Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Signal ; 16(795): eadd9539, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37490546

RESUMEN

Precise synapse formation is essential for normal functioning of the nervous system. Retinal photoreceptors establish selective contacts with bipolar cells, aligning the neurotransmitter release apparatus with postsynaptic signaling cascades. This involves transsynaptic assembly between the dystroglycan-dystrophin complex on the photoreceptor and the orphan receptor GPR179 on the bipolar cell, which is mediated by the extracellular matrix protein pikachurin (also known as EGFLAM). This complex plays a critical role in the synaptic organization of photoreceptors and signal transmission, and mutations affecting its components cause blinding disorders in humans. Here, we investigated the structural organization and molecular mechanisms by which pikachurin orchestrates transsynaptic assembly and solved structures of the human pikachurin domains by x-ray crystallography and of the GPR179-pikachurin complex by single-particle, cryo-electron microscopy. The structures reveal molecular recognition principles of pikachurin by the Cache domains of GPR179 and show how the interaction is involved in the transsynaptic alignment of the signaling machinery. Together, these data provide a structural basis for understanding the synaptic organization of photoreceptors and ocular pathology.


Asunto(s)
Proteínas de la Matriz Extracelular , Sinapsis , Humanos , Proteínas Portadoras/metabolismo , Microscopía por Crioelectrón , Proteínas de la Matriz Extracelular/metabolismo , Células Fotorreceptoras/metabolismo , Sinapsis/metabolismo
2.
Science ; 379(6639): 1352-1358, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36996198

RESUMEN

Glycine is a major neurotransmitter involved in several fundamental neuronal processes. The identity of the metabotropic receptor mediating slow neuromodulatory effects of glycine is unknown. We identified an orphan G protein-coupled receptor, GPR158, as a metabotropic glycine receptor (mGlyR). Glycine and a related modulator, taurine, directly bind to a Cache domain of GPR158, and this event inhibits the activity of the intracellular signaling complex regulator of G protein signaling 7-G protein ß5 (RGS7-Gß5), which is associated with the receptor. Glycine signals through mGlyR to inhibit production of the second messenger adenosine 3',5'-monophosphate. We further show that glycine, but not taurine, acts through mGlyR to regulate neuronal excitability in cortical neurons. These results identify a major neuromodulatory system involved in mediating metabotropic effects of glycine, with implications for understanding cognition and affective states.


Asunto(s)
Glicina , Receptores Acoplados a Proteínas G , Receptores de Glicina , Glicina/metabolismo , Proteínas de Unión al GTP/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glicina/química , Receptores de Glicina/genética , Receptores de Glicina/metabolismo , Transducción de Señal , Humanos , Células HEK293 , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Proteínas RGS/metabolismo , Dominios Proteicos
3.
Science ; 375(6576): 86-91, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34793198

RESUMEN

GPR158 is an orphan G protein­coupled receptor (GPCR) highly expressed in the brain, where it controls synapse formation and function. GPR158 has also been implicated in depression, carcinogenesis, and cognition. However, the structural organization and signaling mechanisms of GPR158 are largely unknown. We used single-particle cryo­electron microscopy (cryo-EM) to determine the structures of human GPR158 alone and bound to an RGS signaling complex. The structures reveal a homodimeric organization stabilized by a pair of phospholipids and the presence of an extracellular Cache domain, an unusual ligand-binding domain in GPCRs. We further demonstrate the structural basis of GPR158 coupling to RGS7-Gß5. Together, these results provide insights into the unusual biology of orphan receptors and the formation of GPCR-RGS complexes.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/química , Proteínas RGS/química , Receptores Acoplados a Proteínas G/química , Sitios de Unión , Microscopía por Crioelectrón , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Humanos , Ligandos , Modelos Moleculares , Fosfolípidos/química , Unión Proteica , Conformación Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos , Multimerización de Proteína , Subunidades de Proteína/química , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
4.
Matrix Biol Plus ; 12: 100074, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34337379

RESUMEN

The Neurofibromatosis type 2 gene encodes the Nf2/merlin tumor suppressor protein that is responsible for the regulation of cell proliferation. Once activated, Nf2/merlin modulates adhesive signaling pathways and thereby inhibits cell growth. Nf2/merlin controls oncogenic gene expression by modulating the Hippo pathway. By responding to several physical and biochemical stimuli, Hippo signaling determines contact inhibition of proliferation as well as organ size. The large tumor suppressor (LATS) serine/threonine-protein kinase is the key enzyme in the highly conserved kinase cascade that negatively regulates the activity and localization of the transcriptional coactivators Yes-associated protein (YAP) and its paralogue transcriptional coactivator with PDZ-binding motif (TAZ). Nf2/merlin belongs to the band 4.1, ezrin, radixin, moesin (FERM) gene family that links the actin cytoskeleton to adherens junctions, remodels adherens junctions during epithelial morphogenesis and maintains organized apical surfaces on the plasma cell membrane. Nf2/merlin and ERM proteins have a globular N-terminal cloverleaf head domain, the FERM domain, that binds to the plasma membrane, a central α-helical domain, and a tail domain that binds to its head domain. Here we present the high-resolution crystal structure of Nf2/merlin bound to LATS1 which shows that LATS1 binding to Nf2/merlin displaces the Nf2/merlin tail domain and causes an allosteric shift in the Nf2/merlin α-helix that extends from its FERM domain. This is consistent with the fact that full-length Nf2/merlin binds LATS1 ~10-fold weaker compared to LATS1 binding to the Nf2/merlin-PIP2 complex. Our data increase our understanding of Nf2/merlin biology by providing mechanistic insights into the Hippo pathway that are relevant to several diseases in particular oncogenic features that are associated with cancers.

5.
SLAS Discov ; 26(9): 1177-1188, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112017

RESUMEN

Regulators of G protein signaling (RGS) proteins serve as critical regulatory nodes to limit the lifetime and extent of signaling via G protein-coupled receptors (GPCRs). Previously, approaches to pharmacologically inhibit RGS activity have mostly focused on the inhibition of GTPase activity by interrupting the interaction of RGS proteins with the G proteins they regulate. However, several RGS proteins are also regulated by association with binding partners. A notable example is the mammalian RGS7 protein, which has prominent roles in metabolic control, vision, reward, and actions of opioid analgesics. In vivo, RGS7 exists in complex with the binding partners type 5 G protein ß subunit (Gß5) and R7 binding protein (R7BP), which control its stability and activity, respectively. Targeting the whole RGS7/Gß5/R7BP protein complex affords the opportunity to allosterically tune opioid receptor signaling following opioid engagement while potentially bypassing undesirable side effects. Hence, we implemented a novel strategy to pharmacologically target the interaction between RGS7/Gß5 and R7BP. To do so, we searched for protein complex inhibitors using a time-resolved fluorescence resonance energy transfer (FRET)-based high-throughput screening (HTS) assay that measures compound-mediated alterations in the FRET signal between RGS7/Gß5 and R7BP. We performed two HTS campaigns, each screening ~100,000 compounds from the Scripps Drug Discovery Library (SDDL). Each screen yielded more than 100 inhibitors, which will be described herein.


Asunto(s)
Descubrimiento de Drogas , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas RGS/metabolismo , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Complejos Multiproteicos/agonistas , Complejos Multiproteicos/antagonistas & inhibidores , Unión Proteica/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas
6.
Cell Rep ; 34(5): 108718, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33535037

RESUMEN

The G protein alpha subunit o (Gαo) is one of the most abundant proteins in the nervous system, and pathogenic mutations in its gene (GNAO1) cause movement disorder. However, the function of Gαo is ill defined mechanistically. Here, we show that Gαo dictates neuromodulatory responsiveness of striatal neurons and is required for movement control. Using in vivo optical sensors and enzymatic assays, we determine that Gαo provides a separate transduction channel that modulates coupling of both inhibitory and stimulatory dopamine receptors to the cyclic AMP (cAMP)-generating enzyme adenylyl cyclase. Through a combination of cell-based assays and rodent models, we demonstrate that GNAO1-associated mutations alter Gαo function in a neuron-type-specific fashion via a combination of a dominant-negative and loss-of-function mechanisms. Overall, our findings suggest that Gαo and its pathological variants function in specific circuits to regulate neuromodulatory signals essential for executing motor programs.


Asunto(s)
AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Trastornos del Movimiento/genética , Animales , Humanos , Ratones
7.
Elife ; 72018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30540250

RESUMEN

Signaling by the G-protein-coupled receptors (GPCRs) plays fundamental role in a vast number of essential physiological functions. Precise control of GPCR signaling requires action of regulators of G protein signaling (RGS) proteins that deactivate heterotrimeric G proteins. RGS proteins are elaborately regulated and comprise multiple domains and subunits, yet structural organization of these assemblies is poorly understood. Here, we report a crystal structure and dynamics analyses of the multisubunit complex of RGS7, a major regulator of neuronal signaling with key roles in controlling a number of drug target GPCRs and links to neuropsychiatric disease, metabolism, and cancer. The crystal structure in combination with molecular dynamics and mass spectrometry analyses reveals unique organizational features of the complex and long-range conformational changes imposed by its constituent subunits during allosteric modulation. Notably, several intermolecular interfaces in the complex work in synergy to provide coordinated modulation of this key GPCR regulator.


Asunto(s)
Proteínas Portadoras/química , Subunidades beta de la Proteína de Unión al GTP/química , Proteínas de Unión al GTP/metabolismo , Simulación de Dinámica Molecular , Neuronas/metabolismo , Proteínas RGS/química , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Medición de Intercambio de Deuterio , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Espectrometría de Masas , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Conformación Proteica , Multimerización de Proteína , Proteínas RGS/genética , Proteínas RGS/metabolismo , Homología de Secuencia de Aminoácido , Células Sf9 , Spodoptera
8.
Cell Rep ; 24(3): 557-568.e5, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30021154

RESUMEN

Despite the wealth of genetic information available, mechanisms underlying pathological effects of disease-associated mutations in components of G protein-coupled receptor (GPCR) signaling cascades remain elusive. In this study, we developed a scalable approach for the functional analysis of clinical variants in GPCR pathways along with a complete analytical framework. We applied the strategy to evaluate an extensive set of dystonia-causing mutations in G protein Gαolf. Our quantitative analysis revealed diverse mechanisms by which pathogenic variants disrupt GPCR signaling, leading to a mechanism-based classification of dystonia. In light of significant clinical heterogeneity, the mechanistic analysis of individual disease-associated variants permits tailoring personalized intervention strategies, which makes it superior to the current phenotype-based approach. We propose that the platform developed in this study can be universally applied to evaluate disease mechanisms for conditions associated with genetic variation in all components of GPCR signaling.


Asunto(s)
Enfermedad/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Adenilil Ciclasas/metabolismo , Animales , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Modelos Moleculares , Mutación/genética , Nucleótidos/metabolismo , Dominios Proteicos , Multimerización de Proteína , Estabilidad Proteica
9.
Proc Natl Acad Sci U S A ; 115(19): 5022-5027, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29686062

RESUMEN

Functional characterization of the GPCR interactome has been focused predominantly on intracellular interactions, yet GPCRs are increasingly found in complex with extracellular proteins. Extracellular leucine-rich repeat fibronectin type III domain containing 1 (ELFN1) was recently reported to physically anchor mGluR6 and mGluR7 across retinal and hippocampal synapses, respectively; however, the consequence of transsynaptic interactions on properties and pharmacology of these receptors are unknown. In the current study, we explore the effects of ELFN1 on mGluR signaling and pharmacology. First, we established the binding specificity of ELFN1 and found it to be recruited selectively to all group III mGluRs (mGluR4, mGluR6, mGluR7, and mGluR8), but not other mGluR species. Using site-directed mutagenesis we mapped binding determinants of this interaction to two distinct sites on the ELFN1 ectodomain. To evaluate functional aspects of the interaction, we developed a transcellular signaling assay in reconstituted HEK293 cells which monitors changes in mGluR activity in one cell following its exposure to separate ELFN1-containing cells. Using this platform, we found that ELFN1 acts as an allosteric modulator of class III mGluR activity in suppressing cAMP accumulation: altering both agonist-induced and constitutive receptor activity. Using bioluminescence resonance energy transfer-based real-time kinetic assays, we established that ELFN1 alters the ability of mGluRs to activate G proteins. Our findings demonstrate that core properties of class III mGluRs can be altered via extracellular interactions with ELFN1 which serves as a transsynaptic allosteric modulator for these receptors. Furthermore, our unique assay platform opens avenues for exploring transcellular/transsynaptic pharmacology of other GPCR transcomplexes.


Asunto(s)
AMP Cíclico/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Sistemas de Mensajero Secundario , Regulación Alostérica/fisiología , Sitios de Unión , AMP Cíclico/genética , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/genética , Receptores de Glutamato Metabotrópico/genética
10.
Assay Drug Dev Technol ; 16(3): 150-161, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29658790

RESUMEN

G protein-coupled receptors (GPCRs) are excellent drug targets exploited by majority of the Food and Drug Administration-approved medications, but when modulated, are often accompanied by significant adverse effects. Targeting of other elements in GPCR pathways for improved safety and efficacy is thus an unmet need. The strength of GPCR signaling is tightly regulated by regulators of G protein signaling (RGS) proteins, making them attractive drug targets. We focused on a prominent RGS complex in the brain consisting of RGS7 and its binding partners Gß5 and R7BP. These complexes play critical roles in regulating multiple GPCRs and essential physiological processes, yet no small molecule modulators are currently available to modify its function. In this study, we report a novel high-throughput approach to screen for small molecule modulators of the intramolecular transitions in the RGS7/Gß5/R7BP complex known to be involved in its allosteric regulation. We developed a time-resolved fluorescence energy transfer-based in vitro assay that utilizes full-length recombinant proteins and shows consistency, excellent assay statistics, and high level of sensitivity. We demonstrated the potential of this approach by screening two compound libraries (LOPAC 1280 and MicroSource Spectrum). This study confirms the feasibility of the chosen strategy for identifying small molecule modulators of RGS7/Gß5/R7BP complex for impacting signaling downstream of the GPCRs.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Ensayos Analíticos de Alto Rendimiento , Proteínas RGS/metabolismo , Animales , Ratones , Factores de Tiempo
11.
Hum Mol Genet ; 26(6): 1078-1086, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087732

RESUMEN

Global developmental delay (GDD), often accompanied by intellectual disability, seizures and other features is a severe, clinically and genetically highly heterogeneous childhood-onset disorder. In cases where genetic causes have been identified, de novo mutations in neuronally expressed genes are a common scenario. These mutations can be best identified by exome sequencing of parent-offspring trios. De novo mutations in the guanine nucleotide-binding protein, beta 1 (GNB1) gene, encoding the Gß1 subunit of heterotrimeric G proteins, have recently been identified as a novel genetic cause of GDD. Using exome sequencing, we identified 14 different novel variants (2 splice site, 2 frameshift and 10 missense changes) in GNB1 in 16 pediatric patients. One mutation (R96L) was recurrently found in three ethnically diverse families with an autosomal dominant mode of inheritance. Ten variants occurred de novo in the patients. Missense changes were functionally tested for their pathogenicity by assaying the impact on complex formation with Gγ and resultant mutant Gßγ with Gα. Signaling properties of G protein complexes carrying mutant Gß1 subunits were further analyzed by their ability to couple to dopamine D1R receptors by real-time bioluminescence resonance energy transfer (BRET) assays. These studies revealed altered functionality of the missense mutations R52G, G64V, A92T, P94S, P96L, A106T and D118G but not for L30F, H91R and K337Q. In conclusion, we demonstrate a pathogenic role of de novo and autosomal dominant mutations in GNB1 as a cause of GDD and provide insights how perturbation in heterotrimeric G protein function contributes to the disease.


Asunto(s)
Discapacidades del Desarrollo/genética , Subunidades beta de la Proteína de Unión al GTP/genética , Mutación Missense/genética , Neuronas/metabolismo , Niño , Preescolar , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Exoma/genética , Femenino , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Lactante , Masculino , Neuronas/patología , Unión Proteica , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo
12.
Genome Biol ; 17(1): 195, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27677260

RESUMEN

BACKGROUND: Neuropsychiatric disorders are common forms of disability in humans. Despite recent progress in deciphering the genetics of these disorders, their phenotypic complexity continues to be a major challenge. Mendelian neuropsychiatric disorders are rare but their study has the potential to unravel novel mechanisms that are relevant to their complex counterparts. RESULTS: In an extended consanguineous family, we identified a novel neuropsychiatric phenotype characterized by severe speech impairment, variable expressivity of attention deficit hyperactivity disorder (ADHD), and motor delay. We identified the disease locus through linkage analysis on 15q21.2, and exome sequencing revealed a novel missense variant in GNB5. GNB5 encodes an atypical ß subunit of the heterotrimeric GTP-binding proteins (Gß5). Gß5 is enriched in the central nervous system where it forms constitutive complexes with members of the regulator of G protein signaling family of proteins to modulate neurotransmitter signaling that affects a number of neurobehavioral outcomes. Here, we show that the S81L mutant form of Gß5 has significantly impaired activity in terminating responses that are elicited by dopamine. CONCLUSIONS: We demonstrate that these deficits originate from the impaired expression of the mutant Gß5 protein, resulting in the decreased ability to stabilize regulator of G protein signaling complexes. Our data suggest that this novel neuropsychiatric phenotype is the human equivalent of Gnb5 deficiency in mice, which manifest motor deficits and hyperactivity, and highlight a critical role of Gß5 in normal behavior as well as language and motor development in humans.

13.
Biochemistry ; 54(17): 2758-68, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25880222

RESUMEN

Vinculin localizes to cellular adhesions where it regulates motility, migration, development, wound healing, and response to force. Importantly, vinculin loss results in cancer phenotypes, cardiovascular disease, and embryonic lethality. At the plasma cell membrane, the most abundant phosphoinositide, phosphatidylinositol 4,5-bisphosphate (PIP2), binds the vinculin tail domain, Vt, and triggers homotypic and heterotypic interactions that amplify binding of vinculin to the actin network. Binding of PIP2 to Vt is necessary for maintaining optimal focal adhesions, for organizing stress fibers, for cell migration and spreading, and for the control of vinculin dynamics and turnover of focal adhesions. While the recently determined Vt/PIP2 crystal structure revealed the conformational changes occurring upon lipid binding and oligomerization, characterization of PIP2-induced vinculin oligomerization has been challenging in the adhesion biology field. Here, via a series of novel biochemical assays not performed in previous studies that relied on chemical cross-linking, we characterize the PIP2-induced vinculin oligomerization. Our results show that Vt/PIP2 forms a tight dimer with Vt or with the muscle-specific vinculin isoform, metavinculin, at sites of adhesion at the cell membrane. Insight into how PIP2 regulates clustering and into mechanisms that regulate cell adhesion allows the development for a more definite sensor for PIP2, and our developed techniques can be applied generally and thus open the door for the characterization of many other protein/PIP2 complexes under physiological conditions.


Asunto(s)
Lípidos/química , Vinculina/química , Adhesión Celular , Membrana Celular/química , Cromatografía en Gel , Dimerización , Resonancia por Plasmón de Superficie , Vinculina/metabolismo
14.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 2): 239-55, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25664734

RESUMEN

KdsC, the third enzyme of the 3-deoxy-D-manno-octulosonic acid (KDO) biosynthetic pathway, catalyzes a substrate-specific reaction to hydrolyze 3-deoxy-D-manno-octulosonate 8-phosphate to generate a molecule of KDO and phosphate. KdsC is a phosphatase that belongs to the C0 subfamily of the HAD superfamily. To understand the molecular basis for the substrate specificity of this tetrameric enzyme, the crystal structures of KdsC from Moraxella catarrhalis (Mc-KdsC) with several combinations of ligands, namely metal ion, citrate and products, were determined. Various transition states of the enzyme have been captured in these crystal forms. The ligand-free and ligand-bound crystal forms reveal that the binding of ligands does not cause any specific conformational changes in the active site. However, the electron-density maps clearly showed that the conformation of KDO as a substrate is different from the conformation adopted by KDO when it binds as a cleaved product. Furthermore, structural evidence for the existence of an intersubunit tunnel has been reported for the first time in the C0 subfamily of enzymes. A role for this tunnel in transferring water molecules from the interior of the tetrameric structure to the active-site cleft has been proposed. At the active site, water molecules are required for the formation of a water bridge that participates as a proton shuttle during the second step of the two-step phosphoryl-transfer reaction. In addition, as the KDO biosynthesis pathway is a potential antibacterial target, pharmacophore-based virtual screening was employed to identify inhibitor molecules for the Mc-KdsC enzyme.


Asunto(s)
Acuaporinas/química , Proteínas Bacterianas/química , Moraxella catarrhalis/enzimología , Infecciones por Moraxellaceae/microbiología , Monoéster Fosfórico Hidrolasas/química , Secuencia de Aminoácidos , Acuaporinas/metabolismo , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Ácido Cítrico/metabolismo , Cristalografía por Rayos X , Humanos , Ligandos , Datos de Secuencia Molecular , Moraxella catarrhalis/química , Moraxella catarrhalis/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Unión Proteica , Conformación Proteica , Alineación de Secuencia , Especificidad por Sustrato
15.
J Cell Biol ; 207(5): 643-56, 2014 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-25488920

RESUMEN

Adherens junctions (AJs) and focal adhesion (FA) complexes are necessary for cell migration and morphogenesis, and for the development, growth, and survival of all metazoans. Vinculin is an essential regulator of both AJs and FAs, where it provides links to the actin cytoskeleton. Phosphatidylinositol 4,5-bisphosphate (PIP2) affects the functions of many targets, including vinculin. Here we report the crystal structure of vinculin in complex with PIP2, which revealed that PIP2 binding alters vinculin structure to direct higher-order oligomerization and suggests that PIP2 and F-actin binding to vinculin are mutually permissive. Forced expression of PIP2-binding-deficient mutants of vinculin in vinculin-null mouse embryonic fibroblasts revealed that PIP2 binding is necessary for maintaining optimal FAs, for organization of actin stress fibers, and for cell migration and spreading. Finally, photobleaching experiments indicated that PIP2 binding is required for the control of vinculin dynamics and turnover in FAs. Thus, through oligomerization, PIP2 directs a transient vinculin sequestration at FAs that is necessary for proper FA function.


Asunto(s)
Fosfatidilinositol 4,5-Difosfato/química , Vinculina/química , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células Cultivadas , Cristalografía por Rayos X , Adhesiones Focales/fisiología , Humanos , Enlace de Hidrógeno , Ratones , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Vinculina/fisiología
16.
PLoS One ; 8(5): e63779, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23717482

RESUMEN

The glycosyl hydrolase 18 (GH18) family consists of active chitinases as well as chitinase like lectins/proteins (CLPs). The CLPs share significant sequence and structural similarities with active chitinases, however, do not display chitinase activity. Some of these proteins are reported to have specific functions and carbohydrate binding property. In the present study, we report a novel chitinase like lectin (TCLL) from Tamarindus indica. The crystal structures of native TCLL and its complex with N-acetyl glucosamine were determined. Similar to the other CLPs of the GH18 members, TCLL lacks chitinase activity due to mutations of key active site residues. Comparison of TCLL with chitinases and other chitin binding CLPs shows that TCLL has substitution of some chitin binding site residues and more open binding cleft due to major differences in the loop region. Interestingly, the biochemical studies suggest that TCLL is an N-acetyl glucosamine specific chi-lectin, which is further confirmed by the complex structure of TCLL with N-acetyl glucosamine complex. TCLL has two distinct N-acetyl glucosamine binding sites S1 and S2 that contain similar polar residues, although interaction pattern with N-acetyl glucosamine varies extensively among them. Moreover, TCLL structure depicts that how plants utilize existing structural scaffolds ingenuously to attain new functions. To date, this is the first structural investigation of a chi-lectin from plants that explore novel carbohydrate binding sites other than chitin binding groove observed in GH18 family members. Consequently, TCLL structure confers evidence for evolutionary link of lectins with chitinases.


Asunto(s)
Quitinasas/química , Hemaglutininas/química , Lectinas de Plantas/química , Proteínas de Plantas/química , Tamarindus/enzimología , Acetilglucosamina/química , Secuencia de Aminoácidos , Dominio Catalítico , Quitinasas/farmacología , Cristalografía por Rayos X , Evolución Molecular , Hemaglutinación , Hemaglutininas/farmacología , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Peso Molecular , Filogenia , Lectinas de Plantas/farmacología , Proteínas de Plantas/farmacología , Unión Proteica , Análisis de Secuencia de Proteína , Homología Estructural de Proteína
17.
Protein Pept Lett ; 20(5): 499-509, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22973842

RESUMEN

Wrightia tinctoria globulin (WTG), one of the major seed storage proteins, was isolated for the first time from seeds of the medicinal plant. WTG was extracted and purified to homogeneity in two steps using anion-exchange and size-exclusion chromatographies. On an SDS-PAGE gel under non-reducing conditions, a major band of ~56 kDa was observed; under reducing conditions, however, two major polypeptides, one with molecular weight ~32-34 kDa and the other with molecular weight ~22-26 kDa were observed. Intact mass determination by MALDI-TOF supported this observation. The N-terminal amino acid sequence of WTG matched in NCBI database with an expressed sequence tag obtained from the c-DNA of developing embryo m-RNA of Wrightia tinctoria. The EST sequence was further substantiated by partial de novo internal sequencing using MALDI-TOF/TOF. The high sequence homology with seed storage protein 11S globulin confirmed that WTG is a type of 11S globulin. Circular dichroism analysis showed that the secondary structure of WTG consists predominantly of ß-sheets (44.2%) and moderate content of α-helices (10.3%). WTG showed hemagglutinating property indicating that the protein may possess lectin-like activity. WTG was crystallized at 20 Å°C by the vapour diffusion method using PEG 400 as precipitant. The crystals belonged to the orthorhombic space group P212121 with cell dimensions of a=109.9Å, b=113.2Å and c=202.2Å with six molecules per asymmetric unit. Diffraction data were collected to a resolution of 2.2Å under cryocondition. Preliminary structure solution of WTG indicated the possibility of a hexameric assembly in its asymmetric unit.


Asunto(s)
Apocynaceae/química , Globulinas/química , Hemaglutinación/efectos de los fármacos , Proteínas de Almacenamiento de Semillas/química , Secuencia de Aminoácidos , Western Blotting , Electroforesis en Gel de Poliacrilamida , Eritrocitos/efectos de los fármacos , Globulinas/aislamiento & purificación , Globulinas/farmacología , Humanos , Datos de Secuencia Molecular , Proteínas de Almacenamiento de Semillas/aislamiento & purificación , Proteínas de Almacenamiento de Semillas/farmacología , Semillas/química , Alineación de Secuencia , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Difracción de Rayos X
18.
FEBS J ; 279(24): 4547-64, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23094997

RESUMEN

A Kunitz type dual inhibitor (TKI) of factor Xa (FXa) and trypsin was found in tamarind. It also shows prolongation of blood coagulation time. The deduced 185 amino acid sequence of TKI by cDNA cloning and sequence analysis revealed that it belongs to the Kunitz type soybean trypsin inhibitor (STI) family; however, it has a distorted Kunitz signature sequence due to insertion of Asn15 in the motif. TKI exhibited a competitive inhibitory activity against both FXa (K(i)  = 220 nm) and porcine pancreatic trypsin (K(i)  = 3.2 nm). The crystal structure of TKI shows a ß-trefoil fold similar to Kunitz STI inhibitors; however, a distinct mobile reactive site, an inserted residue and loop ß7ß8 make it distinct from classical Kunitz inhibitors. The crystal structure of TKI-trypsin and a 3D model of TKI-FXa complex revealed that the distinct reactive site loop probably plays a role in dual inhibition. The reactive site of TKI interacts with an active site and two exosites (36 loop and autolysis loop) of FXa. Apart from Arg66 (P1), Arg64 (P3) is one of the most important residues responsible for the specificity of TKI towards FXa. Along with the reactive site loop (ß4ß5), loops ß1 and ß7ß8 also interact with FXa and could further confer selectivity for FXa. We also present the role of inserted Asn15 in the stabilization of complexes. To the best of our knowledge, this is the first structure of FXa inhibitor belonging to the Kunitz type inhibitor family and its unique structural and sequence features make TKI a novel potent inhibitor. DATABASE: The complete nucleotide of TKI was deposited in the NCBI gene databank with accession no. HQ385502. The atomic coordinates and structure factor files for the structure of TKI and TKI:PPT complex have been deposited in the Protein Data Bank with accession numbers 4AN6 and 4AN7, respectively STRUCTURED DIGITAL ABSTRACT:  TKI and TKI bind by x-ray crystallography (View interaction)  TKI and PPT bind by x-ray crystallography (View interaction).


Asunto(s)
Inhibidores del Factor Xa , Inhibidores de Proteasas/farmacología , Inhibidores de Tripsina/farmacología , Secuencia de Aminoácidos , Modelos Moleculares , Datos de Secuencia Molecular , Inhibidores de Proteasas/química , Conformación Proteica , Homología de Secuencia de Aminoácido , Inhibidores de Tripsina/química
19.
PLoS One ; 7(1): e30102, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22291906

RESUMEN

Expression of KdpFABC, a K(+) pump that restores osmotic balance, is controlled by binding of the response regulator KdpE to a specific DNA sequence (kdpFABC(BS)) via the winged helix-turn-helix type DNA binding domain (KdpE(DBD)). Exploration of E. coli KdpE(DBD) and kdpFABC(BS) interaction resulted in the identification of two conserved, AT-rich 6 bp direct repeats that form half-sites. Despite binding to these half-sites, KdpE(DBD) was incapable of promoting gene expression in vivo. Structure-function studies guided by our 2.5 Å X-ray structure of KdpE(DBD) revealed the importance of residues R193 and R200 in the α-8 DNA recognition helix and T215 in the wing region for DNA binding. Mutation of these residues renders KdpE incapable of inducing expression of the kdpFABC operon. Detailed biophysical analysis of interactions using analytical ultracentrifugation revealed a 2∶1 stoichiometry of protein to DNA with dissociation constants of 200±100 and 350±100 nM at half-sites. Inactivation of one half-site does not influence binding at the other, indicating that KdpE(DBD) binds independently to the half-sites with approximately equal affinity and no discernable cooperativity. To our knowledge, these data are the first to describe in quantitative terms the binding at half-sites under equilibrium conditions for a member of the ubiquitous OmpR/PhoB family of proteins.


Asunto(s)
ADN/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología , Elementos de Respuesta , Transactivadores/química , Transactivadores/metabolismo , Transactivadores/fisiología , Secuencia de Aminoácidos , Sitios de Unión/genética , Cristalografía por Rayos X , Proteínas de Escherichia coli/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Elementos de Respuesta/genética , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Especificidad por Sustrato/genética , Transactivadores/genética
20.
J Biol Chem ; 286(42): 37011-22, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21880718

RESUMEN

Biphenyl dehydrogenase, a member of short-chain dehydrogenase/reductase enzymes, catalyzes the second step of the biphenyl/polychlorinated biphenyls catabolic pathway in bacteria. To understand the molecular basis for the broad substrate specificity of Pandoraea pnomenusa strain B-356 biphenyl dehydrogenase (BphB(B-356)), the crystal structures of the apo-enzyme, the binary complex with NAD(+), and the ternary complexes with NAD(+)-2,3-dihydroxybiphenyl and NAD(+)-4,4'-dihydroxybiphenyl were determined at 2.2-, 2.5-, 2.4-, and 2.1-Å resolutions, respectively. A crystal structure representing an intermediate state of the enzyme was also obtained in which the substrate binding loop was ordered as compared with the apo and binary forms but it was displaced significantly with respect to the ternary structures. These five structures reveal that the substrate binding loop is highly mobile and that its conformation changes during ligand binding, starting from a disorganized loop in the apo state to a well organized loop structure in the ligand-bound form. Conformational changes are induced during ligand binding; forming a well defined cavity to accommodate a wide variety of substrates. This explains the biochemical data that shows BphB(B-356) converts the dihydrodiol metabolites of 3,3'-dichlorobiphenyl, 2,4,4'-trichlorobiphenyl, and 2,6-dichlorobiphenyl to their respective dihydroxy metabolites. For the first time, a combination of structural, biochemical, and molecular docking studies of BphB(B-356) elucidate the unique ability of the enzyme to transform the cis-dihydrodiols of double meta-, para-, and ortho-substituted chlorobiphenyls.


Asunto(s)
Proteínas Bacterianas/química , Burkholderiaceae/enzimología , Oxidorreductasas/química , Clorofenoles/química , Cristalografía por Rayos X , Ligandos , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA