Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Cell ; 80(2): 237-245.e4, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33007200

RESUMEN

Heterotrimeric G proteins communicate signals from activated G protein-coupled receptors to downstream effector proteins. In the phototransduction pathway responsible for vertebrate vision, the G protein-effector complex is composed of the GTP-bound transducin α subunit (GαT·GTP) and the cyclic GMP (cGMP) phosphodiesterase 6 (PDE6), which stimulates cGMP hydrolysis, leading to hyperpolarization of the photoreceptor cell. Here we report a cryo-electron microscopy (cryoEM) structure of PDE6 complexed to GTP-bound GαT. The structure reveals two GαT·GTP subunits engaging the PDE6 hetero-tetramer at both the PDE6 catalytic core and the PDEγ subunits, driving extensive rearrangements to relieve all inhibitory constraints on enzyme catalysis. Analysis of the conformational ensemble in the cryoEM data highlights the dynamic nature of the contacts between the two GαT·GTP subunits and PDE6 that supports an alternating-site catalytic mechanism.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Transducción de Señal , Transducina/metabolismo , Animales , Biocatálisis , Dominio Catalítico , Bovinos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/ultraestructura , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Transducina/química , Transducina/ultraestructura , Diclorhidrato de Vardenafil/química , Diclorhidrato de Vardenafil/metabolismo
2.
Mol Cell ; 75(4): 781-790.e3, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31300275

RESUMEN

Rhodopsin (Rho), a prototypical G-protein-coupled receptor (GPCR) in vertebrate vision, activates the G-protein transducin (GT) by catalyzing GDP-GTP exchange on its α subunit (GαT). To elucidate the determinants of GT coupling and activation, we obtained cryo-EM structures of a fully functional, light-activated Rho-GT complex in the presence and absence of a G-protein-stabilizing nanobody. The structures illustrate how GT overcomes its low basal activity by engaging activated Rho in a conformation distinct from other GPCR-G-protein complexes. Moreover, the nanobody-free structures reveal native conformations of G-protein components and capture three distinct conformers showing the GαT helical domain (αHD) contacting the Gßγ subunits. These findings uncover the molecular underpinnings of G-protein activation by visual rhodopsin and shed new light on the role played by Gßγ during receptor-catalyzed nucleotide exchange.


Asunto(s)
Complejos Multiproteicos/química , Rodopsina/química , Transducina/química , Animales , Bovinos , Microscopía por Crioelectrón , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Dominios Proteicos , Estructura Secundaria de Proteína , Rodopsina/metabolismo , Transducina/metabolismo
3.
J Biol Chem ; 299(8): 104963, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37356720

RESUMEN

Vimentin intermediate filaments form part of the cytoskeleton of mesenchymal cells, but under pathological conditions often associated with inflammation, vimentin filaments depolymerize as the result of phosphorylation or citrullination, and vimentin oligomers are secreted or released into the extracellular environment. In the extracellular space, vimentin can bind surfaces of cells and the extracellular matrix, and the interaction between extracellular vimentin and cells can trigger changes in cellular functions, such as activation of fibroblasts to a fibrotic phenotype. The mechanism by which extracellular vimentin binds external cell membranes and whether vimentin alone can act as an adhesive anchor for cells is largely uncharacterized. Here, we show that various cell types (normal and vimentin null fibroblasts, mesenchymal stem cells, and A549 lung carcinoma cells) attach to and spread on polyacrylamide hydrogel substrates covalently linked to vimentin. Using traction force microscopy and spheroid expansion assays, we characterize how different cell types respond to extracellular vimentin. Cell attachment to and spreading on vimentin-coated surfaces is inhibited by hyaluronic acid degrading enzymes, hyaluronic acid synthase inhibitors, soluble heparin or N-acetyl glucosamine, all of which are treatments that have little or no effect on the same cell types binding to collagen-coated hydrogels. These studies highlight the effectiveness of substrate-bound vimentin as a ligand for cells and suggest that carbohydrate structures, including the glycocalyx and glycosylated cell surface proteins that contain N-acetyl glucosamine, form a novel class of adhesion receptors for extracellular vimentin that can either directly support cell adhesion to a substrate or fine-tune the glycocalyx adhesive properties.


Asunto(s)
Vimentina , Acetilglucosamina/química , Adhesión Celular , Movimiento Celular , Ácido Hialurónico/química , Filamentos Intermedios/metabolismo , Vimentina/metabolismo , Humanos , Línea Celular Tumoral
5.
J Biol Chem ; 298(2): 101564, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34999118

RESUMEN

The mitochondrial enzyme glutaminase C (GAC) is upregulated in many cancer cells to catalyze the first step in glutamine metabolism, the hydrolysis of glutamine to glutamate. The dependence of cancer cells on this transformed metabolic pathway highlights GAC as a potentially important therapeutic target. GAC acquires maximal catalytic activity upon binding to anionic activators such as inorganic phosphate. To delineate the mechanism of GAC activation, we used the tryptophan substitution of tyrosine 466 in the catalytic site of the enzyme as a fluorescent reporter for glutamine binding in the presence and absence of phosphate. We show that in the absence of phosphate, glutamine binding to the Y466W GAC tetramer exhibits positive cooperativity. A high-resolution X-ray structure of tetrameric Y466W GAC bound to glutamine suggests that cooperativity in substrate binding is coupled to tyrosine 249, located at the edge of the catalytic site (i.e., the "lid"), adopting two distinct conformations. In one dimer within the GAC tetramer, the lids are open and glutamine binds weakly, whereas, in the adjoining dimer, the lids are closed over the substrates, resulting in higher affinity interactions. When crystallized in the presence of glutamine and phosphate, all four subunits of the Y466W GAC tetramer exhibited bound glutamine with closed lids. Glutamine can bind with high affinity to each subunit, which subsequently undergo simultaneous catalysis. These findings explain how the regulated transitioning of GAC between different conformational states ensures that maximal catalytic activity is reached in cancer cells only when an allosteric activator is available.


Asunto(s)
Glutaminasa , Glutamina , Mitocondrias , Dominio Catalítico , Glutaminasa/química , Glutaminasa/metabolismo , Glutamina/química , Glutamina/metabolismo , Mitocondrias/enzimología , Mitocondrias/metabolismo , Fosfatos/química , Fosfatos/metabolismo , Conformación Proteica , Tirosina/química , Tirosina/metabolismo
6.
J Biol Chem ; 298(2): 101535, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34954143

RESUMEN

Cancer cells frequently exhibit uncoupling of the glycolytic pathway from the TCA cycle (i.e., the "Warburg effect") and as a result, often become dependent on their ability to increase glutamine catabolism. The mitochondrial enzyme Glutaminase C (GAC) helps to satisfy this 'glutamine addiction' of cancer cells by catalyzing the hydrolysis of glutamine to glutamate, which is then converted to the TCA-cycle intermediate α-ketoglutarate. This makes GAC an intriguing drug target and spurred the molecules derived from bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide (the so-called BPTES class of allosteric GAC inhibitors), including CB-839, which is currently in clinical trials. However, none of the drugs targeting GAC are yet approved for cancer treatment and their mechanism of action is not well understood. Here, we shed new light on the underlying basis for the differential potencies exhibited by members of the BPTES/CB-839 family of compounds, which could not previously be explained with standard cryo-cooled X-ray crystal structures of GAC bound to CB-839 or its analogs. Using an emerging technique known as serial room temperature crystallography, we were able to observe clear differences between the binding conformations of inhibitors with significantly different potencies. We also developed a computational model to further elucidate the molecular basis of differential inhibitor potency. We then corroborated the results from our modeling efforts using recently established fluorescence assays that directly read out inhibitor binding to GAC. Together, these findings should aid in future design of more potent GAC inhibitors with better clinical outlook.


Asunto(s)
Inhibidores Enzimáticos , Glutaminasa , Neoplasias , Sulfuros , Tiadiazoles , Cristalografía , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glutaminasa/antagonistas & inhibidores , Glutaminasa/química , Glutaminasa/metabolismo , Glutamina/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Sulfuros/química , Sulfuros/farmacología , Temperatura , Tiadiazoles/química , Tiadiazoles/farmacología
7.
J Biol Chem ; 295(5): 1328-1337, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31871054

RESUMEN

The glutaminase C (GAC) isoform of mitochondrial glutaminase is overexpressed in many cancer cells and therefore represents a potential therapeutic target. Understanding the regulation of GAC activity has been guided by the development of spectroscopic approaches that measure glutaminase activity in real time. Previously, we engineered a GAC protein (GAC(F327W)) in which a tryptophan residue is substituted for phenylalanine in an activation loop to explore the role of this loop in enzyme activity. We showed that the fluorescence emission of Trp-327 is enhanced in response to activator binding, but quenched by inhibitors of the BPTES class that bind to the GAC tetramer and contact the activation loop, thereby constraining it in an inactive conformation. In the present work, we took advantage of a tryptophan substitution at position 471, proximal to the GAC catalytic site, to examine the conformational coupling between the activation loop and the substrate-binding cleft, separated by ∼16 Å. Comparison of glutamine binding in the presence or absence of the BPTES analog CB-839 revealed a reciprocal relationship between the constraints imposed on the activation loop position and the affinity of GAC for substrate. Binding of the inhibitor weakened the affinity of GAC for glutamine, whereas activating anions such as Pi increased this affinity. These results indicate that the conformations of the activation loop and the substrate-binding cleft in GAC are allosterically coupled and that this coupling determines substrate affinity and enzymatic activity and explains the activities of CB-839, which is currently in clinical trials.


Asunto(s)
Bencenoacetamidas/farmacología , Glutaminasa/química , Glutamina/metabolismo , Mitocondrias/enzimología , Tiadiazoles/farmacología , Regulación Alostérica/genética , Sitio Alostérico/genética , Sustitución de Aminoácidos/genética , Animales , Ingeniería Biomédica , Dominio Catalítico/genética , Glutaminasa/metabolismo , Cinética , Ratones , Mitocondrias/química , Modelos Moleculares , Mutación , Isoformas de Proteínas , Estructura Terciaria de Proteína/genética , Proteínas Recombinantes , Sulfuros/farmacología
8.
J Biol Chem ; 293(46): 17941-17952, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30266806

RESUMEN

Two regions on the α subunits of heterotrimeric GTP-binding proteins (G-proteins), the Switch II/α2 helix (which changes conformation upon GDP-GTP exchange) and the α3 helix, have been shown to contain the binding sites for their effector proteins. However, how the binding of Gα subunits to their effector proteins is translated into the stimulation of effector activity is still poorly understood. Here, we took advantage of a reconstituted rhodopsin-coupled phototransduction system to address this question and identified a distinct surface and an essential residue on the α subunit of the G-protein transducin (αT) that is necessary to fully activate its effector enzyme, the cGMP phosphodiesterase (PDE). We started with a chimeric G-protein α subunit (αT*) comprising residues mainly from αT and a short stretch of residues from the Gi1 α subunit (αi1), which only weakly stimulates PDE activity. We then reinstated the αT residues by systematically replacing the corresponding αi1 residues within αT* with the aim of fully restoring PDE stimulatory activity. These experiments revealed that the αG/α4 loop and a phenylalanine residue at position 283 are essential for conferring the αT* subunit with full PDE stimulatory capability. We further demonstrated that this same region and amino acid within the α subunit of the Gs protein (αs) are necessary for full adenylyl cyclase activation. These findings highlight the importance of the αG/α4 loop and of an essential phenylalanine residue within this region on Gα subunits αT and αs as being pivotal for their selective and optimal stimulation of effector activity.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Fenilalanina/química , Transducina/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Bovinos , Cromograninas/metabolismo , Activación Enzimática , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Mutación con Ganancia de Función , Células HEK293 , Humanos , Conformación Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Rodopsina/metabolismo , Transducina/genética
9.
J Biol Chem ; 292(34): 14280-14289, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28655769

RESUMEN

The visual photo-transduction cascade is a prototypical G protein-coupled receptor (GPCR) signaling system, in which light-activated rhodopsin (Rho*) is the GPCR catalyzing the exchange of GDP for GTP on the heterotrimeric G protein transducin (GT). This results in the dissociation of GT into its component αT-GTP and ß1γ1 subunit complex. Structural information for the Rho*-GT complex will be essential for understanding the molecular mechanism of visual photo-transduction. Moreover, it will shed light on how GPCRs selectively couple to and activate their G protein signaling partners. Here, we report on the preparation of a stable detergent-solubilized complex between Rho* and a heterotrimer (GT*) comprising a GαT/Gαi1 chimera (αT*) and ß1γ1 The complex was formed on native rod outer segment membranes upon light activation, solubilized in lauryl maltose neopentyl glycol, and purified with a combination of affinity and size-exclusion chromatography. We found that the complex is fully functional and that the stoichiometry of Rho* to GαT* is 1:1. The molecular weight of the complex was calculated from small-angle X-ray scattering data and was in good agreement with a model consisting of one Rho* and one GT*. The complex was visualized by negative-stain electron microscopy, which revealed an architecture similar to that of the ß2-adrenergic receptor-GS complex, including a flexible αT* helical domain. The stability and high yield of the purified complex should allow for further efforts toward obtaining a high-resolution structure of this important signaling complex.


Asunto(s)
Proteínas del Ojo/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Modelos Moleculares , Rodopsina/metabolismo , Transducina/metabolismo , Animales , Bovinos , Cristalografía por Rayos X , Detergentes/química , Proteínas del Ojo/química , Proteínas del Ojo/genética , Proteínas del Ojo/aislamiento & purificación , Subunidades beta de la Proteína de Unión al GTP/química , Subunidades beta de la Proteína de Unión al GTP/aislamiento & purificación , Subunidades gamma de la Proteína de Unión al GTP/química , Subunidades gamma de la Proteína de Unión al GTP/aislamiento & purificación , Luz , Microscopía Electrónica , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/metabolismo , Conformación Proteica/efectos de la radiación , Multimerización de Proteína/efectos de la radiación , Estabilidad Proteica/efectos de la radiación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Retina/enzimología , Retina/metabolismo , Retina/efectos de la radiación , Rodopsina/química , Rodopsina/aislamiento & purificación , Segmento Externo de la Célula en Bastón/enzimología , Segmento Externo de la Célula en Bastón/metabolismo , Segmento Externo de la Célula en Bastón/efectos de la radiación , Dispersión del Ángulo Pequeño , Solubilidad , Transducina/química , Transducina/genética , Transducina/aislamiento & purificación , Difracción de Rayos X
10.
Proc Natl Acad Sci U S A ; 112(2): 394-9, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25548170

RESUMEN

The mitochondrial enzyme glutaminase C (GAC) catalyzes the hydrolysis of glutamine to glutamate plus ammonia, a key step in the metabolism of glutamine by cancer cells. Recently, we discovered a class of allosteric inhibitors of GAC that inhibit cancer cell growth without affecting their normal cellular counterparts, with the lead compound being the bromo-benzophenanthridinone 968. Here, we take advantage of mouse embryonic fibroblasts transformed by oncogenic Dbl, which hyperactivates Rho GTPases, together with (13)C-labeled glutamine and stable-isotope tracing methods, to establish that 968 selectively blocks the enhancement in glutaminolysis necessary for satisfying the glutamine addiction of cancer cells. We then determine how 968 inhibits the catalytic activity of GAC. First, we developed a FRET assay to examine the effects of 968 on the ability of GAC to undergo the dimer-to-tetramer transition necessary for enzyme activation. We next demonstrate how the fluorescence of a reporter group attached to GAC provides a direct read-out of the binding of 968 and related compounds to the enzyme. By combining these fluorescence assays with newly developed GAC mutants trapped in either the monomeric or dimeric state, we show that 968 has the highest affinity for monomeric GAC and that the dose-dependent binding of 968 to GAC monomers directly matches its dose-dependent inhibition of enzyme activity and cellular transformation. Together, these findings highlight the requirement of tetramer formation as the mechanism of GAC activation and shed new light on how a distinct class of allosteric GAC inhibitors impacts the metabolic program of transformed cells.


Asunto(s)
Glutamina/metabolismo , Regulación Alostérica , Sustitución de Aminoácidos , Animales , Benzofenantridinas/farmacología , Transformación Celular Neoplásica/metabolismo , Inhibidores Enzimáticos/farmacología , Transferencia Resonante de Energía de Fluorescencia , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Estructura Cuaternaria de Proteína/efectos de los fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Transaminasas/antagonistas & inhibidores , Transaminasas/química , Transaminasas/genética
11.
J Biol Chem ; 291(40): 20900-20910, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27542409

RESUMEN

Glutamine-derived carbon becomes available for anabolic biosynthesis in cancer cells via the hydrolysis of glutamine to glutamate, as catalyzed by GAC, a splice variant of kidney-type glutaminase (GLS). Thus, there is significant interest in understanding the regulation of GAC activity, with the suggestion being that higher order oligomerization is required for its activation. We used x-ray crystallography, together with site-directed mutagenesis, to determine the minimal enzymatic unit capable of robust catalytic activity. Mutagenesis of the helical interface between the two pairs of dimers comprising a GAC tetramer yielded a non-active, GAC dimer whose x-ray structure displays a stationary loop ("activation loop") essential for coupling the binding of allosteric activators like inorganic phosphate to catalytic activity. Further mutagenesis that removed constraints on the activation loop yielded a constitutively active dimer, providing clues regarding how the activation loop communicates with the active site, as well as with a peptide segment that serves as a "lid" to close off the active site following substrate binding. Our studies show that the formation of large GAC oligomers is not a pre-requisite for full enzymatic activity. They also offer a mechanism by which the binding of activators like inorganic phosphate enables the activation loop to communicate with the active site to ensure maximal rates of catalysis, and promotes the opening of the lid to achieve optimal product release. Moreover, these findings provide new insights into how other regulatory events might induce GAC activation within cancer cells.


Asunto(s)
Glutaminasa/metabolismo , Glutamina/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Multimerización de Proteína , Animales , Línea Celular Tumoral , Activación Enzimática , Glutaminasa/química , Glutaminasa/genética , Glutamina/química , Glutamina/genética , Humanos , Ratones , Células 3T3 NIH , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Estructura Secundaria de Proteína
12.
Biochemistry ; 51(15): 3232-40, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22448927

RESUMEN

The activation of Gα subunits of heterotrimeric G proteins by G protein-coupled receptors (GPCRs) is a critical event underlying a variety of biological responses. Understanding how G proteins are activated will require structural and biochemical analyses of GPCRs complexed to their G protein partners, together with structure-function studies of Gα mutants that shed light on the different steps in the activation pathway. Previously, we reported that the substitution of a glycine for a proline at position 56 within the linker region connecting the helical and GTP-binding domains of a Gα chimera, designated αT*, yields a more readily exchangeable state for guanine nucleotides. Here we show that GDP-GTP exchange on αT*(G56P), in the presence of the light-activated GPCR, rhodopsin (R*), is less sensitive to the ß1γ1 subunit complex than to wild-type αT*. We determined the X-ray crystal structure for the αT*(G56P) mutant and found that the G56P substitution leads to concerted changes that are transmitted to the conformationally sensitive switch regions, the α4-ß6 loop, and the ß6 strand. The α4-ß6 loop has been proposed to be a GPCR contact site that signals to the TCAT motif and weakens the binding of the guanine ring of GDP, whereas the switch regions are the contact sites for the ß1γ1 complex. Collectively, these biochemical and structural data lead us to suggest that αT*(G56P) may be adopting a conformation that is normally induced within Gα subunits by the combined actions of a GPCR and a Gßγ subunit complex during the G protein activation event.


Asunto(s)
Proteínas de Unión al GTP/química , Arginina/genética , Arginina/metabolismo , Cristalografía por Rayos X , Subunidades alfa de la Proteína de Unión al GTP/química , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Proteínas de Unión al GTP/metabolismo , Lisina/genética , Lisina/metabolismo , Modelos Moleculares , Conformación Proteica , Rodopsina/química , Rodopsina/metabolismo , Relación Estructura-Actividad
13.
J Biol Chem ; 286(14): 12702-11, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21285355

RESUMEN

Residues comprising the guanine nucleotide-binding sites of the α subunits of heterotrimeric (large) G-proteins (Gα subunits), as well as the Ras-related (small) G-proteins, are highly conserved. This is especially the case for the phosphate-binding loop (P-loop) where both Gα subunits and Ras-related G-proteins have a conserved serine or threonine residue. Substitutions for this residue in Ras and related (small) G-proteins yield nucleotide-depleted, dominant-negative mutants. Here we have examined the consequences of changing the conserved serine residue in the P-loop to asparagine, within a chimeric Gα subunit (designated αT*) that is mainly comprised of the α subunit of the retinal G-protein transducin and a limited region from the α subunit of Gi1. The αT*(S43N) mutant exhibits a significantly higher rate of intrinsic GDP-GTP exchange compared with wild-type αT*, with light-activated rhodopsin (R*) causing only a moderate increase in the kinetics of nucleotide exchange on αT*(S43N). The αT*(S43N) mutant, when bound to either GDP or GTP, was able to significantly slow the rate of R*-catalyzed GDP-GTP exchange on wild-type αT*. Thus, GTP-bound αT*(S43N), as well as the GDP-bound mutant, is capable of forming a stable complex with R*. αT*(S43N) activated the cGMP phosphodiesterase (PDE) with a dose-response similar to wild-type αT*. Activation of the PDE by αT*(S43N) was unaffected if either R* or ß1γ1 alone was present, whereas it was inhibited when R* and the ß1γ1 subunit were added together. Overall, our studies suggest that the S43N substitution on αT* stabilizes an intermediate on the G-protein activation pathway consisting of an activated G-protein-coupled receptor, a GTP-bound Gα subunit, and the ß1γ1 complex.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Guanosina Trifosfato/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Rodopsina/metabolismo , Transducina/metabolismo , Animales , Bovinos , Polarización de Fluorescencia , Subunidades alfa de la Proteína de Unión al GTP/genética , Técnicas In Vitro , Receptores Acoplados a Proteínas G/genética , Rodopsina/genética , Transducina/genética
14.
Biochemistry ; 50(6): 1070-80, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21033699

RESUMEN

Guanine nucleotide exchange factors (GEFs) activate Rho GTPases by catalyzing the exchange of bound GDP for GTP, thereby resulting in downstream effector recognition. Two metazoan families of GEFs have been described: Dbl-GEF family members that share conserved Dbl homology (DH) and Pleckstrin homology (PH) domains and the more recently described Dock180 family members that share little sequence homology with the Dbl family and are characterized by conserved Dock homology regions 1 and 2 (DHR-1 and -2, respectively). While extensive characterization of the Dbl family has been performed, less is known about how Dock180 family members act as GEFs, with only a single X-ray structure having recently been reported for the Dock9-Cdc42 complex. To learn more about the mechanisms used by the founding member of the family, Dock180, to act as a Rac-specific GEF, we set out to identify and characterize its limit functional GEF domain. A C-terminal portion of the DHR-2 domain, composed of approximately 300 residues (designated as Dock180(DHR-2c)), is shown to be necessary and sufficient for robust Rac-specific GEF activity both in vitro and in vivo. We further show that Dock180(DHR-2c) binds to Rac in a manner distinct from that of Rac-GEFs of the Dbl family. Specifically, Ala(27) and Trp(56) of Rac appear to provide a bipartite binding site for the specific recognition of Dock180(DHR-2c), whereas for Dbl family Rac-GEFs, Trp(56) of Rac is the sole primary determinant of GEF specificity. On the basis of our findings, we are able to define the core of Dock180 responsible for its Rac-GEF activity as well as highlight key recognition sites that distinguish different Dock180 family members and determine their corresponding GTPase specificities.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Proteínas de Unión al GTP rac/química , Sitios de Unión , Catálisis , Técnica del Anticuerpo Fluorescente Indirecta , Factores de Intercambio de Guanina Nucleótido/metabolismo , Estructura Terciaria de Proteína , Proteínas de Unión al GTP rac/metabolismo
15.
Methods Mol Biol ; 2009: 307-315, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31152413

RESUMEN

G protein-coupled receptors (GPCRs) comprise the largest family of transmembrane receptors and are targets for over 30% of all drugs on the market. Structural information of GPCRs and more importantly that of the complex between GPCRs and their signaling partner heterotrimeric G proteins is of great importance. Here we present a method for the large-scale purification of the rhodopsin-transducin complex, the GPCR-G protein signaling complex in visual phototransduction, directly from their native retinal membrane using native proteins purified from bovine retinae. Formation of the complex on native membrane is orchestrated in part by the proper engagement of lipid-modified rhodopsin and transducin (i.e., palmitoylation of the rhodopsin C-terminus, myristoylation and farnesylation of the αT and γ1, respectively). The resulting complex is of high purity and stability and has proved suitable for further biophysical and structural studies. The methods described here should be applicable to other recombinantly expressed receptors from insect cells or mamalian cells by forming stable, functional complexes directly on purified cell membranes.


Asunto(s)
Membrana Celular/química , Complejos Multiproteicos , Retina/química , Rodopsina , Transducina , Animales , Bovinos , Complejos Multiproteicos/química , Complejos Multiproteicos/aislamiento & purificación , Estructura Cuaternaria de Proteína , Rodopsina/química , Rodopsina/aislamiento & purificación , Transducina/química , Transducina/aislamiento & purificación
16.
Methods Mol Biol ; 2009: 317-324, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31152414

RESUMEN

Transmembrane proteins, such as G protein-coupled receptors (GPCR), require solubilization in detergents prior to purification. The recent development of novel detergents has allowed for the stabilization of GPCRs, which typically have a high degree of structural flexibility and are otherwise subject to denaturation. However, the detergent micelle environment is still very different from the native lipid membrane and the activity of GPCRs can be profoundly affected by interactions with annular lipid molecules. Moreover, GPCRs are often palmitoylated at their intracellular side, and a lipid bilayer environment would allow for proper orientation of these lipid modifications. Therefore, a reconstituted lipid bilayer environment would best mimic the physiological receptor microenvironment for biophysical studies of GPCRs and nanodiscs provide a methodology to address this aim. Nanodiscs are lipid bilayer discs stabilized by amphipathic membrane scaffolding proteins (MSP) where detergent-solubilized transmembrane proteins can be incorporated into them through a self-assembly process. Here we present a method for reconstituting the purified detergent-solubilized rhodopsin-transducin complex, the GPCR-G protein complex in visual phototransduction, into nanodiscs. The resulting complex incorporated into lipid nanodiscs can be used in biophysical studies including small-angle X-ray scattering and electron microscopy. This method is applicable to integral membrane proteins that mediate protein lipidation, including the zDHHC-family of S-acyltransferases and membrane-bound O-acyltransferases.


Asunto(s)
Membrana Dobles de Lípidos/química , Nanoestructuras/química , Rodopsina/química , Transducina/química , Animales , Detergentes/química
17.
Cell Rep ; 29(1): 76-88.e7, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31577957

RESUMEN

Efforts to target glutamine metabolism for cancer therapy have focused on the glutaminase isozyme GLS. The importance of the other isozyme, GLS2, in cancer has remained unclear, and it has been described as a tumor suppressor in some contexts. Here, we report that GLS2 is upregulated and essential in luminal-subtype breast tumors, which account for >70% of breast cancer incidence. We show that GLS2 expression is elevated by GATA3 in luminal-subtype cells but suppressed by promoter methylation in basal-subtype cells. Although luminal breast cancers resist GLS-selective inhibitors, we find that they can be targeted with a dual-GLS/GLS2 inhibitor. These results establish a critical role for GLS2 in mammary tumorigenesis and advance our understanding of how to target glutamine metabolism in cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glutaminasa/metabolismo , Hígado/metabolismo , Animales , Neoplasias de la Mama/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular , Línea Celular Tumoral , Metilación de ADN/genética , Femenino , Factor de Transcripción GATA3/metabolismo , Genes Supresores de Tumor/fisiología , Glutamina/metabolismo , Células HEK293 , Humanos , Células MCF-7 , Ratones , Regiones Promotoras Genéticas/genética
18.
Nat Commun ; 8: 15163, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28516903

RESUMEN

Heterotrimeric G proteins are quintessential signalling switches activated by nucleotide exchange on Gα. Although activation is predominantly carried out by G-protein-coupled receptors (GPCRs), non-receptor guanine-nucleotide exchange factors (GEFs) have emerged as critical signalling molecules and therapeutic targets. Here we characterize the molecular mechanism of G-protein activation by a family of non-receptor GEFs containing a Gα-binding and -activating (GBA) motif. We combine NMR spectroscopy, computational modelling and biochemistry to map changes in Gα caused by binding of GBA proteins with residue-level resolution. We find that the GBA motif binds to the SwitchII/α3 cleft of Gα and induces changes in the G-1/P-loop and G-2 boxes (involved in phosphate binding), but not in the G-4/G-5 boxes (guanine binding). Our findings reveal that G-protein-binding and activation mechanisms are fundamentally different between GBA proteins and GPCRs, and that GEF-mediated perturbation of nucleotide phosphate binding is sufficient for Gα activation.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Guanosina Difosfato/metabolismo , Proteínas de Microfilamentos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Secuencias de Aminoácidos/fisiología , Línea Celular , Activación Enzimática/fisiología , Células HEK293 , Humanos , Resonancia Magnética Nuclear Biomolecular , Unión Proteica/fisiología , Transducción de Señal/fisiología
20.
PLoS One ; 8(9): e74783, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098668

RESUMEN

Glutaminase C is a key metabolic enzyme, which is unregulated in many cancer systems and believed to play a central role in the Warburg effect, whereby cancer cells undergo changes to an altered metabolic profile. A long-standing hypothesis links enzymatic activity to the protein oligomeric state, hence the study of the solution behavior in general and the oligomer state in particular of glutaminase C is important for the understanding of the mechanism of protein activation and inhibition. In this report, this is extensively investigated in correlation to enzyme concentration or phosphate level, using a high-throughput microfluidic-mixing chip for the SAXS data collection, and we confirm that the oligomeric state correlates with activity. The in-depth solution behavior analysis further reveals the structural behavior of flexible regions of the protein in the dimeric, tetrameric and octameric state and investigates the C-terminal influence on the enzyme solution behavior. Our data enable SAXS-based rigid body modeling of the full-length tetramer states, thereby presenting the first ever experimentally derived structural model of mitochondrial glutaminase C including the N- and C-termini of the enzyme.


Asunto(s)
Glutaminasa/química , Proteínas Mitocondriales/química , Modelos Moleculares , Conformación Proteica , Área Bajo la Curva , Glutaminasa/genética , Humanos , Microfluídica , Proteínas Mitocondriales/genética , Oligonucleótidos/genética , Dispersión del Ángulo Pequeño
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA