Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(46): 29090-29100, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33122432

RESUMEN

TRPM3 channels play important roles in the detection of noxious heat and in inflammatory thermal hyperalgesia. The activity of these ion channels in somatosensory neurons is tightly regulated by µ-opioid receptors through the signaling of Gßγ proteins, thereby reducing TRPM3-mediated pain. We show here that Gßγ directly binds to a domain of 10 amino acids in TRPM3 and solve a cocrystal structure of this domain together with Gßγ. Using these data and mutational analysis of full-length proteins, we pinpoint three amino acids in TRPM3 and their interacting partners in Gß1 that are individually necessary for TRPM3 inhibition by Gßγ. The 10-amino-acid Gßγ-interacting domain in TRPM3 is subject to alternative splicing. Its inclusion in or exclusion from TRPM3 channel proteins therefore provides a mechanism for switching on or off the inhibitory action that Gßγ proteins exert on TRPM3 channels.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Canales Catiónicos TRPM/química , Canales Catiónicos TRPM/efectos de los fármacos , Canales Catiónicos TRPM/metabolismo , Sitios de Unión , Calcio/metabolismo , Subunidades beta de la Proteína de Unión al GTP/química , Subunidades gamma de la Proteína de Unión al GTP/química , Células HEK293 , Humanos , Hiperalgesia/metabolismo , Modelos Moleculares , Mutación , Neuronas/metabolismo , Dolor/metabolismo , Receptores Opioides/metabolismo , Canales Catiónicos TRPM/genética
2.
Elife ; 62017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28829742

RESUMEN

Transient receptor potential melastatin 3 (TRPM3) channels are activated by heat, and chemical ligands such as pregnenolone sulphate (PregS) and CIM0216. Here, we show that activation of receptors coupled to heterotrimeric Gi/o proteins inhibits TRPM3 channels. This inhibition was alleviated by co-expression of proteins that bind the ßγ subunits of heterotrimeric G-proteins (Gßγ). Co-expression of Gßγ, but not constitutively active Gαi or Gαo, inhibited TRPM3 currents. TRPM3 co-immunoprecipitated with Gß, and purified Gßγ proteins applied to excised inside-out patches inhibited TRPM3 currents, indicating a direct effect. Baclofen and somatostatin, agonists of Gi-coupled receptors, inhibited Ca2+ signals induced by PregS and CIM0216 in mouse dorsal root ganglion (DRG) neurons. The GABAB receptor agonist baclofen also inhibited inward currents induced by CIM0216 in DRG neurons, and nocifensive responses elicited by this TRPM3 agonist in mice. Our data uncover a novel signaling mechanism regulating TRPM3 channels.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Canales Catiónicos TRPM/efectos de los fármacos , Animales , Baclofeno/antagonistas & inhibidores , Escala de Evaluación de la Conducta , Calcio/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/farmacología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Células HEK293 , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Pregnenolona/farmacología , Somatostatina/antagonistas & inhibidores
3.
Elife ; 62017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28826482

RESUMEN

Opioids, agonists of µ-opioid receptors (µORs), are the strongest pain killers clinically available. Their action includes a strong central component, which also causes important adverse effects. However, µORs are also found on the peripheral endings of nociceptors and their activation there produces meaningful analgesia. The cellular mechanisms downstream of peripheral µORs are not well understood. Here, we show in neurons of murine dorsal root ganglia that pro-nociceptive TRPM3 channels, present in the peripheral parts of nociceptors, are strongly inhibited by µOR activation, much more than other TRP channels in the same compartment, like TRPV1 and TRPA1. Inhibition of TRPM3 channels occurs via a short signaling cascade involving Gßγ proteins, which form a complex with TRPM3. Accordingly, activation of peripheral µORs in vivo strongly attenuates TRPM3-dependent pain. Our data establish TRPM3 inhibition as important consequence of peripheral µOR activation indicating that pharmacologically antagonizing TRPM3 may be a useful analgesic strategy.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Receptores Opioides mu/metabolismo , Canales Catiónicos TRPM/efectos de los fármacos , Analgésicos Opioides/agonistas , Animales , Escala de Evaluación de la Conducta , Calcio/metabolismo , Señalización del Calcio/fisiología , Ganglios Espinales/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Nociceptores/fisiología , Dolor/metabolismo , Receptores Opioides/metabolismo , Canal Catiónico TRPA1/metabolismo , Canales Catiónicos TRPV/metabolismo
4.
J Pharmacol Exp Ther ; 352(1): 148-55, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25339760

RESUMEN

The α1-subunit containing glycine receptors (GlyRs) is potentiated by ethanol, in part, by intracellular Gßγ actions. Previous studies have suggested that molecular requirements in the large intracellular domain are involved; however, the lack of structural data about this region has made it difficult to describe a detailed mechanism. Using circular dichroism and molecular modeling, we generated a full model of the α1-GlyR, which includes the large intracellular domain and provides new information on structural requirements for allosteric modulation by ethanol and Gßγ. The data strongly suggest the existence of an α-helical conformation in the regions near transmembrane (TM)-3 and TM4 of the large intracellular domain. The secondary structure in the N-terminal region of the large intracellular domain near TM3 appeared critical for ethanol action, and this was tested using the homologous domain of the γ2-subunit of the GABAA receptor predicted to have little helical conformation. This region of γ2 was able to bind Gßγ and form a functional channel when combined with α1-GlyR, but it was not sensitive to ethanol. Mutations in the N- and C-terminal regions introduced to replace corresponding amino acids of the α1-GlyR sequence restored the ability to be modulated by ethanol and Gßγ. Recovery of the sensitivity to ethanol was associated with the existence of a helical conformation similar to α1-GlyR, thus being an essential secondary structural requirement for GlyR modulation by ethanol and G protein.


Asunto(s)
Etanol/farmacología , Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Espacio Intracelular/metabolismo , Receptores de Glicina/química , Receptores de Glicina/metabolismo , Secuencia de Aminoácidos , Animales , Relación Dosis-Respuesta a Droga , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Ratas , Receptores de GABA-A/metabolismo
5.
Int J Biochem Cell Biol ; 42(6): 1052-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20348012

RESUMEN

The hematopoietic-specific G(q) subfamily members, Galpha(16) and Galpha(14) proteins have recently been shown to be capable of stimulating the signal transducer and activator of transcription 3 (STAT3) as well as STAT1. In the present study we examined whether this activation was STAT-member specific as well as determining the possible involvement of Gbetagamma dimers. Despite clear stimulation of STAT3, the constitutively active mutants of Galpha(16) (Galpha(16)QL) and Galpha(14) (Galpha(14)QL) failed to induce the phosphorylation of several STAT family members, including STAT2, STAT4 and STAT5 in human embryonic kidney 293 cells. On the other hand, transient expression of specific combinations of Gbetagamma complexes induced STAT3 phosphorylation. Among the 48 combinations tested, 13 permutations of Gbetagamma stimulated STAT3 phosphorylation and all of them contain the neuronal-specific Ggamma(2), Ggamma(4), Ggamma(7) and Ggamma(9). These results suggested that the activation of STAT family members by Galpha(16) or Galpha(14) was selective and that distinct combinations of Gbetagamma complexes can also regulate the STAT signaling pathway.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Factor de Transcripción STAT3/metabolismo , Dimerización , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/farmacología , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Células HeLa , Hematopoyesis , Humanos , Mutación/genética , Neuronas/metabolismo , Fosforilación , Unión Proteica , Transducción de Señal , Transgenes/genética
6.
J Pharmacol Exp Ther ; 333(2): 393-403, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20110378

RESUMEN

Signaling through G protein-coupled receptors (GPCRs) promotes breast cancer metastasis. G proteins convey GPCR signals by dissociating into Galpha and Gbetagamma subunits. The aim of the present study was to determine whether blockade of Gbetagamma signaling suppresses breast cancer cell migration and invasion, which are critical components of metastasis. Conditioned media (CM) of NIH-3T3 fibroblasts are widely used as chemoattractants in in vitro cancer metastasis studies. Expression of a Gbetagamma scavenger peptide attenuated NIH-3T3 CM-induced migration and invasion of both metastatic breast cancer MDA-MB-231 and MDA-MB-436 cells by 40 to 50% without effects on cell viability. Migration and invasion of cells in response to NIH-3T3 CM were also blocked by 8-(4,5,6-trihydroxy-3-oxo-3H-xanthen-9-yl)-1-naph-thalene-carboxylic acid) (M119K), a Gbetagamma inhibitor, with maximum inhibition exceeding 80% and half-maximal inhibitory concentration (IC50) values of 1 to 2 microM. M119K also attenuated Rac-dependent formation of lamellipodia, a key structure required for metastasis. Constitutively active Rac1 rescued Gbetagamma blockade-mediated inhibition of breast cancer cell migration, whereas dominant negative Rac1 inhibited cell migration similar to Gbetagamma blockade. Furthermore, M119K suppressed Gi protein-coupled CXC chemokine receptor 4 (CXCR4)-dependent MDA-MB-231 cell migration by 80% with an IC50 value of 1 microM, whereas tyrosine kinase receptor-dependent cell migration was significantly less inhibited. However, CXCR4-dependent inhibition of adenylyl cyclase, a Gialpha-mediated response in MDA-MB-231 cells, was not blocked by M119K but was blocked by pertussis toxin, which selectively inactivates Gialpha. This report is the first to directly demonstrate the role of Gbetagamma in cancer cell migration and invasion and suggests that targeting Gbetagamma signaling pathways may provide a novel strategy for suppressing breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Movimiento Celular/fisiología , Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Invasividad Neoplásica/fisiopatología , Adenilil Ciclasas/efectos de los fármacos , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Ciclohexanos/farmacología , Femenino , Humanos , Microscopía Fluorescente , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/fisiopatología , Fragmentos de Péptidos/fisiología , Seudópodos/efectos de los fármacos , Receptores CXCR4/fisiología , Proteínas Recombinantes , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Xantenos/farmacología , Proteínas de Unión al GTP rac/efectos de los fármacos , Proteínas de Unión al GTP rac/fisiología
7.
Blood ; 108(1): 160-6, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16537812

RESUMEN

Vascular endothelial growth factor (VEGF) is a major mediator of pathologic angiogenesis, a process necessary for the formation of new blood vessels to support tumor growth. Historically, VEGF has been thought to signal via receptor tyrosine kinases, which are not typically considered to be G protein dependent. Here, we show that targeted knockdown of the G protein gng2 gene (Ggamma2) blocks the normal angiogenic process in developing zebrafish embryos. Moreover, loss of gng2 function inhibits the ability of VEGF to promote the angiogenic sprouting of blood vessels by attenuating VEGF induced phosphorylation of phospholipase C-gamma1 (PLCgamma1) and serine/threonine kinase (AKT). Collectively, these results demonstrate a novel interaction between Ggamma2- and VEGF-dependent pathways to regulate the angiogenic process in a whole-animal model. Blocking VEGF function using a humanized anti-VEGF antibody has emerged as a promising treatment for colorectal, non-small lung cell, and breast cancers. However, this treatment may cause considerable side effects. Our findings provide a new opportunity for cotargeting G protein- and VEGF-dependent pathways to synergistically block pathologic angiogenesis, which may lead to a safer and more efficacious therapeutic regimen to fight cancer.


Asunto(s)
Subunidades gamma de la Proteína de Unión al GTP/farmacología , Proteínas de Unión al GTP/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Subunidades de Proteína/farmacología , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Proteínas de Pez Cebra/farmacología , Animales , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/fisiología , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/fisiología , Perfilación de la Expresión Génica , Modelos Animales , Neovascularización Fisiológica/fisiología , Fenotipo , Fosfolipasa C gamma/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
8.
Cell Signal ; 18(1): 62-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15925485

RESUMEN

The Ca2+-activated adenylyl cyclase type VIII (AC-VIII) has been implicated in several forms of neural plasticity, including drug addiction and learning and memory. It has not been clear whether Gi/o proteins and G-protein coupled receptors regulate the activity of AC-VIII. Here we show in intact mammalian cell system that AC-VIII is inhibited by mu-opioid receptor activation and that this inhibition is pertussis toxin sensitive. Moreover, we show that G(betagamma) subunits inhibit AC-VIII activity, while constitutively active alphai/o subunits do not. Different Gbeta isoforms varied in their efficacies, with Gbeta1gamma2 or Gbeta2gamma2 being more efficient than Gbeta3gamma2 and Gbeta4gamma2, while Gbeta5 (transfected with gamma2) had no effect. As for the Ggamma subunits, Gbeta1 inhibited AC-VIII activity in the presence of all gamma subunits tested except for gamma5 that had only a marginal activity. Moreover, cotransfection with proteins known to serve as scavengers of Gbetagamma dimers, or to reduce Gbetagamma plasma membrane anchorage, markedly attenuated the mu-opioid receptor-induced inhibition of AC-VIII. These results demonstrate that Gbetagamma (originating from agonist activation of these receptors) and probably not Galphai/o subunits are involved in the agonist inhibition of AC-VIII.


Asunto(s)
Adenilil Ciclasas/metabolismo , Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Inhibidores de Adenilato Ciclasa , Animales , Células COS , Chlorocebus aethiops , Colforsina/farmacología , Dimerización , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Ionomicina/farmacología , Morfina/farmacología , Toxina del Pertussis/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo
9.
Nat Neurosci ; 8(4): 421-5, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15778713

RESUMEN

The activation of G protein-coupled receptors (GPCRs) can result in an inhibition of Ca(2+)-dependent hormone and neurotransmitter secretion. This has been attributed in part to G protein inhibition of Ca(2+) influx. However, a frequently dominant inhibitory effect, of unknown mechanism, also occurs distal to Ca(2+) entry. Here we characterize direct inhibitory actions of G protein betagamma (Gbetagamma) on Ca(2+)-triggered vesicle exocytosis in permeable PC12 cells. Gbetagamma inhibition was rapid (<1 s) and was attenuated by cleavage of synaptosome-associated protein of 25 kD (SNAP25). Gbetagamma bound soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes, and binding was reduced to SNARE complexes containing cleaved SNAP25 or by Ca(2+)-dependent synaptotagmin binding. Here we show inhibitory coupling between GPCRs and vesicle exocytosis mediated directly by Gbetagamma interactions with the Ca(2+)-dependent fusion machinery.


Asunto(s)
Exocitosis/fisiología , Subunidades beta de la Proteína de Unión al GTP/fisiología , Subunidades gamma de la Proteína de Unión al GTP/fisiología , Vesículas Secretoras/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Western Blotting/métodos , Calcio/farmacología , Proteínas de Unión al Calcio/metabolismo , Bovinos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Exocitosis/efectos de los fármacos , Proteínas del Ojo/farmacología , Reguladores de Proteínas de Unión al GTP , Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/farmacología , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/farmacología , Inhibición Neural/efectos de los fármacos , Norepinefrina/metabolismo , Células PC12 , Fosfoproteínas/farmacología , Unión Proteica/efectos de los fármacos , Ratas , Tiempo de Reacción/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Proteínas SNARE , Vesículas Secretoras/efectos de los fármacos , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida , Proteína 25 Asociada a Sinaptosomas , Sinaptotagminas , Factores de Tiempo
10.
FEBS Lett ; 556(1-3): 187-92, 2004 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-14706848

RESUMEN

The p21-activated kinase (PAK) family is homologous to the yeast sterile 20 (Ste20) and regulates a wide variety of cellular responses, including cell morphology, proliferation, and survival. In this study we examined the activation of PAK1 by Gbetagamma subunits. Co-transfection of COS7 cells with Gbeta1gamma2 or Gbeta1gamma5 was sufficient to induce agonist-independent activation of PAK1. Expression of dominant/negative Rac, Cdc42, or Ras did not inhibit this Gbetagamma-dependent activation. Wortmannin, which inhibits phosphoinositide 3-kinase (PI3-kinase) activity, and expression of a dominant/negative form of Akt were sufficient to abrogate the activation of PAK1 that was induced by Gbetagamma. These results reveal that stimulation of PAK1 by Gbetagamma can occur via a PI3-kinase and Akt pathway that does not require Rac1 or Cdc42.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Androstadienos/farmacología , Animales , Western Blotting , Células COS , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Lisofosfolípidos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Pruebas de Precipitina , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/agonistas , Proteínas Proto-Oncogénicas c-akt , Transfección , Wortmanina , Quinasas p21 Activadas
11.
Circ Res ; 93(9): 848-56, 2003 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-14512443

RESUMEN

Rho GTPases integrate the intracellular signaling in a wide range of cellular processes. Activation of these G proteins is tightly controlled by a number of guanine nucleotide exchange factors (GEFs). In this study, we addressed the functional role of the recently identified p114RhoGEF in in vivo experiments. Activation of endogenous G protein-coupled receptors with lysophosphatidic acid resulted in activation of a transcription factor, serum response element (SRE), that was enhanced by p114RhoGEF. This stimulation was inhibited by the functional scavenger of Gbetagamma subunits, transducin. We have determined that Gbetagamma subunits but not Galpha subunits of heterotrimeric G proteins stimulated p114RhoGEF-dependent SRE activity. Using coimmunoprecipitation assay, we have determined that Gbetagamma subunits interacted with full-length and DH/PH domain of p114RhoGEF. Similarly, Gbetagamma subunits stimulated SRE activity induced by full-length and DH/PH domain of p114RhoGEF. Using in vivo pull-down assays and dominant-negative mutants of Rho GTPases, we have determined that p114RhoGEF activated RhoA and Rac1 but not Cdc42 proteins. Functional significance of RhoA activation was established by the ability of p114RhoGEF to induce actin stress fibers and cell rounding. Functional significance of Rac1 activation was established by the ability of p114RhoGEF to induce production of reactive oxygen species (ROS) followed by activation of NADPH oxidase enzyme complex. In summary, our data showed that the novel guanine nucleotide exchange factor p114RhoGEF regulates the activity of RhoA and Rac1, and that Gbetagamma subunits of heterotrimeric G proteins are activators of p114RhoGEF under physiological conditions. The findings help to explain the integrated effects of LPA and other G-protein receptor-coupled agonists on actin stress fiber formation, cell shape change, and ROS production.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Empalme Alternativo , Animales , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Tamaño de la Célula/fisiología , Subunidades beta de la Proteína de Unión al GTP/farmacología , Subunidades gamma de la Proteína de Unión al GTP/farmacología , Genes Reporteros , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Lisofosfolípidos/farmacología , Ratones , NADPH Oxidasas/metabolismo , Células 3T3 NIH , Especificidad de Órganos , Fosfoproteínas/metabolismo , Estructura Terciaria de Proteína/fisiología , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Elemento de Respuesta al Suero/genética , Fibras de Estrés/efectos de los fármacos , Fibras de Estrés/metabolismo , Trombina/farmacología , Proteína de Unión al GTP cdc42/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA