Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 10(8)2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34440613

RESUMEN

Sepsis and septic shock are associated with acute and sustained impairment in the function of the cardiovascular system, kidneys, lungs, liver, and brain, among others. Despite the significant advances in prevention and treatment, sepsis and septic shock sepsis remain global health problems with elevated mortality rates. Rho proteins can interact with a considerable number of targets, directly affecting cellular contractility, actin filament assembly and growing, cell motility and migration, cytoskeleton rearrangement, and actin polymerization, physiological functions that are intensively impaired during inflammatory conditions, such as the one that occurs in sepsis. In the last few decades, Rho proteins and their downstream pathways have been investigated in sepsis-associated experimental models. The most frequently used experimental design included the exposure to bacterial lipopolysaccharide (LPS), in both in vitro and in vivo approaches, but experiments using the cecal ligation and puncture (CLP) model of sepsis have also been performed. The findings described in this review indicate that Rho proteins, mainly RhoA and Rac1, are associated with the development of crucial sepsis-associated dysfunction in different systems and cells, including the endothelium, vessels, and heart. Notably, the data found in the literature suggest that either the inhibition or activation of Rho proteins and associated pathways might be desirable in sepsis and septic shock, accordingly with the cellular system evaluated. This review included the main findings, relevance, and limitations of the current knowledge connecting Rho proteins and sepsis-associated experimental models.


Asunto(s)
Sepsis/enzimología , Choque Séptico/enzimología , Proteínas de Unión al GTP rho/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Terapia Molecular Dirigida , Sepsis/tratamiento farmacológico , Sepsis/patología , Choque Séptico/tratamiento farmacológico , Choque Séptico/patología , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
2.
ACS Chem Biol ; 13(6): 1514-1524, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29746086

RESUMEN

Ras and Ras-related small GTPases are key regulators of diverse cellular functions that impact cell growth, survival, motility, morphogenesis, and differentiation. They are important targets for studies of disease mechanisms as well as drug discovery. Here, we report the characterization of small molecule agonists of one or more of six Rho, Rab, and Ras family GTPases that were first identified through flow cytometry-based, multiplexed high-throughput screening of 200000 compounds. The activators were categorized into three distinct chemical families that are represented by three lead compounds having the highest activity. Virtual screening predicted additional compounds with potential GTPase activating properties. Secondary dose-response assays performed on compounds identified through these screens confirmed agonist activity of 43 compounds. While the lead and second most active small molecules acted as pan activators of multiple GTPase subfamilies, others showed partial selectivity for Ras and Rab proteins. The compounds did not stimulate nucleotide exchange by guanine nucleotide exchange factors and did not protect against GAP-stimulated GTP hydrolysis. The activating properties were caused by a reversible stabilization of the GTP-bound state and prolonged effector protein interactions. Notably, these compounds were active both in vitro and in cell-based assays, and small molecule-mediated changes in Rho GTPase activities were directly coupled to measurable changes in cytoskeletal rearrangements that dictate cell morphology.


Asunto(s)
Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas de Unión al GTP rho/agonistas , Actinas/metabolismo , Animales , Activación Enzimática/efectos de los fármacos , Pruebas de Enzimas , Células HeLa , Humanos , Ratones , Estructura Molecular , Ratas , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Células 3T3 Swiss
3.
PLoS One ; 12(2): e0172613, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28222172

RESUMEN

OBJECTIVE: Epidermal stem cells (ESCs) play a critical role in wound healing, but the mechanism underlying ESC proliferation is not well defined. Here, we explore the effects of RhoA on ESC proliferation and the possible underlying mechanism. METHODS: Human ESCs were enriched by rapid adhesion to collagen IV. RhoA(+/+)(G14V), RhoA(-/-)(T19N) and pGFP control plasmids were transfected into human ESCs. The effect of RhoA on cell proliferation was detected by cell proliferation and DNA synthesis assays. Induction of PKN1 activity by RhoA was determined by immunoblot analysis, and the effects of PKN1 on RhoA in terms of inducing cell proliferation and cyclin D1 expression were detected using specific siRNA targeting PKN1. The effects of U-46619 (a RhoA agonist) and C3 transferase (a RhoA antagonist) on ESC proliferation were observed in vivo. RESULTS: RhoA had a positive effect on ESC proliferation, and PKN1 activity was up-regulated by the active RhoA mutant (G14V) and suppressed by RhoA T19N. Moreover, the ability of RhoA to promote ESC proliferation and DNA synthesis was interrupted by PKN1 siRNA. Additionally, cyclin D1 protein and mRNA expression levels were up-regulated by RhoA G14V, and these effects were inhibited by siRNA-mediated knock-down of PKN1. RhoA also promoted ESC proliferation via PKN in vivo. CONCLUSION: This study shows that the effect of RhoA on ESC proliferation is mediated by activation of the PKN1-cyclin D1 pathway in vitro, suggesting that RhoA may serve as a new therapeutic target for wound healing.


Asunto(s)
Ciclina D1/fisiología , Células Epidérmicas , Células Epiteliales/metabolismo , Proteína Quinasa C/fisiología , Transducción de Señal/fisiología , Células Madre/metabolismo , Cicatrización de Heridas , Proteína de Unión al GTP rhoA/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , ADP Ribosa Transferasas/farmacología , Animales , Toxinas Botulínicas/farmacología , Quemaduras/fisiopatología , Quemaduras/terapia , Carbazoles/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclina D1/biosíntesis , Ciclina D1/genética , Replicación del ADN/efectos de los fármacos , Células Epiteliales/citología , Humanos , Alcaloides Indólicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Cultivo Primario de Células , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Interferencia de ARN , ARN Interferente Pequeño , Distribución Aleatoria , Células Madre/citología , Transfección , Cicatrización de Heridas/fisiología , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/deficiencia , Proteína de Unión al GTP rhoA/genética
4.
Thromb Haemost ; 116(3): 506-16, 2016 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-27345948

RESUMEN

Leukemia-Associated RhoGEF (LARG) is highly expressed in platelets, which are essential for maintaining normal haemostasis. We studied the function of LARG in murine and human megakaryocytes and platelets with Larg knockout (KO), shRNA-mediated knockdown and small molecule-mediated inhibition. We found that LARG is important for human, but not murine, megakaryocyte maturation. Larg KO mice exhibit macrothrombocytopenia, internal bleeding in the ovaries and prolonged bleeding times. KO platelets have impaired aggregation, α-granule release and integrin α2bß3 activation in response to thrombin and thromboxane, but not to ADP. The same agonist-specific reductions in platelet aggregation occur in human platelets treated with a LARG inhibitor. Larg KO platelets have reduced RhoA activation and myosin light chain phosphorylation, suggesting that Larg plays an agonist-specific role in platelet signal transduction. Using two different in vivo assays, Larg KO mice are protected from in vivo thrombus formation. Together, these results establish that LARG regulates human megakaryocyte maturation, and is critical for platelet function in both humans and mice.


Asunto(s)
Plaquetas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/sangre , Proteínas de Unión al GTP rho/sangre , Proteína de Unión al GTP rhoA/sangre , Animales , Tiempo de Sangría , Plaquetas/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Ligeras de Miosina/sangre , Pruebas de Función Plaquetaria , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido Rho/deficiencia , Factores de Intercambio de Guanina Nucleótido Rho/genética , Trombina/metabolismo , Trombina/farmacología , Trombopoyesis/genética , Trombopoyesis/fisiología , Tromboxanos/sangre , Tromboxanos/farmacología , Proteínas de Unión al GTP rho/agonistas , Proteína de Unión al GTP rhoA/agonistas
5.
Neuropharmacology ; 109: 357-365, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27350290

RESUMEN

Recent evidence suggests an early involvement of dopaminergic (DA) processes and terminals in Parkinson's disease (PD). The arborization of neurons depends on the actin cytoskeleton, which in turn is regulated by small GTPases of the Rho family, encompassing Rho, Rac and Cdc42 subfamilies. Indeed, some reports point to a role for Rac and Cdc42 signaling in the pathophysiology of inherited parkinsonisms. We thus investigated the potential therapeutic effect of the modulation of cerebral Rho GTPases in PD. Cytotoxic necrotizing factor 1 (CNF1), a 114 kDa protein toxin produced by Escherichia coli, permanently activates RhoA, Rac1 and Cdc42 in intact cells. We report that the modulation of Rho GTPases by CNF1 results in hypertrophy of DA cell processes of cultured substantia nigra neurons, including increase in length, branching and varicosity. In vivo, the treatment corrects long-standing motor and biochemical asymmetries and restores degenerated nigrostriatal DA tissue after 6-hydroxydopamine lesion. We conclude that the pharmacological modulation of Rho GTPases shows neurorestorative potential and represents a promising avenue in the treatment PD. The study also suggests that naturally occurring molecules acting on Rho GTPase signaling, such as some bacterial protein toxins, might play a role in the development of PD.


Asunto(s)
Toxinas Bacterianas/uso terapéutico , Proteínas de Escherichia coli/uso terapéutico , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/enzimología , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/metabolismo , Animales , Toxinas Bacterianas/farmacología , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Proteínas de Escherichia coli/farmacología , Masculino , Neuronas/efectos de los fármacos , Neuronas/enzimología , Ratas , Ratas Wistar , Sustancia Negra/efectos de los fármacos , Sustancia Negra/enzimología , Resultado del Tratamiento
6.
Hum Reprod ; 31(4): 723-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26874360

RESUMEN

STUDY QUESTION: Is phosphatase of regenerating liver-3 (PRL-3) associated with increased motility of endometriotic cells from endometrioma? SUMMARY ANSWER: Elevated PRL-3 promotes cytoskeleton reorganization, cell migration and invasion of endometrial stromal cells (ESCs) from endometrioma. WHAT IS KNOWN ALREADY: Overexpression of PRL-3 is associated with cancer cell migration, invasion and metastatic phenotype. STUDY DESIGN, SIZE, DURATION: Primary human ESCs were isolated from eutopic endometrium of women without endometriosis (EuCo, n = 10), with histologically proven endometrioma (EuEM, n = 19) and from the cyst wall of ovarian endometriosis (OvEM, n = 26). PARTICIPANTS/MATERIALS, SETTING, METHODS: The expression of PRL-3 in ESCs derived from EuCo, EuEM and OvEM at different phases of menstrual cycle were compared. The protein and mRNA levels of PRL-3 were examined by western blot and RT-qPCR, respectively. ESCs from OvEM were transfected with/without short hairpin RNA (shRNA) or small interfering RNA (siRNA). Additionally, a plasmid-mediated delivery system was used to achieve PRL-3 overexpression in ESCs from EuEM. The cellular distribution of F-actin and α-tubulin were examined by immunocytochemistry. Cell motility was evaluated by a transwell migration/invasion assay. MAIN RESULTS AND THE ROLE OF CHANCE: The protein and mRNA levels of PRL-3 are significantly elevated in ESCs from OvEM compared with EuCo and EuEM. The expression of PRL-3 was not altered between proliferative phase and secretory phase in ESCs from all groups. Knockdown of PRL-3 significantly modified the distribution of F-actin and α-tubulin cytoskeleton, inhibited cell migration and invasion. Endogenous inhibition of PRL-3 attenuated the expression of Ras homolog gene family members A and C (RhoA, RhoC), Rho-associated coiled-coil-containing protein kinase 1 (ROCK1) and matrix metalloproteinase (MMP) 9, but not MMP2 in ESCs from OvEM. Additionally, overexpression of PRL-3 in ESCs from EuEM up-regulates cell migration and invasion, and increases the expression of RhoA, RhoC, ROCK1 and MMP9. LIMITATIONS, REASONS FOR CAUTION: Lack of in vivo animal studies is the major limitation of our report. Our results should be further confirmed in a larger cohort of patients and extended to include eutopic and ectopic endometrium from patients with peritoneal endometriosis at different stages of the disease. WIDER IMPLICATIONS OF THE FINDINGS: Our study describes that elevated expression of PRL-3 contributes to the cell motility of ESCs from endometrioma. The results emphasize the importance of metastatic-related factor PRL-3 in the pathogenesis of endometrioma. STUDY FUNDING/COMPETING INTEREST: This work was supported by National Natural Science Foundation of China (No. 81170546) and Zhejiang Medicine Science and Technology Projects (No. Y13H040003). The authors declare no conflict of interest.


Asunto(s)
Citoesqueleto/metabolismo , Endometriosis/metabolismo , Endometrio/metabolismo , Inducción Enzimática , Proteínas de Neoplasias/metabolismo , Enfermedades del Ovario/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Células del Estroma/metabolismo , Movimiento Celular , Células Cultivadas , Citoesqueleto/patología , Endometriosis/patología , Endometrio/citología , Endometrio/patología , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Metaloproteinasa 9 de la Matriz/química , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ciclo Menstrual/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Enfermedades del Ovario/patología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células del Estroma/citología , Células del Estroma/patología , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/química , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/agonistas , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Proteína rhoC de Unión a GTP
7.
J Biol Chem ; 291(7): 3552-68, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26683376

RESUMEN

Both type 1 and type 2 diabetes are associated with increased risk of cardiovascular disease. This is in part attributed to the effects of hyperglycemia on vascular endothelial and smooth muscle cells, but the underlying mechanisms are not fully understood. In diabetic animal models, hyperglycemia results in hypercontractility of vascular smooth muscle possibly due to increased activation of Rho-kinase. The aim of the present study was to investigate the regulation of contractile smooth muscle markers by glucose and to determine the signaling pathways that are activated by hyperglycemia in smooth muscle cells. Microarray, quantitative PCR, and Western blot analyses revealed that both mRNA and protein expression of contractile smooth muscle markers were increased in isolated smooth muscle cells cultured under high compared with low glucose conditions. This effect was also observed in hyperglycemic Akita mice and in diabetic patients. Elevated glucose activated the protein kinase C and Rho/Rho-kinase signaling pathways and stimulated actin polymerization. Glucose-induced expression of contractile smooth muscle markers in cultured cells could be partially or completely repressed by inhibitors of advanced glycation end products, L-type calcium channels, protein kinase C, Rho-kinase, actin polymerization, and myocardin-related transcription factors. Furthermore, genetic ablation of the miR-143/145 cluster prevented the effects of glucose on smooth muscle marker expression. In conclusion, these data demonstrate a possible link between hyperglycemia and vascular disease states associated with smooth muscle contractility.


Asunto(s)
Aterosclerosis/metabolismo , Angiopatías Diabéticas/metabolismo , Regulación de la Expresión Génica , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Transducción de Señal , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Anciano , Animales , Aterosclerosis/enzimología , Aterosclerosis/patología , Células Cultivadas , Proteínas Contráctiles/agonistas , Proteínas Contráctiles/genética , Proteínas Contráctiles/metabolismo , Proteínas del Citoesqueleto/agonistas , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Angiopatías Diabéticas/enzimología , Angiopatías Diabéticas/patología , Humanos , Masculino , Ratones Noqueados , Ratones Mutantes , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Proteína Quinasa C/química , Proteína Quinasa C/metabolismo , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/química , Quinasas Asociadas a rho/metabolismo
8.
PLoS One ; 8(10): e76541, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098526

RESUMEN

Peroxisome proliferator activator receptors (PPAR) ligands such as 15-Δ12,13-prostaglandin L(2) [PJ] and all trans retinoic acid (ATRA) have been shown to inhibit the development of liver fibrosis. The role of ligands of retinoic X receptor (RXR) and its ligand, 9-cis, is less clear. The purpose of this study was to investigate the effects of combined treatment of the three ligends, PJ, ATRA and 9-cis, on key events during liver fibrosis in rat primary hepatic stellate cells (HSCs). We found that the anti-proliferative effect of the combined treatment of PJ, ATRA and 9-cis on HSCs was additive. Further experiments revealed that this inhibition was due to cell cycle arrest at the G0/G1 phase as demonstrated by FACS analysis. In addition, the combined treatment reduced cyclin D1 expression and increased p21 and p27 protein levels. Furthermore, we found that the three ligands down regulated the phosphorylation of mTOR and p70(S6K). The activation of HSCs was also inhibited by the three ligands as shown by inhibition of vitamin A lipid droplets depletion from HSCs. Studies using real time PCR and western blot analysis showed marked inhibition of collagen Iα1 and αSMA by the combination of the three ligands. These findings suggest that the combined use of PJ, ATRA and 9-cis causes inhibition of cell proliferation by cell cycle arrest and down-regulation of fibrotic markers to a greater extent compared to each of the ligands alone.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Prostaglandina D2/análogos & derivados , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Actinas/antagonistas & inhibidores , Actinas/genética , Actinas/metabolismo , Alitretinoína , Animales , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/antagonistas & inhibidores , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Ciclina D1/antagonistas & inhibidores , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/agonistas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Sinergismo Farmacológico , Fase G1/efectos de los fármacos , Fase G1/genética , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Masculino , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , PPAR gamma/metabolismo , Cultivo Primario de Células , Prostaglandina D2/farmacología , Ratas , Ratas Wistar , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptores X Retinoide/antagonistas & inhibidores , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
9.
PLoS One ; 7(4): e35883, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22558255

RESUMEN

BACKGROUND: Neurons extend their dendrites and axons to build functional neural circuits, which are regulated by both positive and negative signals during development. Brain-derived neurotrophic factor (BDNF) is a positive regulator for neurite outgrowth and neuronal survival but the functions of its precursor (proBDNF) are less characterized. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that proBDNF collapses neurite outgrowth in murine dorsal root ganglion (DRG) neurons and cortical neurons by activating RhoA via the p75 neurotrophin receptor (p75NTR). We demonstrated that the receptor proteins for proBDNF, p75NTR and sortilin, were highly expressed in cultured DRG or cortical neurons. ProBDNF caused a dramatic neurite collapse in a dose-dependent manner and this effect was about 500 fold more potent than myelin-associated glycoprotein. Neutralization of endogenous proBDNF by using antibodies enhanced neurite outgrowth in vitro and in vivo, but this effect was lost in p75NTR(-/-) mice. The neurite outgrowth of cortical neurons from p75NTR deficient (p75NTR(-/-)) mice was insensitive to proBDNF. There was a time-dependent reduction of length and number of filopodia in response to proBDNF which was accompanied with a polarized RhoA activation in growth cones. Moreover, proBDNF treatment of cortical neurons resulted in a time-dependent activation of RhoA but not Cdc42 and the effect was absent in p75NTR(-/-) neurons. Rho kinase (ROCK) and the collapsin response mediator protein-2 (CRMP-2) were also involved in the proBDNF action. CONCLUSIONS: proBDNF has an opposing role in neurite outgrowth to that of mature BDNF. Our observations suggest that proBDNF collapses neurites outgrowth and filopodial growth cones by activating RhoA through the p75NTR signaling pathway.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Ganglios Espinales/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Precursores de Proteínas/fisiología , Proteínas de Unión al GTP rho/agonistas , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Anticuerpos/farmacología , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Ratones , Ratones Noqueados , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/fisiología , Neuritas/efectos de los fármacos , Neuritas/fisiología , Precursores de Proteínas/farmacología , Seudópodos/efectos de los fármacos , Seudópodos/fisiología , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal/fisiología , Imagen de Lapso de Tiempo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
10.
Methods Mol Biol ; 827: 271-82, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22144281

RESUMEN

Extensive knowledge about protein-protein interactions is fundamental to fully understand signaling pathways and for the development of new drugs. Rho GTPases are key molecules in cellular signaling processes and their deregulation is implicated in the development of a variety of diseases such as neurofibromatosis type 2 and cancer. Here, we describe a bead-based protein-protein interaction assay for overexpressed HA-tagged Rho GTPases to study the GTPγS-dependent interaction with the regulatory protein RhoGDIα. This assay provides a useful tool for the analysis of both macromolecular and small molecule activators and inhibitors of the protein-protein interactions of Rho GTPases with their regulatory proteins in a multiplexed miniaturized format.


Asunto(s)
Análisis por Matrices de Proteínas/métodos , Mapeo de Interacción de Proteínas/métodos , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/metabolismo , Proteínas Bacterianas/metabolismo , Unión Competitiva , Cisteína Endopeptidasas/metabolismo , Activación Enzimática/efectos de los fármacos , Microesferas , Nucleótidos/química , Nucleótidos/metabolismo , Unión Proteica , Proteolisis
11.
J Pharmacol Exp Ther ; 339(2): 403-11, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21828260

RESUMEN

Anacardic acid (6-pentadecylsalicylic acid), a natural inhibitor of histone acetyltransferase from Amphipterygium adstringens, has been shown to have anti-inflammatory, anticancer, antioxidative, and antimicrobial functions. However, whether this salicylic acid could block angiogenesis has not been elucidated to date. Here, we postulate that anacardic acid affects multiple steps of tumor angiogenesis to contribute to tumor inhibition. In this study, we found that vascular endothelial growth factor (VEGF)-induced cell proliferation, migration, and adhesion and capillary-like structure formation of primary cultured human umbilical vascular endothelial cells (HUVECs) could all be significantly suppressed by anacardic acid in vitro, without detectable cellular toxicity. Furthermore, anacardic acid effectively inhibited vascular development in chick embryo chorioallantoic membrane ex vivo (n = 10) and VEGF-triggered corneal neovascularization in vivo (n = 10). A mechanistic study revealed that anacardic acid blocked activities of Src and FAK kinases in concentration- and time-dependent manners in HUVECs, resulting in activation of RhoA-GTPase and inactivation of Rac1- and Cdc42-GTPases. Of note, when anacardic acid (2 mg/kg per day) was subcutaneously administrated to mice bearing human prostate tumor xenografts (n = 6-7), the volume and weight of solid tumors were significantly retarded. Src, Ki-67, and CD31 immunohistochemical staining further revealed that Src protein expression, tumor cell proliferation, and microvessel density could be remarkably suppressed by anacardic acid. Taken together, our findings demonstrate for the first time that anacardic acid functions as a potent tumor angiogenesis inhibitor by targeting the Src/FAK/Rho GTPase signaling pathway, leading to significant suppression of prostate tumor growth.


Asunto(s)
Ácidos Anacárdicos/farmacología , Inhibidores de la Angiogénesis/farmacología , GTP Fosfohidrolasas/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Ácidos Anacárdicos/uso terapéutico , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/patología , Neovascularización de la Córnea/tratamiento farmacológico , Neovascularización de la Córnea/metabolismo , Relación Dosis-Respuesta a Droga , Quinasa 1 de Adhesión Focal/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Neovascularización Patológica/metabolismo , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión al GTP rho/agonistas , Quinasas Asociadas a rho/metabolismo , Familia-src Quinasas/metabolismo
12.
Clin Cancer Res ; 15(12): 4242-9, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19509159

RESUMEN

PURPOSE: Tocopherols are lipophilic antioxidants present in vegetable oils. Although the antioxidant and anticancer activities of alpha-tocopherol (vitamin E) have been studied for decades, recent intervention studies with alpha-tocopherol have been negative for protection from cancer in humans. The tocopherols consist of four isoforms, which are the alpha, beta, gamma, and delta variants, and recent attention is being given to other isoforms. In the present study, we investigated the inhibitory effect of a tocopherol mixture rich in gamma- and delta-tocopherols against mammary tumorigenesis. EXPERIMENTAL DESIGN: Female Sprague Dawley rats were treated with N-methyl-N-nitrosourea (NMU), and then fed diets containing 0.1%, 0.3%, or 0.5% mixed tocopherols rich in gamma- and delta-tocopherols for 9 weeks. Tumor burden and multiplicity were determined, and the levels of markers of inflammation, proliferation, and apoptosis were evaluated in the serum and in mammary tumors. The regulation of nuclear receptor signaling by tocopherols was studied in mammary tumors and in breast cancer cells. RESULTS: Dietary administration of 0.1%, 0.3%, or 0.5% mixed tocopherols suppressed mammary tumor growth by 38%, 50%, or 80%, respectively. Tumor multiplicity was also significantly reduced in all three mixed tocopherol groups. Mixed tocopherols increased the expression of p21, p27, caspase-3, and peroxisome proliferator activated receptor-gamma, and inhibited AKT and estrogen signaling in mammary tumors. Our mechanistic study found that gamma- and delta-tocopherols, but not alpha-tocopherol, activated peroxisome proliferator activated receptor-gamma and antagonized estrogen action in breast cancer. CONCLUSION: The results suggest that gamma- and delta-tocopherols may be effective agents for the prevention of breast cancer.


Asunto(s)
Anticarcinógenos/administración & dosificación , Neoplasias Mamarias Experimentales/prevención & control , Tocoferoles/administración & dosificación , Vitaminas/administración & dosificación , Alquilantes/farmacología , Animales , Anticarcinógenos/química , Apoptosis , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Suplementos Dietéticos , Femenino , Humanos , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/patología , Metilnitrosourea/toxicidad , PPAR gamma/agonistas , PPAR gamma/metabolismo , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Tocoferoles/química , Vitaminas/química , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/metabolismo
13.
J Immunol ; 176(9): 5314-20, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16621998

RESUMEN

RhoG is a Rho family small GTPase implicated in cytoskeletal regulation, acting either upstream of or in parallel to Rac1. The precise function(s) of RhoG in vivo has not yet been defined. We have identified a novel role for RhoG in signaling the neutrophil respiratory burst stimulated by G protein-coupled receptor agonists. Bone marrow-derived neutrophils from RhoG knockout (RhoG(-/-)) mice exhibited a marked impairment of oxidant generation in response to C5a or fMLP, but normal responses to PMA or opsonized zymosan and normal bacterial killing. Activation of Rac1 and Rac2 by fMLP was diminished in RhoG(-/-) neutrophils only at very early (5 s) time points (by 25 and 32%, respectively), whereas chemotaxis in response to soluble agonists was unaffected by lack of RhoG. Additionally, fMLP-stimulated phosphorylation of protein kinase B and p38MAPK, activation of phospholipase D, and calcium fluxes were equivalent in wild-type and RhoG(-/-) neutrophils. Our results define RhoG as a critical component of G protein-coupled receptor-stimulated signaling cascades in murine neutrophils, acting either via a subset of total cellular Rac relevant to oxidase activation and/or by a novel and as yet undefined interaction with the neutrophil NADPH oxidase.


Asunto(s)
NADPH Oxidasas/metabolismo , Neutrófilos/enzimología , Proteínas de Unión al GTP rho/metabolismo , Animales , Calcio/metabolismo , Catecolaminas/farmacología , Células Cultivadas , Quimiotaxis de Leucocito , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Humanos , Imidazolinas/farmacología , Ratones , Ratones Noqueados , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Fosfolipasa D/metabolismo , Transducción de Señal , Solubilidad , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/deficiencia , Proteínas de Unión al GTP rho/genética
14.
Mol Pharmacol ; 69(6): 2068-75, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16554409

RESUMEN

Because phospholipase C epsilon (PLC-epsilon) is activated by Galpha(12/13) and Rho family GTPases, we investigated whether these G proteins contribute to the increased inositol lipid hydrolysis observed in COS-7 cells after activation of certain G protein-coupled receptors. Stimulation of inositol lipid hydrolysis by endogenous lysophosphatidic acid (LPA) or thrombin receptors was markedly enhanced by the expression of PLC-epsilon. Expression of the LPA(1) or PAR1 receptor increased inositol phosphate production in response to LPA or SFLLRN, respectively, and these agonist-stimulated responses were markedly enhanced by coexpression of PLC-epsilon. Both LPA(1) and PAR1 receptor-mediated activation of PLC-epsilon was inhibited by coexpression of the regulator of G protein signaling (RGS) domain of p115RhoGEF, a GTPase-activating protein for Galpha(12/13) but not by expression of the RGS domain of GRK2, which inhibits Galpha(q) signaling. In contrast, activation of the G(q)-coupled M1 muscarinic or P2Y(2) purinergic receptor was neither enhanced by coexpression with PLC-epsilon nor inhibited by the RGS domain of p115RhoGEF but was blocked by expression of the RGS domain of GRK2. Expression of the Rho inhibitor C3 botulinum toxin did not affect LPA- or SFLLRN-stimulated inositol lipid hydrolysis in the absence of PLC-epsilon but completely prevented the PLC-epsilon-dependent increase in inositol phosphate accumulation. Likewise, C3 toxin blocked the PLC-epsilon-dependent stimulatory effects of the LPA(1), LPA(2), LPA(3), or PAR1 receptor but had no effect on the agonist-promoted inositol phosphate response of the M1 or P2Y(2) receptor. Moreover, PLC-epsilon-dependent stimulation of inositol phosphate accumulation by activation of the epidermal growth factor receptor, which involves Ras- but not Rho-mediated activation of the phospholipase, was unaffected by C3 toxin. These studies illustrate that specific LPA and thrombin receptors promote inositol lipid signaling via activation of Galpha(12/13) and Rho.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Receptor PAR-1/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Fosfolipasas de Tipo C/metabolismo , Proteínas de Unión al GTP rho/metabolismo , ADP Ribosa Transferasas/farmacología , Animales , Toxinas Botulínicas/farmacología , Células COS , Chlorocebus aethiops , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Hidrólisis , Fosfatos de Inositol/metabolismo , Metabolismo de los Lípidos , Fragmentos de Péptidos/farmacología , Fosfoinositido Fosfolipasa C , Receptores de Trombina/metabolismo , Fosfolipasas de Tipo C/antagonistas & inhibidores , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/antagonistas & inhibidores
15.
Biochem Biophys Res Commun ; 323(2): 395-401, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15369765

RESUMEN

Rho GTPases acting through effector proteins regulate actin dynamics and cytoskeletal structure. In Hermissenda Csp24 is a cytoskeletal-related protein that contributes to the development of intermediate-term memory, and is homologous to other beta-thymosin-like repeat proteins containing multiple actin-binding domains. We have examined the role of Rho GTPase activity and its downstream target ROCK, and cyclin-dependent kinase 5 (Cdk5) on the phosphorylation of Csp24 using 32PO4 labeling of proteins separated with 2-D PAGE. The ROCK inhibitor Y-27632 significantly increased Csp24 phosphorylation, and the Rho activator lysophosphatidic acid (LPA) or the Cdk5 inhibitor butyrolactone significantly decreased Csp24 phosphorylation. Pretreatment with Y-27632 before LPA application significantly reduced the decreased phosphorylation of Csp24 normally detected in nervous systems exposed to LPA. Using a pull-down assay we found that LPA treatments activated Rho and exposure to 5-HT decreased Rho activity. Our results indicate that the Rho/ROCK and Cdk5 signaling pathways contribute to the regulation of Csp24 phosphorylation.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Microfilamentos/metabolismo , Moluscos/enzimología , Fosfoproteínas/metabolismo , Timosina/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Amidas/farmacología , Animales , Butirofenonas/farmacología , Quinasa 5 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular , Lisofosfolípidos/farmacología , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/enzimología , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/farmacología , Timosina/análogos & derivados , Proteínas de Unión al GTP rho/agonistas , Quinasas Asociadas a rho
16.
Pigment Cell Res ; 16(2): 139-48, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12622791

RESUMEN

A hallmark of melanocytic cells is their ability to form dendrites in response to growth factors and to ultraviolet irradiation. It is known that the cyclic adenosine monophosphate (cAMP) second messenger pathway stimulates melanocyte dendrite formation because agents that increase cAMP such as forskolin and dibutyrl cAMP induce dendrite formation in normal human and murine melanocytes and melanoma cell lines. The Rho family of guanosine triphosphate (GTP)-binding proteins regulates cytoskeletal reorganization in all cells tested and Rac and Rho have both been shown to regulate melanocyte dendrite formation. In this report, we analyzed the effect of cAMP on the activation of Rac and Rho and show that elevation of cAMP stimulates Rac and inhibits Rho in B16F10 cells. The Rho GTP-binding proteins have also been shown to either cross-activate or inhibit each other and in this report we show that Rac activates Rho in B16F10 cells. Microinjection of C3 botulinum exoenzyme toxin, an agent that specifically inactivates Rho or microinjection of constitutively active mutant Rac protein-induced dendricity in human melanocytes and in B16F10 and B16F1 murine melanoma cell lines. We conclude that cAMP-mediated dendrite formation in melanocytic cells is mediated through upregulation of Rac activity and downregulation of Rho activity.


Asunto(s)
Diferenciación Celular/fisiología , AMP Cíclico/metabolismo , Dendritas/metabolismo , Melanocitos/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Toxinas Botulínicas/farmacología , Diferenciación Celular/efectos de los fármacos , AMP Cíclico/farmacología , Dendritas/efectos de los fármacos , Dendritas/ultraestructura , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Melanocitos/citología , Melanocitos/efectos de los fármacos , Melanoma/genética , Melanoma/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteínas de Unión al GTP rac/efectos de los fármacos , Proteínas de Unión al GTP rho/agonistas , Proteínas de Unión al GTP rho/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...