein signaling by stimulating the slow intrinsic GTP hydrolysis (GTPase) reaction. The specificity of the majority of the members of RHOGAP family is largely uncharacterized. Here, we comprehensively investigated the sequence-structure-function relationship between RHOGAPs and RHO proteins by combining our in vitro data with in silico data. The activity of 14 representatives of the RHOGAP family toward 12 RHO family proteins was determined in real time. We identified and structurally verified hot spots in the interface between RHOGAPs and RHO proteins as critical determinants for binding and catalysis. We have found that the RHOGAP domain itself is nonselective and in some cases rather inefficient under cell-free conditions. Thus, we propose that other domains of RHOGAPs confer substrate specificity and fine-tune their catalytic efficiency in cells.


Asunto(s)
Proteínas Activadoras de GTPasa/química , Proteínas de Unión al GTP rho/química , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Dominios Proteicos , Relación Estructura-Actividad , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
3.
EMBO J ; 32(7): 1023-35, 2013 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-23455152

RESUMEN

Dynactin is a protein complex required for the in vivo function of cytoplasmic dynein, a microtubule (MT)-based motor. Dynactin binds both dynein and MTs via its p150(Glued) subunit, but little is known about the 'pointed-end complex' that includes the protein subunits Arp11, p62 and the p27/p25 heterodimer. Here, we show that the p27/p25 heterodimer undergoes mitotic phosphorylation by cyclin-dependent kinase 1 (Cdk1) at a single site, p27 Thr186, to generate an anchoring site for polo-like kinase 1 (Plk1) at kinetochores. Removal of p27/p25 from dynactin results in reduced levels of Plk1 and its phosphorylated substrates at kinetochores in prometaphase, which correlates with aberrant kinetochore-MT interactions, improper chromosome alignment and abbreviated mitosis. To investigate the structural implications of p27 phosphorylation, we determined the structure of human p27. This revealed an unusual left-handed ß-helix domain, with the phosphorylation site located within a disordered, C-terminal segment. We conclude that dynactin plays a previously undescribed regulatory role in the spindle assembly checkpoint by recruiting Plk1 to kinetochores and facilitating phosphorylation of important downstream targets.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Bovinos , Proteínas de Ciclo Celular/genética , Línea Celular , Embrión de Pollo , Complejo Dinactina , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Microtúbulos/metabolismo , Fosforilación/fisiología , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Subunidades de Proteína/genética , Proteínas Proto-Oncogénicas/genética , Huso Acromático/genética , Huso Acromático/metabolismo , Quinasa Tipo Polo 1
4.
J Biol Chem ; 288(47): 34030-34040, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24106280

RESUMEN

Many agonists, acting through G-protein-coupled receptors and Gα subunits of the heterotrimeric G-proteins, induce contraction of smooth muscle through an increase of [Ca(2+)]i as well as activation of the RhoA/RhoA-activated kinase pathway that amplifies the contractile force, a phenomenon known as Ca(2+) sensitization. Gα12/13 subunits are known to activate the regulator of G-protein signaling-like family of guanine nucleotide exchange factors (RhoGEFs), which includes PDZ-RhoGEF (PRG) and leukemia-associated RhoGEF (LARG). However, their contributions to Ca(2+)-sensitized force are not well understood. Using permeabilized blood vessels from PRG(-/-) mice and a new method to silence LARG in organ-cultured blood vessels, we show that both RhoGEFs are activated by the physiologically and pathophysiologically important thromboxane A2 and endothelin-1 receptors. The co-activation is the result of direct and independent activation of both RhoGEFs as well as their co-recruitment due to heterodimerization. The isolated recombinant C-terminal domain of PRG, which is responsible for heterodimerization with LARG, strongly inhibited Ca(2+)-sensitized force. We used photolysis of caged phenylephrine, caged guanosine 5'-O-(thiotriphosphate) (GTPγS) in solution, and caged GTPγS or caged GTP loaded on the RhoA·RhoGDI complex to show that the recruitment and activation of RhoGEFs is the cause of a significant time lag between the initial Ca(2+) transient and phasic force components and the onset of Ca(2+)-sensitized force.


Asunto(s)
Calcio/metabolismo , Factores de Intercambio de Guanina Nucleótido/agonistas , Guanosina 5'-O-(3-Tiotrifosfato)/análogos & derivados , Fenilefrina/análogos & derivados , Factores de Intercambio de Guanina Nucleótido Rho/agonistas , Animales , Línea Celular , Silenciador del Gen/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Humanos , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Fenilefrina/farmacología , Multimerización de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína , Conejos , Ratas , Receptor de Endotelina A/genética , Receptor de Endotelina A/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/genética , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico/genética , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
5.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 9): 2420-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25195755

RESUMEN

Ebolavirus (EBOV) causes severe hemorrhagic fever with a mortality rate of up to 90%. EBOV is a member of the order Mononegavirales and, like other viruses in this taxonomic group, contains a negative-sense single-stranded (ss) RNA. The EBOV ssRNA encodes seven distinct proteins. One of them, the nucleoprotein (NP), is the most abundant viral protein in the infected cell and within the viral nucleocapsid. Like other EBOV proteins, NP is multifunctional. It is tightly associated with the viral genome and is essential for viral transcription, RNA replication, genome packaging and nucleocapsid assembly prior to membrane encapsulation. NP is unusual among the Mononegavirales in that it contains two distinct regions, or putative domains, the C-terminal of which shows no homology to any known proteins and is purported to be a hub for protein-protein interactions within the nucleocapsid. The atomic structure of NP remains unknown. Here, the boundaries of the N- and C-terminal domains of NP from Zaire EBOV are defined, it is shown that they can be expressed as highly stable recombinant proteins in Escherichia coli, and the atomic structure of the C-terminal domain (residues 641-739) derived from analysis of two distinct crystal forms at 1.98 and 1.75 Šresolution is described. The structure reveals a novel tertiary fold that is distantly reminiscent of the ß-grasp architecture.


Asunto(s)
Ebolavirus/química , Nucleoproteínas/química , Proteínas Virales/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Ebolavirus/fisiología , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Pliegue de Proteína , Homología de Secuencia de Aminoácido
6.
Acta Crystallogr D Struct Biol ; 80(Pt 7): 551-562, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38941144

RESUMEN

Tryptophan is the most prominent amino acid found in proteins, with multiple functional roles. Its side chain is made up of the hydrophobic indole moiety, with two groups that act as donors in hydrogen bonds: the Nϵ-H group, which is a potent donor in canonical hydrogen bonds, and a polarized Cδ1-H group, which is capable of forming weaker, noncanonical hydrogen bonds. Due to adjacent electron-withdrawing moieties, C-H...O hydrogen bonds are ubiquitous in macromolecules, albeit contingent on the polarization of the donor C-H group. Consequently, Cα-H groups (adjacent to the carbonyl and amino groups of flanking peptide bonds), as well as the Cϵ1-H and Cδ2-H groups of histidines (adjacent to imidazole N atoms), are known to serve as donors in hydrogen bonds, for example stabilizing parallel and antiparallel ß-sheets. However, the nature and the functional role of interactions involving the Cδ1-H group of the indole ring of tryptophan are not well characterized. Here, data mining of high-resolution (r ≤ 1.5 Å) crystal structures from the Protein Data Bank was performed and ubiquitous close contacts between the Cδ1-H groups of tryptophan and a range of electronegative acceptors were identified, specifically main-chain carbonyl O atoms immediately upstream and downstream in the polypeptide chain. The stereochemical analysis shows that most of the interactions bear all of the hallmarks of proper hydrogen bonds. At the same time, their cohesive nature is confirmed by quantum-chemical calculations, which reveal interaction energies of 1.5-3.0 kcal mol-1, depending on the specific stereochemistry.


Asunto(s)
Enlace de Hidrógeno , Proteínas , Triptófano , Triptófano/química , Proteínas/química , Modelos Moleculares , Cristalografía por Rayos X/métodos , Conformación Proteica
7.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 2): 266-75, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23385462

RESUMEN

Members of the RSK family of kinases constitute attractive targets for drug design, but a lack of structural information regarding the mechanism of selective inhibitors impedes progress in this field. The crystal structure of the N-terminal kinase domain (residues 45-346) of mouse RSK2, or RSK2(NTKD), has recently been described in complex with one of only two known selective inhibitors, a rare naturally occurring flavonol glycoside, kaempferol 3-O-(3'',4''-di-O-acetyl-α-L-rhamnopyranoside), known as SL0101. Based on this structure, it was hypothesized that quercitrin (quercetin 3-O-α-L-rhamnopyranoside), a related but ubiquitous and inexpensive compound, might also act as an RSK inhibitor. Here, it is demonstrated that quercitrin binds to RSK2(NTKD) with a dissociation constant (K(d)) of 5.8 µM as determined by isothermal titration calorimetry, and a crystal structure of the binary complex at 1.8 Å resolution is reported. The crystal structure reveals a very similar mode of binding to that recently reported for SL0101. Closer inspection shows a number of small but significant differences that explain the slightly higher K(d) for quercitrin compared with SL0101. It is also shown that quercitrin can effectively substitute for SL0101 in a biological assay, in which it significantly suppresses the contractile force in rabbit pulmonary artery smooth muscle in response to Ca(2+).


Asunto(s)
Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Quercetina/análogos & derivados , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 90-kDa/química , Animales , Cristalografía por Rayos X , Ratones , Fragmentos de Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/metabolismo , Quercetina/metabolismo , Quercetina/farmacología , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Termodinámica
8.
Circ Res ; 109(9): 993-1002, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21885830

RESUMEN

RATIONALE: In normal and diseased vascular smooth muscle (SM), the RhoA pathway, which is activated by multiple agonists through G protein-coupled receptors (GPCRs), plays a central role in regulating basal tone and peripheral resistance. This occurs through inhibition of myosin light chain phosphatase, leading to increased phosphorylation of the myosin regulatory light chain. Although it is thought that specific agonists and GPCRs may couple to distinct RhoA guanine nucleotide exchange factors (GEFs), thus raising the possibility of selective targeting of specific GEFs for therapeutic use, this notion is largely unexplored for SM contraction. OBJECTIVE: We examine whether p63RhoGEF, known to couple specifically to Gα(q/11) in vitro, is functional in blood vessels as a mediator of RhoA activation and if it is selectively activated by Gα(q/11) coupled agonists. METHODS AND RESULTS: We find that p63RhoGEF is present across SM tissues and demonstrate that silencing of the endogenous p63RhoGEF in mouse portal vein inhibits contractile force induced by endothelin-1 to a greater extent than the predominantly Gα(12/13)-mediated thromboxane analog U46619. This is because endothelin-1 acts on Gα(q/11) as well as Gα(12/13). Introduction of the exogenous isolated pleckstrin-homology (PH) domain of p63RhoGEF (residues 331-580) into permeabilized rabbit portal vein inhibited Ca2+ sensitized force and activation of RhoA, when phenylephrine was used as an agonist. This reinforces the results based on endothelin-1, because phenylephrine is thought to act exclusively through Gα(q/11). CONCLUSION: We demonstrate that p63RhoGEF selectively couples Gα(q/11) but not Gα(12/13), to RhoA activation in blood vessels and cultured cells and thus mediates the physiologically important Ca2+ sensitization of force induced with Gα(q/11)-coupled agonists. Our results suggest that signaling through p63RhoGEF provides a novel mechanism for selective regulation of blood pressure.


Asunto(s)
Calcio/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Contracción Muscular/fisiología , Músculo Liso Vascular/fisiología , Transducción de Señal/fisiología , Animales , Células Cultivadas , Endotelina-1/farmacología , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/genética , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Fenilefrina/farmacología , Vena Porta/fisiología , Conejos , Ratas , Factores de Intercambio de Guanina Nucleótido Rho , Vasoconstrictores/farmacología , Proteína de Unión al GTP rhoA/fisiología
9.
Biochemistry ; 51(33): 6499-510, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22846040

RESUMEN

The p90 ribosomal S6 family of kinases (RSK) are potential drug targets, due to their involvement in cancer and other pathologies. There are currently only two known selective inhibitors of RSK, but the basis for selectivity is not known. One of these inhibitors is a naturally occurring kaempferol-α-L-diacetylrhamnoside, SL0101. Here, we report the crystal structure of the complex of the N-terminal kinase domain of the RSK2 isoform with SL0101 at 1.5 Å resolution. The refined atomic model reveals unprecedented structural reorganization of the protein moiety, as compared to the nucleotide-bound form. The entire N-lobe, the hinge region, and the αD-helix undergo dramatic conformational changes resulting in a rearrangement of the nucleotide binding site with concomitant formation of a highly hydrophobic pocket spatially suited to accommodate SL0101. These unexpected results will be invaluable in further optimization of the SL0101 scaffold as a promising lead for a novel class of kinase inhibitors.


Asunto(s)
Benzopiranos/farmacología , Monosacáridos/farmacología , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Sitios de Unión , Cristalización , Cristalografía por Rayos X , Manósidos/farmacología , Modelos Moleculares , Proantocianidinas/farmacología , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
10.
J Biol Chem ; 286(40): 35163-75, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21816819

RESUMEN

PDZRhoGEF (PRG) belongs to a small family of RhoA-specific nucleotide exchange factors that mediates signaling through select G-protein-coupled receptors via Gα(12/13) and activates RhoA by catalyzing the exchange of GDP to GTP. PRG is a multidomain protein composed of PDZ, regulators of G-protein signaling-like (RGSL), Dbl-homology (DH), and pleckstrin-homology (PH) domains. It is autoinhibited in cytosol and is believed to undergo a conformational rearrangement and translocation to the membrane for full activation, although the molecular details of the regulation mechanism are not clear. It has been shown recently that the main autoregulatory elements of PDZRhoGEF, the autoinhibitory "activation box" and the "GEF switch," which is required for full activation, are located directly upstream of the catalytic DH domain and its RhoA binding surface, emphasizing the functional role of the RGSL-DH linker. Here, using a combination of biophysical and biochemical methods, we show that the mechanism of PRG regulation is yet more complex and may involve an additional autoinhibitory element in the form of a molten globule region within the linker between RGSL and DH domains. We propose a novel, two-tier model of autoinhibition where the activation box and the molten globule region act synergistically to impair the ability of RhoA to bind to the catalytic DH-PH tandem. The molten globule region and the activation box become less ordered in the PRG-RhoA complex and dissociate from the RhoA-binding site, which may constitute a critical step leading to PRG activation.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Secuencia de Aminoácidos , Sitios de Unión , Dicroismo Circular , Humanos , Luz , Modelos Estadísticos , Datos de Secuencia Molecular , Conformación Proteica , Estructura Terciaria de Proteína , Factores de Intercambio de Guanina Nucleótido Rho , Dispersión de Radiación , Homología de Secuencia de Aminoácido , Rayos Ultravioleta , Rayos X , Proteína de Unión al GTP rhoA/química
11.
Biochemistry ; 48(36): 8664-71, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19653655

RESUMEN

The DUF1094 family contains over 100 bacterial proteins, all containing a conserved CXC motif, with unknown function. We solved the crystal structure of the Bacillus subtilis representative, the product of the yphP gene. The protein shows remarkable structural similarity to thioredoxins, with a canonical alphabetaalphabetaalphabetabetaalpha topology, despite low amino acid sequence identity to thioredoxin. The CXC motif is found in the loop immediately downstream of the first beta-strand, in a location equivalent to the CXXC motif of thioredoxins, with the first Cys occupying a position equivalent to the first Cys in canonical thioredoxin. The experimentally determined reduction potential of YphP is E degrees' = -130 mV, significantly higher than that of thioredoxin and consistent with disulfide isomerase activity. Functional assays confirmed that the protein displays a level of isomerase activity that might be biologically significant. We propose a mechanism by which the members of this family catalyze isomerization using the CXC catalytic site.


Asunto(s)
Bacillus subtilis/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Dominio Catalítico/fisiología , Proteína Disulfuro Isomerasas/química , Proteína Disulfuro Isomerasas/fisiología , Secuencias de Aminoácidos/genética , Secuencias de Aminoácidos/fisiología , Secuencia de Aminoácidos , Bacillus subtilis/genética , Proteínas Bacterianas/genética , Catálisis , Dominio Catalítico/genética , Secuencia Conservada/genética , Cristalografía por Rayos X , Cisteína/química , Cisteína/genética , Isomerismo , Datos de Secuencia Molecular , Familia de Multigenes , Mutagénesis Sitio-Dirigida , Proteína Disulfuro Isomerasas/genética , Alineación de Secuencia , Tiorredoxinas/química , Tiorredoxinas/metabolismo , Tiorredoxinas/fisiología
12.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 4): 356-65, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19307717

RESUMEN

The GntR superfamily of dimeric transcription factors, with more than 6200 members encoded in bacterial genomes, are characterized by N-terminal winged-helix DNA-binding domains and diverse C-terminal regulatory domains which provide a basis for the classification of the constituent families. The largest of these families, FadR, contains nearly 3000 proteins with all-alpha-helical regulatory domains classified into two related Pfam families: FadR_C and FCD. Only two crystal structures of FadR-family members, those of Escherichia coli FadR protein and LldR from Corynebacterium glutamicum, have been described to date in the literature. Here, the crystal structure of TM0439, a GntR regulator with an FCD domain found in the Thermotoga maritima genome, is described. The FCD domain is similar to that of the LldR regulator and contains a buried metal-binding site. Using atomic absorption spectroscopy and Trp fluorescence, it is shown that the recombinant protein contains bound Ni(2+) ions but that it is able to bind Zn(2+) with K(d) < 70 nM. It is concluded that Zn(2+) is the likely physiological metal and that it may perform either structural or regulatory roles or both. Finally, the TM0439 structure is compared with two other FadR-family structures recently deposited by structural genomics consortia. The results call for a revision in the classification of the FadR family of transcription factors.


Asunto(s)
Proteínas Bacterianas/química , Proteínas de Unión al ADN/química , Proteínas Represoras/química , Thermotoga maritima/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Secuencia Conservada , Cristalografía por Rayos X , Proteínas de Unión al ADN/clasificación , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dimerización , Regulación Bacteriana de la Expresión Génica , Modelos Moleculares , Familia de Multigenes , Níquel/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/clasificación , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Thermotoga maritima/genética , Zinc/metabolismo
13.
BMC Struct Biol ; 9: 36, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19460155

RESUMEN

BACKGROUND: The Dbl-family of guanine nucleotide exchange factors (GEFs) activate the cytosolic GTPases of the Rho family by enhancing the rate of exchange of GTP for GDP on the cognate GTPase. This catalytic activity resides in the DH (Dbl-homology) domain, but typically GEFs are multidomain proteins containing other modules. It is believed that GEFs are autoinhibited in the cytosol due to supramodular architecture, and become activated in diverse signaling pathways through conformational change and exposure of the DH domain, as the protein is translocated to the membrane. A small family of RhoA-specific GEFs, containing the RGSL (regulators of G-protein signaling-like) domain, act as effectors of select GPCRs via Galpha12/13, although the molecular mechanism by which this pathway operates is not known. These GEFs include p115, LARG and PDZRhoGEF (PRG). RESULTS: Here we show that the autoinhibition of PRG is caused largely by an interaction of a short negatively charged sequence motif, immediately upstream of the DH-domain and including residues Asp706, Glu708, Glu710 and Asp712, with a patch on the catalytic surface of the DH-domain including Arg867 and Arg868. In the absence of both PDZ and RGSL domains, the DH-PH tandem with additional 21 residues upstream, is 50% autoinhibited. However, within the full-length protein, the PDZ and/or RGSL domains significantly restore autoinhibition. CONCLUSION: Our results suggest a mechanism for autoinhibition of RGSL family of GEFs, in which the RGSL domain and a unique sequence motif upstream of the DH domain, act cooperatively to reduce the ability of the DH domain to bind the nucleotide free RhoA. The activation mechanism is likely to involve two independent steps, i.e. displacement of the RGSL domain and conformational change involving the autoinhibitory sequence motif containing several negatively charged residues.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/química , Modelos Químicos , Animales , Dominio Catalítico , Humanos , Ratones , Mutación , Células 3T3 NIH , Dominios PDZ , Factores de Intercambio de Guanina Nucleótido Rho
14.
BMC Biochem ; 10: 24, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19835610

RESUMEN

BACKGROUND: Helical repeat motifs are common among regulatory subunits for type-1 and type-2A protein Ser/Thr phosphatases. Yeast Sit4 is a distinctive type-2A phosphatase that has dedicated regulatory subunits named Sit4-Associated Proteins (SAPS). These subunits are conserved, and three human SAPS-related proteins are known to associate with PP6 phosphatase, the Sit4 human homologue. RESULTS: Here we show that endogenous SAPS subunit PP6R3 co-precipitates half of PP6 in cell extracts, and the SAPS region of PP6R3 is sufficient for binding PP6. The SAPS domain of recombinant GST-PP6R3 is relatively resistant to trypsin despite having many K and R residues, and the purified SAPS domain (residues 1-513) has a circular dichroic spectrum indicative of mostly alpha helical structure. We used sequence alignments and 3D-jury methods to develop alternative models for the SAPS domain, based on available structures of other helical repeat proteins. The models were used to select sites for charge-reversal substitutions in the SAPS domain of PP6R3 that were tested by co-precipitation of endogenous PP6c with FLAG-tagged PP6R3 from mammalian cells. Mutations that reduced binding with PP6 suggest that SAPS adopts a helical repeat similar to the structure of p115 golgin, but distinct from the PP2A-A subunit. These mutations did not cause perturbations in overall PP6R3 conformation, evidenced by no change in kinetics or preferential cleavage by chymotrypsin. CONCLUSION: The conserved SAPS domain in PP6R3 forms helical repeats similar to those in golgin p115 and negatively charged residues in interhelical loops are used to associate specifically with PP6. The results advance understanding of how distinctive helical repeat subunits uniquely distribute and differentially regulate closely related Ser/Thr phosphatases.


Asunto(s)
Secuencias de Aminoácidos/genética , Modelos Moleculares , Fosfoproteínas Fosfatasas/análisis , Proteína Fosfatasa 2/análisis , Secuencias Repetitivas de Aminoácido/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Línea Celular Tumoral , Biblioteca de Genes , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Unión Proteica , Ingeniería de Proteínas , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Estructura Secundaria de Proteína , Alineación de Secuencia , Eliminación de Secuencia , Transgenes/genética
15.
Structure ; 15(11): 1467-81, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17997972

RESUMEN

Ndel1 and Nde1 are homologous and evolutionarily conserved proteins, with critical roles in cell division, neuronal migration, and other physiological phenomena. These functions are dependent on their interactions with the retrograde microtubule motor dynein and with its regulator Lis1--a product of the causal gene for isolated lissencephaly sequence (ILS) and Miller-Dieker lissencephaly. The molecular basis of the interactions of Ndel1 and Nde1 with Lis1 is not known. Here, we present a crystallographic study of two fragments of the coiled-coil domain of Ndel1, one of which reveals contiguous high-quality electron density for residues 10-166, the longest such structure reported by X-ray diffraction at high resolution. Together with complementary solution studies, our structures reveal how the Ndel1 coiled coil forms a stable parallel homodimer and suggest mechanisms by which the Lis1-interacting domain can be regulated to maintain a conformation in which two supercoiled alpha helices cooperatively bind to a Lis1 homodimer.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/química , Proteínas Portadoras/química , Proteínas Asociadas a Microtúbulos/química , 1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Secuencia de Aminoácidos , Proteínas Portadoras/metabolismo , Dicroismo Circular , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/metabolismo , Cristalografía por Rayos X , Dimerización , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia
16.
Acta Biochim Pol ; 55(2): 269-80, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18542831

RESUMEN

PDZ domains are ubiquitous protein-protein interaction modules which bind short, usually carboxyterminal fragments of receptors, other integral or membrane-associated proteins, and occasionally cytosolic proteins. Their role in organizing multiprotein complexes at the cellular membrane is crucial for many signaling pathways, but the rules defining their binding specificity are still poorly understood and do not readily explain the observed diversity of their known binding partners. Two homologous RhoA-specific, multidomain nucleotide exchange factors PDZRhoGEF and LARG contain PDZ domains which show a particularly broad recognition profile, as suggested by the identification of five diverse biological targets. To investigate the molecular roots of this phenomenon, we constructed a phage display library of random carboxyterminal hexapeptides. Peptide variants corresponding to the sequences identified in library selection were synthesized and their affinities for both PDZ domains were measured and compared with those of peptides derived from sequences of natural partners. Based on the analysis of the binding sequences identified for PDZRhoGEF, we propose a sequence for an 'optimal' binding partner. Our results support the hypothesis that PDZ-peptide interactions may be best understood when one considers the sum of entropic and dynamic effects for each peptide as a whole entity, rather than preferences for specific residues at a given position.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/metabolismo , Dominios PDZ/fisiología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , ADN/genética , Factores de Intercambio de Guanina Nucleótido/genética , Ligandos , Mutagénesis Sitio-Dirigida , Dominios PDZ/genética , Biblioteca de Péptidos , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Proteína de Unión al GTP rhoA/química , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
17.
Sci Signal ; 11(554)2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30377223

RESUMEN

Smooth muscle contraction is triggered when Ca2+/calmodulin-dependent myosin light chain kinase (MLCK) phosphorylates the regulatory light chain of myosin (RLC20). However, blood vessels from Mlck-deficient mouse embryos retain the ability to contract, suggesting the existence of additional regulatory mechanisms. We showed that the p90 ribosomal S6 kinase 2 (RSK2) also phosphorylated RLC20 to promote smooth muscle contractility. Active, phosphorylated RSK2 was present in mouse resistance arteries under normal basal tone, and phosphorylation of RSK2 increased with myogenic vasoconstriction or agonist stimulation. Resistance arteries from Rsk2-deficient mice were dilated and showed reduced myogenic tone and RLC20 phosphorylation. RSK2 phosphorylated Ser19 in RLC in vitro. In addition, RSK2 phosphorylated an activating site in the Na+/H+ exchanger (NHE-1), resulting in cytosolic alkalinization and an increase in intracellular Ca2+ that promotes vasoconstriction. NHE-1 activity increased upon myogenic constriction, and the increase in intracellular pH was suppressed in Rsk2-deficient mice. In pressured arteries, RSK2-dependent activation of NHE-1 was associated with increased intracellular Ca2+ transients, which would be expected to increase MLCK activity, thereby contributing to basal tone and myogenic responses. Accordingly, Rsk2-deficient mice had lower blood pressure than normal littermates. Thus, RSK2 mediates a procontractile signaling pathway that contributes to the regulation of basal vascular tone, myogenic vasoconstriction, and blood pressure and may be a potential therapeutic target in smooth muscle contractility disorders.


Asunto(s)
Arterias/patología , Músculo Liso/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Miosinas del Músculo Liso/metabolismo , Intercambiador 1 de Sodio-Hidrógeno/metabolismo , Actinas/metabolismo , Animales , Aorta/citología , Calcio/metabolismo , Células Cultivadas , Femenino , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Noqueados , Desarrollo de Músculos , Miocitos del Músculo Liso/citología , Miografía , Quinasa de Cadena Ligera de Miosina/metabolismo , Fenilefrina/farmacología , Fosforilación , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Vasoconstricción
18.
Acta Crystallogr D Struct Biol ; 74(Pt 7): 681-689, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29968677

RESUMEN

The vast majority of platforms for the detection of viral or bacterial antigens rely on immunoassays, typically ELISA or sandwich ELISA, that are contingent on the availability of suitable monoclonal antibodies (mAbs). This is a major bottleneck, since the generation and production of mAbs is time-consuming and expensive. Synthetic antibody fragments (sFabs) generated by phage-display selection offer an alternative with many advantages over Fabs obtained from natural antibodies using hybridoma technology. Unlike mAbs, sFabs are generated using phage display, allowing selection for binding to specific strains or for pan-specificity, for identification of structural epitopes or unique protein conformations and even for complexes. Further, they can easily be produced in Escherichia coli in large quantities and engineered for purposes of detection technologies and other applications. Here, the use of phage-display selection to generate a pan-specific Fab (MJ20), based on a Herceptin Fab scaffold, with the ability to bind selectively and with high affinity to the C-terminal domains of the nucleoproteins (NPs) from all five known strains of the Ebola virus is reported. The high-resolution crystal structure of the complex of MJ20 with the antigen from the Bundibugyo strain of the Ebola virus reveals the basis for pan-specificity and illustrates how the phage-display technology can be used to manufacture suitable Fabs for use in diagnostic or therapeutic applications.


Asunto(s)
Complejo Antígeno-Anticuerpo/química , Ebolavirus/química , Fragmentos Fab de Inmunoglobulinas/química , Nucleoproteínas/química , Técnicas de Visualización de Superficie Celular , Cristalografía por Rayos X , Humanos , Fragmentos de Inmunoglobulinas/química , Biblioteca de Péptidos , Unión Proteica , Dominios Proteicos
19.
Acta Crystallogr D Struct Biol ; 73(Pt 9): 767-774, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28876240

RESUMEN

Two nonstructural proteins encoded by Zika virus strain MR766 RNA, a methyltransferase and a helicase, were crystallized and their structures were solved and refined at 2.10 and 2.01 Šresolution, respectively. The NS5 methyltransferase contains a bound S-adenosyl-L-methionine (SAM) co-substrate. The NS3 helicase is in the apo form. Comparison with published crystal structures of the helicase in the apo, nucleotide-bound and single-stranded RNA (ssRNA)-bound states suggests that binding of ssRNA to the helicase may occur through conformational selection rather than induced fit.


Asunto(s)
Metiltransferasas/química , ARN Helicasas/química , Proteínas no Estructurales Virales/química , Virus Zika/química , Virus Zika/enzimología , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Proteica , S-Adenosilmetionina/química , Uganda , Infección por el Virus Zika/virología
20.
Proteins ; 62(1): 144-51, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16287140

RESUMEN

The crystal structure of the product of the Bacillus subtilis ykuD gene was solved by the multiwavelength anomalous dispersion (MAD) method and refined using data to 2.0 A resolution. The ykuD protein is a representative of a distinctly prokaryotic and ubiquitous family found among both pathogenic and nonpathogenic Gram-positive and Gram-negative bacteria. The deduced amino acid sequence reveals the presence of an N-terminal LysM domain, which occurs among enzymes involved in cell wall metabolism, and a novel, putative catalytic domain with a highly conserved His/Cys-containing motif of hitherto unknown structure. As the wild-type protein did not crystallize, a double mutant was designed (Lys117Ala/Gln118Ala) to reduce excess surface conformational entropy. As expected, the structure of the LysM domain is similar to the NMR structure reported for an analogous domain from Escherichia coli murein transglycosylase MltD. The molecular model also shows that the 112-residue-long C-terminal domain has a novel tertiary fold consisting of a beta-sandwich with two mixed sheets, one containing five strands and the other, six strands. The two beta-sheets form a cradle capped by an alpha-helix. This domain contains a putative catalytic site with a tetrad of invariant His123, Gly124, Cys139, and Arg141. The stereochemistry of this active site shows similarities to peptidotransferases and sortases, and suggests that the enzymes of the ykuD family may play an important role in cell wall biology.


Asunto(s)
Bacillus subtilis/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Enzimas/química , Enzimas/metabolismo , Secuencia de Aminoácidos , Secuencia de Consenso , Cristalización , Dipéptidos/química , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Alineación de Secuencia , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA