Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Acta Pharmacol Sin ; 44(10): 1977-1988, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37217602

RESUMEN

Atherosclerotic diseases remain the leading cause of adult mortality and impose heavy burdens on health systems globally. Our previous study found that disturbed flow enhanced YAP activity to provoke endothelial activation and atherosclerosis, and targeting YAP alleviated endothelial inflammation and atherogenesis. Therefore, we established a luciferase reporter assay-based drug screening platform to seek out new YAP inhibitors for anti-atherosclerotic treatment. By screening the FDA-approved drug library, we identified that an anti-psychotic drug thioridazine markedly suppressed YAP activity in human endothelial cells. Thioridazine inhibited disturbed flow-induced endothelial inflammatory response in vivo and in vitro. We verified that the anti-inflammatory effects of thioridazine were mediated by inhibition of YAP. Thioridazine regulated YAP activity via restraining RhoA. Moreover, administration of thioridazine attenuated partial carotid ligation- and western diet-induced atherosclerosis in two mouse models. Overall, this work opens up the possibility of repurposing thioridazine for intervention of atherosclerotic diseases. This study also shed light on the underlying mechanisms that thioridazine inhibited endothelial activation and atherogenesis via repression of RhoA-YAP axis. As a new YAP inhibitor, thioridazine might need further investigation and development for the treatment of atherosclerotic diseases in clinical practice.


Asunto(s)
Aterosclerosis , Células Endoteliales , Tioridazina , Animales , Humanos , Ratones , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/prevención & control , Inflamación/etiología , Proteína de Unión al GTP rhoA/efectos de los fármacos , Tioridazina/uso terapéutico , Proteínas Señalizadoras YAP/efectos de los fármacos
2.
Acta Diabetol ; 60(6): 805-815, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36920548

RESUMEN

AIMS: Diabetic peripheral neuropathy (DPN) is one of the most important complications of diabetes with a poor prognosis. Saikosaponin d (SSD) is a triterpenoid saponin isolated from Radix Bupleuri that has multiple pharmacological activities. However, whether SSD affects DPN is unclarified. METHODS: Sprague Dawley rats were treated with streptozotocin (STZ) and high-fat diet (HFD) to induce DPN, in the presence or absence of SSD, with or without transfection of lentivirus vectors carrying siRNA targeting aquaporin 1 (si-AQP1). The body weight, plasma glucose levels, mechanical and thermal hyperalgesia, and nerve conductive velocity (NCV) of rats were measured. Hematoxylin-Eosin staining was used for histopathological observation of sciatic nerves. RT-qPCR and western blotting were utilized for measuring expression levels of AQP1 and ras homolog family member A/Rho-associated protein kinase (RhoA/ROCK) signaling pathway-related markers in dorsal root ganglion (DRG) of rats. RESULTS: SSD increased the body weight, decreased plasma glucose levels, attenuated mechanical and thermal hyperalgesia, enhanced NCV and reduced proinflammatory cytokine levels in DPN rats. AQP1 displayed a high level in DPN rats and SSD treatment repressed the expression of AQP1. SSD enhanced the protective effect of AQP1 knockdown on the pathological changes of DPN. AQP1 depletion suppressed the activation of RhoA/ROCK signaling pathway in DPN rats. CONCLUSION: SSD alleviates STZ/HFD-induced DPN in rats by inhibiting the AQP1/RhoA/ROCK signaling pathway.


Asunto(s)
Diabetes Mellitus Experimental , Neuropatías Diabéticas , Saponinas , Animales , Ratas , Acuaporina 1/efectos de los fármacos , Acuaporina 1/genética , Acuaporina 1/metabolismo , Glucemia , Peso Corporal , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/etiología , Hiperalgesia/complicaciones , Hiperalgesia/metabolismo , Ratas Sprague-Dawley , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo , Saponinas/farmacología , Saponinas/uso terapéutico , Transducción de Señal , Estreptozocina/efectos adversos , Estreptozocina/farmacología , Quinasas Asociadas a rho/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
3.
Drug Deliv ; 28(1): 2051-2061, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34595978

RESUMEN

Fibrotic diseases pose significant clinical challenges due to their broadness and complexity. Thus, a better understanding of fibrogenesis and the development of more effective treatments is imperative. Recent evidence suggests a significant antifibrotic potential of an endogenous glycoprotein, endostatin. While endostatin has been widely studied for its role as an anticancer adjuvant by inhibiting tumor angiogenesis, its possible implication in fibrosis remains largely unclear. Here, we review the role of endostatin in various cellular processes and highlight its antifibrotic activity. We hypothesize that endostatin conveys a homeostatic function in the process of fibrosis by regulating (a) TGF-ß1 and its downstream signaling; (b) RhoA/ROCK pathway; (c) NF-κB signaling pathway; (d) expression of EGR-1; (e) PDGF/PDGFR pathway; (f) autophagy-related pathways; (g) pathways associated with cell proliferation and apoptosis. Finally, we propose a schematic model of the antifibrotic roles and mechanisms of endostatin; also, we outline future research directions of endostatin and aim to present a potential therapeutic approach for fibrosis.


Asunto(s)
Endostatinas/farmacología , Fibrosis/tratamiento farmacológico , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos
4.
Exp Neurol ; 337: 113554, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33309746

RESUMEN

This study aims to investigate whether escin ameliorates the impairments of neurological function by ameliorating systemic inflammation instead of targeting the brain directly in intracerebral hemorrhage (ICH) mice. It showed that escin did not cross the blood brain barrier (BBB). Compared with the ICH group, the Garcia test scores in the escin groups were significantly increased. Brain water contents and Evans blue extravasation of the right basal ganglia in the ICH group were augmented, and significantly reduced by escin. Escin abated the increases of monocyte counts and serum IL-1ß levels induced by ICH. IL-1ß administration reversed the effect of escin on Garcia test scores, the brain water contents, and the Evans blue extravasation. Escin ameliorated the increasing levels of RhoA, ROCK1, nuclear NF-κB and the decreasing expression of IκBα, cytosolic NF-κB, occludin, claudin-5 in the ICH group. IL-1ß administration blocked not only escin-mediated increases of IκBα, cytosolic NF-κB, occludin, and claudin-5, but also escin-caused decreases of RhoA, ROCK1, and nuclear NF-κB. The results indicate that escin improves neurological outcomes and the BBB function in ICH mice, which is associated with attenuating ICH-induced peripheral system inflammation, and therefore, inhibiting IL-1ß/RhoA/NF-κB signaling pathway in BBB, at least in part. These findings suggest that it may be useful to ameliorate brain injury by inhibiting systemic inflammation instead of aiming to target the brain directly after ICH.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Fármacos Cardiovasculares/uso terapéutico , Hemorragia Cerebral/tratamiento farmacológico , Escina/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Animales , Barrera Hematoencefálica/fisiopatología , Agua Corporal/metabolismo , Hemorragia Cerebral/fisiopatología , Interleucina-1beta/sangre , Interleucina-1beta/farmacología , Masculino , Ratones , Monocitos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos
5.
BMC Nephrol ; 21(1): 402, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32948146

RESUMEN

BACKGROUND: Dipeptidyl peptidase-4 (DPP-4) is a serine protease that inhibits the degradation of glucagon-like peptide 1. DPP-4 inhibitors are used worldwide to treat type 2 diabetes mellitus and were recently shown to have pleiotropic effects such as anti-oxidant, anti-inflammatory, and anti-fibrotic actions. DPP-4 inhibitors improve albuminuria and renal injury including glomerular damage independent of its hypoglycemic effect. Although DPP-4 is mainly expressed in the kidney, the physiological function of DPP-4 remains unclear. METHODS: The localization of renal DPP-4 activity was determined in human renal biopsy specimens with glycyl-1-prolyl-4-methoxy-2-naphthylamide and the effects of a DPP-4 inhibitor were examined in human cultured podocyte. RESULTS: DPP-4 activity under normal conditions was observed in some Bowman's capsular epithelial cells and proximal tubules, but not in the glomerulus. DPP-4 activity was observed in crescent formation in anti-neutrophil myeloperoxidase cytoplasmic antigen antibody nephritis, nodular lesions in diabetic nephropathy, and some podocytes in focal segmental glomerulosclerosis. Notably, the DPP-4 inhibitor saxagliptin suppressed DPP-4 activity in podocytes and the proximal tubules. To assess the effect of DPP-4 inhibitor on podocytes, human cultured podocytes were injured by Adriamycin, which increased DPP-4 activity; this activity was dose-dependently suppressed by saxagliptin. Treatment with saxagliptin maintained the structure of synaptopodin and RhoA. Saxagliptin also improved the detachment of podocytes. CONCLUSIONS: DPP-4 activity induces degradation of synaptopodin and reduction of RhoA, resulting in destruction of the podocyte cytoskeleton. Saxagliptin may have pleiotropic effects to prevent podocyte injury.


Asunto(s)
Adamantano/análogos & derivados , Nefropatías Diabéticas/metabolismo , Dipéptidos/farmacología , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Riñón/metabolismo , Nefritis/metabolismo , Podocitos/efectos de los fármacos , Adamantano/farmacología , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Cápsula Glomerular/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Doxorrubicina/farmacología , Femenino , Humanos , Técnicas In Vitro , Glomérulos Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Masculino , Proteínas de Microfilamentos/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Nefritis/inmunología , Podocitos/metabolismo , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
6.
Acta Neurobiol Exp (Wars) ; 80(3): 225-244, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32990282

RESUMEN

Alzheimer's disease (AD) has become the most prevalent neurodegenerative disorder. Given the pathogenesis of AD is unclear, there is currently no drug approved to halt or delay the progression of AD. Therefore, it is pressing to explore new targets and drugs for AD. In China, polyphenolic Chinese herbal medicine has been used for thousands of years in clinical application, and no toxic effects have been reported. In the present study, using D­galactose and aluminum­induced rat model, the effects of paeonol on AD were validated via the Morris water maze test, open field test, and elevated plus maze test. Neuronal morphology in frontal cortex was assessed using ImageJ's Sholl plugin and RESCONSTRUCT software. RhoA/Rock2/Limk1/cofilin1 signaling pathway­related molecules were determined by Western blotting. Cofilin1 and p­cofilin1 were analyzed by immunofluorescence. Results showed that pre­treatment with paeonol attenuated D­galactose and aluminum­induced behavioral dysfunction and AD­like pathological alterations in the frontal cortex. Accompanied by these changes were the alterations in the dendrite and dendritic spine densities, especially the mushroom­type and filopodia­type spines in the apical dendrites, as well as actin filaments. In addition, the activity and intracellular distribution of cofilin1 and the molecules RhoA/Rock2/Limk1 that regulate the signaling pathway for cofilin1 phosphorylation have also changed. Our data suggests that paeonol may be through reducing Aß levels to alleviate the loss of fibrillar actin and dendrites and dendritic spines via the Rho/Rock2/Limk1/cofilin1 signaling pathway in the frontal cortex, and ultimately improving AD­like behavior.


Asunto(s)
Aluminio/farmacología , Enfermedad de Alzheimer/metabolismo , Espinas Dendríticas/metabolismo , Galactosa/farmacología , Proteína de Unión al GTP rhoA/metabolismo , Enfermedad de Alzheimer/patología , Animales , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/patología , Hipocampo/efectos de los fármacos , Quinasas Lim/efectos de los fármacos , Quinasas Lim/metabolismo , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos
7.
Am J Obstet Gynecol ; 223(5): 733.e1-733.e14, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32417359

RESUMEN

BACKGROUND: Uterine leiomyomas, the most common tumors of the female reproductive system, are characterized by excessive deposition of disordered stiff extracellular matrix and fundamental alteration in the mechanical signaling pathways. Specifically, these alterations affect the normal dynamic state of responsiveness to mechanical cues in the extracellular environment. These mechanical cues are converted through integrins, cell membrane receptors, to biochemical signals including cytoskeletal signaling pathways to maintain mechanical homeostasis. Leiomyoma cells overexpress ß1 integrin and other downstream mechanical signaling proteins. We previously reported that simvastatin, an antihyperlipidemic drug, has antileiomyoma effects through cellular, animal model, and epidemiologic studies. OBJECTIVE: This study aimed to examine the hypothesis that simvastatin might influence altered mechanotransduction in leiomyoma cells. STUDY DESIGN: This is a laboratory-based experimental study. Primary leiomyoma cells were isolated from 5 patients who underwent hysterectomy at the Department of Gynecology and Obstetrics of the Johns Hopkins University Hospital. Primary and immortalized human uterine leiomyoma cells were treated with simvastatin at increasing concentrations (0.001, 0.01, 0.1, and 1 µM, or control) for 48 hours. Protein and mRNA levels of ß1 integrin and extracellular matrix components involved in mechanical signaling were quantified by quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. In addition, we examined the effect of simvastatin on the activity of Ras homolog family member A using pull-down assay and gel contraction. RESULTS: We found that simvastatin significantly reduced the protein expression of ß1 integrin by 44% and type I collagen by 60% compared with untreated leiomyoma cells. Simvastatin-treated cells reduced phosphorylation of focal adhesion kinase down to 26%-60% of control, whereas it increased total focal adhesion kinase protein expression. Using a Ras homolog family member A pull-down activation assay, we observed reduced levels of active Ras homolog family member A in simvastatin-treated cells by 45%-85% compared with control. Consistent with impaired Ras homolog family member A activation, simvastatin treatment reduced tumor gel contraction where gel area was 122%-153% larger than control. Furthermore, simvastatin treatment led to reduced levels of mechanical signaling proteins involved in ß1 integrin downstream signaling, such as A-kinase anchor protein 13, Rho-associated protein kinase 1, myosin light-chain kinase, and cyclin D1. CONCLUSION: The results of this study suggest a possible therapeutic role of simvastatin in restoring the altered state of mechanotransduction signaling in leiomyoma. Collectively, these findings are aligned with previous epidemiologic studies and other reports and support the need for clinical trials.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Leiomioma/genética , Mecanotransducción Celular/efectos de los fármacos , Simvastatina/farmacología , Neoplasias Uterinas/genética , Proteínas de Anclaje a la Quinasa A/efectos de los fármacos , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Colágeno Tipo I/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Integrina beta1/efectos de los fármacos , Integrina beta1/genética , Integrina beta1/metabolismo , Leiomioma/metabolismo , Mecanotransducción Celular/genética , Antígenos de Histocompatibilidad Menor/efectos de los fármacos , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Quinasa de Cadena Ligera de Miosina/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosforilación , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Neoplasias Uterinas/metabolismo , Quinasas Asociadas a rho/efectos de los fármacos , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
8.
BMC Anesthesiol ; 20(1): 43, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32085741

RESUMEN

BACKGROUND: Substantial clinical and preclinical evidence have indicated the association between amide-linked local anesthesia and the long-term outcomes of cancer patients. However, the potential effects of local anesthesia on cancer recurrence are inconclusive and the underlying mechanisms remain poorly understood. METHODS: We systematically examined the effects of three commonly used local anesthetics in melanoma cells and analyzed the underlying mechanisms focusing on small GTPases. RESULTS: Ropivacaine and lidocaine but not bupivacaine inhibited migration and proliferation, and induced apoptosis in melanoma cells. In addition, ropivacaine and lidocaine but not bupivacaine significantly augmented the in vitro efficacy of vemurafenib (a B-Raf inhibitor for melanoma with BRAF V600E mutation) and dacarbazine (a chemotherapeutic drug). Mechanistically, ropivacaine but not bupivacaine decreased the activities of Ras superfamily members with the dominant inhibitory effects on RhoA and Ras, independent of sodium channel blockade. Rescue studies using constitutively active Ras and Rho activator calpeptin demonstrated that ropivacaine inhibited migration mainly through RhoA whereas growth and survival were mainly inhibited through Ras in melanoma cells. We further detected a global reduction of downstream signaling of Ras and RhoA in ropivacaine-treated melanoma cells. CONCLUSION: Our study is the first to demonstrate the anti-melanoma activity of ropivacaine and lidocaine but not bupivacaine, via targeting small GTPases. Our findings provide preclinical evidence on how amide-linked local anesthetics could affect melanoma patients.


Asunto(s)
Anestésicos Locales/farmacología , Melanoma/metabolismo , Bloqueadores de los Canales de Sodio/metabolismo , Proteínas ras/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos , Bupivacaína/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Técnicas In Vitro , Lidocaína/farmacología , Melanoma/tratamiento farmacológico , Ropivacaína/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas ras/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
9.
Front Endocrinol (Lausanne) ; 11: 607968, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33597925

RESUMEN

Transforming growth factor-ß (TGF-ß)-induced differentiation of orbital fibroblasts into myofibroblasts is an important pathogenesis of Graves' ophthalmopathy (GO) and leads to orbital tissue fibrosis. In the present study, we explored the antifibrotic effects of simvastatin and ROCK inhibitor Y-27632 in primary cultured GO orbital fibroblasts and tried to explain the molecular mechanisms behind these effects. Both simvastatin and Y-27632 inhibited TGF-ß-induced α-smooth muscle actin (α-SMA) expression, which serves as a marker of fibrosis. The inhibitory effect of simvastatin on TGF-ß-induced RhoA, ROCK1, and α-SMA expression could be reversed by geranylgeranyl pyrophosphate, an intermediate in the biosynthesis of cholesterol. This suggested that the mechanism of simvastatin-mediated antifibrotic effects may involve RhoA/ROCK signaling. Furthermore, simvastatin and Y-27632 suppressed TGF-ß-induced phosphorylation of ERK and p38. The TGF-ß-mediated α-SMA expression was suppressed by pharmacological inhibitors of p38 and ERK. These results suggested that simvastatin inhibits TGF-ß-induced myofibroblast differentiation via suppression of the RhoA/ROCK/ERK and p38 MAPK signaling pathways. Thus, our study provides evidence that simvastatin and ROCK inhibitors may be potential therapeutic drugs for the prevention and treatment of orbital fibrosis in GO.


Asunto(s)
Amidas/farmacología , Oftalmopatía de Graves/patología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Piridinas/farmacología , Simvastatina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/efectos de los fármacos , Adulto , Diferenciación Celular/efectos de los fármacos , Femenino , Fibrosis/prevención & control , Humanos , Masculino , Persona de Mediana Edad , Fosforilación , Cultivo Primario de Células , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/farmacología
10.
J Am Soc Nephrol ; 30(5): 795-810, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30988011

RESUMEN

BACKGROUND: Arginine-vasopressin (AVP) binding to vasopressin V2 receptors promotes redistribution of the water channel aquaporin-2 (AQP2) from intracellular vesicles into the plasma membrane of renal collecting duct principal cells. This pathway fine-tunes renal water reabsorption and urinary concentration, and its perturbation is associated with diabetes insipidus. Previously, we identified the antimycotic drug fluconazole as a potential modulator of AQP2 localization. METHODS: We assessed the influence of fluconazole on AQP2 localization in vitro and in vivo as well as the drug's effects on AQP2 phosphorylation and RhoA (a small GTPase, which under resting conditions, maintains F-actin to block AQP2-bearing vesicles from reaching the plasma membrane). We also tested fluconazole's effects on water flow across epithelia of isolated mouse collecting ducts and on urine output in mice treated with tolvaptan, a VR2 blocker that causes a nephrogenic diabetes insipidus-like excessive loss of hypotonic urine. RESULTS: Fluconazole increased plasma membrane localization of AQP2 in principal cells independent of AVP. It also led to an increased AQP2 abundance associated with alterations in phosphorylation status and ubiquitination as well as inhibition of RhoA. In isolated mouse collecting ducts, fluconazole increased transepithelial water reabsorption. In mice, fluconazole increased collecting duct AQP2 plasma membrane localization and reduced urinary output. Fluconazole also reduced urinary output in tolvaptan-treated mice. CONCLUSIONS: Fluconazole promotes collecting duct AQP2 plasma membrane localization in the absence of AVP. Therefore, it might have utility in treating forms of diabetes insipidus (e.g., X-linked nephrogenic diabetes insipidus) in which the kidney responds inappropriately to AVP.


Asunto(s)
Acuaporina 2/metabolismo , Transporte Biológico/genética , Colforsina/farmacología , Diabetes Insípida Nefrogénica/tratamiento farmacológico , Fluconazol/farmacología , Proteína de Unión al GTP rhoA/efectos de los fármacos , Análisis de Varianza , Animales , Membrana Celular/metabolismo , Células Cultivadas , Diabetes Insípida Nefrogénica/metabolismo , Modelos Animales de Enfermedad , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fosforilación/genética , Distribución Aleatoria , Transducción de Señal , Estadísticas no Paramétricas
11.
Neurosci Lett ; 696: 191-196, 2019 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-30597230

RESUMEN

Transforming growth factor (TGF)-ß stimulates release of interleukin (IL)-6, which is recognized to function as both a pro- and anti- inflammatory cytokine in the central nervous system, from astrocytes. It is generally recognized that effects of TGF-ß are mediated through Smad-independent as well as Smad-dependent pathways. Small GTPases regulate a variety of cell functions. In the present study, we investigated whether or not Rho-kinase, a downstream effector of Rho, and Rac are implicated in TGF-ß-stimulated IL-6 release from astrocytes (C8D1A cells). Y-27632 or fasudil (Rho-kinase inhibitors) or NSC23766 (an inhibitor of Rac-guanine nucleotide exchange factor interaction) significantly enhanced TGF-ß-stimulated IL-6 release from these cells. TGF-ß-stimulated IL-6 release was markedly upregulated in RhoA- or Rac-knockdown C8D1A cells. We found that SIS3 (a specific inhibitor of TGF-ß-dependent Smad3 phosphorylation) or LY364947 (a TGF-ß type I receptor kinase inhibitor) significantly reduced the IL-6 release. However, TGF-ß-induced-Smad2 and Smad3 phosphorylation was not affected by Y-27632, fasudil or NSC23766. In conclusion, our results strongly suggest that Rho-kinase and Rac limit TGF-ß-induced IL-6 release from astrocytes, and the suppressive effects are exerted independently of the Smad pathway or at a point downstream of Smad2/3 complex.


Asunto(s)
Amidas/farmacología , Aminoquinolinas/farmacología , Astrocitos/efectos de los fármacos , Interleucina-6/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Quinasas Asociadas a rho/efectos de los fármacos , Animales , Astrocitos/metabolismo , Ratones , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
12.
JCI Insight ; 3(21)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30385725

RESUMEN

Patients with severe, treatment-refractory asthma are at risk for death from acute exacerbations. The cytokine IL17A has been associated with airway inflammation in severe asthma, and novel therapeutic targets within this pathway are urgently needed. We recently showed that IL17A increases airway contractility by activating the procontractile GTPase RhoA. Here, we explore the therapeutic potential of targeting the RhoA pathway activated by IL17A by inhibiting RhoA guanine nucleotide exchange factors (RhoGEFs), intracellular activators of RhoA. We first used a ribosomal pulldown approach to profile mouse airway smooth muscle by qPCR and identified Arhgef12 as highly expressed among a panel of RhoGEFs. ARHGEF12 was also the most highly expressed RhoGEF in patients with asthma, as found by RNA sequencing. Tracheal rings from Arhgef12-KO mice and WT rings treated with a RhoGEF inhibitor had evidence of decreased contractility and RhoA activation in response to IL17A treatment. In a house dust mite model of allergic sensitization, Arhgef12-KO mice had decreased airway hyperresponsiveness without effects on airway inflammation. Taken together, our results show that Arhgef12 is necessary for IL17A-induced airway contractility and identify a therapeutic target for severe asthma.


Asunto(s)
Asma/metabolismo , Interleucina-17/metabolismo , Contracción Muscular/efectos de los fármacos , Hipersensibilidad Respiratoria/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína de Unión al GTP rhoA/efectos de los fármacos , Anciano , Animales , Asma/tratamiento farmacológico , Asma/fisiopatología , Progresión de la Enfermedad , Femenino , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/farmacología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Contracción Muscular/fisiología , Hipersensibilidad Respiratoria/tratamiento farmacológico , Análisis de Secuencia de ARN/métodos , Índice de Severidad de la Enfermedad , Proteína de Unión al GTP rhoA/metabolismo
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(8): 997-1001, 2018 Jul 30.
Artículo en Chino | MEDLINE | ID: mdl-30187871

RESUMEN

OBJECTIVE: To observe effects of Biejiajian Pills on hepatocarcinoma (HCC) cell vasculogenic mimicry (VM) and explore the molecular mechanism by which Biejiajian Pills inhibits HCC metastasis and invasion. METHODS: Forty male SD rats were randomly divided into 4 groups for gastric lavage of normal saline or high, moderate or low doses of Biejiajian Pills (twice daily) for 4 consecutive days. The sera were collected from the rats for treatment of cultured human HCC HepG2 cells. VM formation in the cells was detected using an image acquisition and analysis system 24 h after incubation of the cells with the sera and with the RhoA/ROCK inhibitor Y-27632(P). The expression levels of RhoA and ROCK1 in the cells were detected using Western blotting, and the contents of VE-cadherin and PI3K in the culture supernatant were determined using ELISA. RESULTS: Treatment with the sera from Biejiajian Pills-treated rats significantly inhibited formation of VM in HepG2 cells, and the diameters of VM formed were significantly greater than those in the positive control group (P < 0.01). Y-27632 completely inhibited the formation of VM in HepG2 cells (P < 0.01). Treatments with Biejiajian Pills and Y-27632 both inhibited the expression of RhoA and ROCK1 (P < 0.05) and significantly lowered the contents of VE-cadherin and PI3K in the culture supernatant (P < 0.05). CONCLUSIONS: Biejiajian Pills can inhibit the formation of VM in HCC cells in vitro possibly by inhibiting the RhoA/ROCK pathways and the expressions of VE-cadherin and PI3K.


Asunto(s)
Carcinoma Hepatocelular/irrigación sanguínea , Medicamentos Herbarios Chinos/farmacología , Neoplasias Hepáticas/irrigación sanguínea , Neovascularización Patológica/prevención & control , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Quinasas Asociadas a rho/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos
14.
J Pharm Pharmacol ; 70(12): 1713-1722, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30251312

RESUMEN

OBJECTIVES: Total flavones of Rhododendron simsii Planch flower (TFR) are an effective part extracted from the flower. The present study was designed to investigate the protective effect of TFR in isolated rat heart following global ischaemia-reperfusion and the possible underlying mechanisms. METHODS: Langendorff perfusion apparatus was used to perfuse isolated rat heart which was subjected to global ischaemia-reperfusion. The hemodynamic parameters were continuously monitored. Coronary flow as well as lactate dehydrogenase (LDH), creatine phosphokinase-MB (CK-MB) and cardiac troponin I (cTnI) in coronary effluents was measured. RhoA activity and urotensin receptor (UTR) and Rho-related coiled-coil-forming protein kinase (ROCK) protein expressions in rat myocardium were examined, respectively. Cardiac dysfunction was indicated by the alterations of hemodynamic parameters and the reduced coronary flow. KEY FINDINGS: Total flavones of Rhododendron simsii Planch flower significantly improved ischaemia-reperfusion-induced cardiac dysfunction and leakages of LDH, CK-MB and cTnI, and inhibited myocardial ischaemia-reperfusion-increased RhoA activity and UTR, ROCK1 and ROCK2 protein expressions. The improvement of TFR in the cardiac dysfunction and the leakage of LDH, CK-MB and cTnI were markedly attenuated under the UTR blockade and ROCK inhibition. TFR-inhibited RhoA activity was decreased under the UTR blockade. CONCLUSIONS: Total flavones of Rhododendron simsii Planch flower had a protective effect on ischaemia-reperfusion injury in isolated rat heart, which may be attributed to the blocking of UTR and subsequent inhibition of the RhoA-ROCK pathway.


Asunto(s)
Flavonas/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Extractos Vegetales/farmacología , Rhododendron , Animales , Circulación Coronaria/efectos de los fármacos , Forma MB de la Creatina-Quinasa/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Flavonas/administración & dosificación , Flores , L-Lactato Deshidrogenasa/efectos de los fármacos , Masculino , Extractos Vegetales/administración & dosificación , Sustancias Protectoras , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/efectos de los fármacos , Troponina I/efectos de los fármacos , Verapamilo/farmacología , Quinasas Asociadas a rho/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos
15.
Cell Prolif ; 51(6): e12503, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30091500

RESUMEN

OBJECTIVES: The main purpose of current study was to explore the effects of tetrahedral DNA nanostructures (TDNs) on neuroectodermal (NE-4C) stem cells migration and unveil the potential mechanisms. MATERIALS AND METHODS: The successfully self-assembled TDNs were also determined by dynamic light scattering (DLS). A bidirectional wound-healing assay and transwell chamber assay were employed to test the migrating behaviour of NE-4C stem cells cultured under different conditions. RESULTS: Through an in vitro study, we found that stem cells could internalize TDNs quickly, and the cells' parallel and vertical migration was promoted effectively. Besides, the effects of TDNs were found being exerted by upregulating the gene and protein expression levels of RhoA, Rock2 and Vinculin, indicating that the RHOA/ROCK2 pathway was activated by the TDNs during the cell migration. CONCLUSIONS: In conclusion, TDNs could enter NSCs without the aid of other transfection reagents in large amounts, whereas only small amounts of ssDNA could enter the cells. TDNs taken up by NSCs activated the RHOA/ROCK2 signalling pathway, which had effects on the relevant genes and proteins expression, eventually promoting the migration of NE-4C stem cells. These findings suggested that TDNs have great potential in application for the repair and regeneration of neural tissue.


Asunto(s)
Movimiento Celular/efectos de los fármacos , ADN/farmacología , Quinasas Asociadas a rho/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos , Animales , Nanoestructuras , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transfección/métodos , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética
16.
Diabetes Obes Metab ; 18(12): 1263-1273, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27561953

RESUMEN

AIMS: The novel cannabinoid receptor GPR55 is expressed by rodent islets and it has been implicated in ß-cell function in response to a range of ligands. This study evaluated the effects of GPR55 ligands on intracellular calcium ([Ca2+ ]i ) levels and insulin secretion from islets isolated from GPR55 knockout (GPR55 -/- ) mice, age-matched wildtype (WT) mice and human pancreas. MATERIALS AND METHODS: GPR55 expression was determined by Western blotting and fluorescent immunohistochemistry. Changes in [Ca2+ ]i were measured by Fura-2 microfluorimetry. Dynamic insulin secretion was quantified by radioimmunoassay following perifusion of isolated islets. RhoA activity was monitored using a Rho binding domain pull down assay. RESULTS: Western blotting indicated that MIN6 ß-cells, mouse and human islets express GPR55 and its localization on human ß-cells was demonstrated by fluorescent immunohistochemistry. The pharmacological GPR55 agonist O-1602 (10 µM) significantly stimulated [Ca2+ ]i and insulin secretion from WT mouse islets and these stimulatory effects were abolished in islets isolated from GPR55 -/- mice. In contrast, while the putative endogenous GPR55 agonist lysophosphatidylinositol (LPI, 5 µM) and the GPR55 antagonist cannabidiol (CBD, 1 µM) also elevated [Ca2+ ]i and insulin secretion, these effects were sustained in islets from GPR55 -/- mice. Stimulatory effects of O-1602 on [Ca2+ ]i and insulin secretion were also observed in experiments using human islets, but O-1602 did not activate RhoA in MIN6 ß-cells. CONCLUSIONS: Our results therefore suggest that GPR55 plays an important role in the regulation of mouse and human islet physiology, but LPI and CBD exert stimulatory effects on islet function by a GPR55-independent pathway(s).


Asunto(s)
Cannabidiol/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Antagonistas de Receptores de Cannabinoides/farmacología , Ciclohexanos/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Lisofosfolípidos/farmacología , Receptores de Cannabinoides/genética , Receptores Acoplados a Proteínas G/metabolismo , Resorcinoles/farmacología , Animales , Western Blotting , Calcio/metabolismo , Línea Celular , Citofotometría , Electroforesis en Gel de Poliacrilamida , Humanos , Inmunohistoquímica , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Imagen Óptica , Receptores de Cannabinoides/metabolismo , Análisis de la Célula Individual , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
17.
J Craniofac Surg ; 27(2): 516-20, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26963302

RESUMEN

UNLABELLED: Angiogenesis is the development of new capillaries from existing blood vessels and is a prerequisite for the wound-healing process. Many lines of scientific evidences have shown that complicated roles of small guanosine triphosphatases (GTPases) (ras-related C3 botulinum toxin substrate 1 [Rac1], cell division control protein 42 [Cdc42], and ras homolog gene family, member A [RhoA]) in regulation of signal transduction pathways exist to transmit distinct cellular effects on the modulation of actin cytoskeleton remodeling such as cell cycle progression, cell survival, and cell motility. In addition, these small GTPases activate mitogen-activated protein kinase kinase kinases (MAP3Ks) leading to activated mitogen-activated protein kinase kinases (MAPKK), mitogen-activated protein kinase (MAPK), and various transcription factors such as vascular endothelial growth factor with involvement of MAPK signaling pathways.In this study, the authors hypothesized that botulinum toxin A increases angiogenesis via the expression of small GTPases in vivo and in vitro studies.In vivo experiment, 24 Sprague-Dawley rats were randomly divided into 2 groups: a control group and a botulinum toxin A group. Five days prior to superiorly based transverse rectus abdominis myocutaneous flap elevation, the botulinum toxin A (BoTA) group was pretreated with BoTA, while the control group was pretreated with normal saline. quantitative real-time polymerase chain reaction was performed to evaluate the expression of Rac1, RhoA, and Cdc42.The angiogenic effects of botulinum toxin A on human dermal fibroblasts were measured in vitro experiment. To understand the mechanism of botulinum toxin A on small GTPases production of fibroblasts, Rac1, Cdc42, and RhoA were measured using qRT-PCR.The relative messenger ribonucleic acid expression of Rac1, RhoA, and Cdc42 was significantly higher in the BoTA group than in the control group, in every zone and pedicle muscle, on postoperative days 1, 3, and 5. Levels of these molecules increased significantly in human dermal fibroblasts grown in the presence of BoTA compared with control group over 5 IU.Our in vivo and in vitro studies suggest that administration of BoTA upregulates the expression of RhoA, Rac1, and Cdc42 in a dose-dependent manner. MAPK signaling pathway might be involved in BoTA-induced angiogenesis mechanism. LEVEL OF EVIDENCE: N/A.


Asunto(s)
Toxinas Botulínicas Tipo A/farmacología , Proteína de Unión al GTP cdc42/efectos de los fármacos , Proteína de Unión al GTP rac1/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos , Inductores de la Angiogénesis/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Supervivencia de Injerto/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Colgajo Miocutáneo/irrigación sanguínea , Colgajo Miocutáneo/cirugía , Neovascularización Fisiológica/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Recto del Abdomen/irrigación sanguínea , Recto del Abdomen/cirugía , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
18.
Lipids Health Dis ; 14: 4, 2015 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-25971815

RESUMEN

BACKGROUND: Omega-3 polyunsaturated fatty acids (n-3 PUFAs) show beneficial effects on cardiovascular health and cognitive functions, but the underlying molecular mechanisms are not completely understood. Because of the fact that cytoskeleton dynamics affect almost every cellular process, the regulation of cytoskeletal dynamics could be a new pathway by which n-3 PUFAs exert their effects on cellular level. METHODS: A 12-week open-label intervention study with 12 healthy men was conducted to determine the effects of 2.7 g/d n-3 PUFA on changes in mRNA expression of cytoskeleton-associated genes by quantitative real-time PCR in whole blood. Furthermore, the actin content in red blood cells was analyzed by immunofluorescence imaging. RESULTS: N-3 PUFA supplementation resulted in a significant down-regulation of cytoskeleton-associated genes, in particular three GTPases (RAC1, RHOA, CDC42), three kinases (ROCK1, PAK2, LIMK), two Wiskott-Aldrich syndrome proteins (WASL, WASF2) as well as actin related protein 2/3 complex (ARPC2, ARPC3) and cofilin (CFL1). Variability in F-actin content between subjects was high; reduced actin content was only reduced within group evaluation. CONCLUSIONS: Reduced cytoskeleton-associated gene expression after n-3 PUFA supplementation suggests that regulation of cytoskeleton dynamics might be an additional way by which n-3 PUFAs exert their cellular effects. Concerning F-actin, this analysis did not reveal unmistakable results impeding a generalized conclusion.


Asunto(s)
Citoesqueleto/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Complejo 2-3 Proteico Relacionado con la Actina/efectos de los fármacos , Adulto , Cofilina 1/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Expresión Génica/efectos de los fármacos , Humanos , Quinasas Lim/efectos de los fármacos , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Familia de Proteínas del Síndrome de Wiskott-Aldrich/efectos de los fármacos , Proteína Neuronal del Síndrome de Wiskott-Aldrich/efectos de los fármacos , Proteína de Unión al GTP cdc42/efectos de los fármacos , Quinasas p21 Activadas/efectos de los fármacos , Proteína de Unión al GTP rac1/efectos de los fármacos , Quinasas Asociadas a rho/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos
19.
Hypertension ; 65(6): 1273-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870189

RESUMEN

Although a causative role for RhoA-Rho kinase has been recognized in the development of human hypertension, the molecular mechanism(s) and the RhoA guanine exchange factor(s) responsible for the overactivation of RhoA remain unknown. Arhgef1 was identified as a RhoA guanine exchange factor involved in angiotensin II (Ang II)-mediated regulation of vascular tone and hypertension in mice. The aim of this study was to determine whether Arhgef1 is activated and involved in the activation of RhoA-Rho kinase signaling by Ang II in humans. In vitro stimulation of human coronary artery smooth muscle cells and human peripheral blood mononuclear cells by Ang II (0.1 µmol/L) induced activation of Arhgef1 attested by its increased tyrosine phosphorylation. Silencing of Arhgef1 expression by siRNA inhibited Ang II-induced activation of RhoA-Rho kinase signaling. In normotensive subjects, activation of the renin-angiotensin system by a low-salt diet for 7 days increased RhoA-Rho kinase signaling and stimulated Arhgef1 activity in peripheral blood mononuclear cells. In conclusion, our results strongly suggest that Arhgef1 mediates Ang II-induced RhoA activation in humans. Moreover, they show that measurement of RhoA guanine exchange factor activity in peripheral blood mononuclear cells might be a useful method to evaluate RhoA guanine exchange factor activity in humans.


Asunto(s)
Angiotensina II/farmacología , Leucocitos Mononucleares/metabolismo , Músculo Liso Vascular/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Western Blotting , Células Cultivadas , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Leucocitos Mononucleares/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , ARN Mensajero/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/efectos de los fármacos , Transducción de Señal , Estadísticas no Paramétricas , Proteína de Unión al GTP rhoA/efectos de los fármacos
20.
J Neurochem ; 132(3): 354-66, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25156524

RESUMEN

HIV-associated neurocognitive disorders afflict approximately half of HIV-infected patients. HIV-infected cells within the CNS release neurotoxic viral proteins such as the transactivator of transcription (Tat). Tat caused a biphasic change in NMDAR function; NMDA-evoked increases in intracellular Ca(2+) were initially potentiated following 16 h exposure to Tat and then adapted by gradually returning to baseline by 24 h. Following Tat-induced NMDAR potentiation, a RhoA/Rho-associated protein kinase (ROCK) signaling pathway was activated; a subsequent remodeling of the actin cytoskeleton reduced NMDA-evoked increases in intracellular Ca(2+) . Pharmacologic or genetic inhibition of RhoA or ROCK failed to affect potentiation, but prevented adaptation of NMDAR function. Activation of RhoA/ROCK signaling increases the formation of filamentous actin. Drugs that prevent changes to filamentous actin blocked adaptation of NMDAR function following Tat-induced potentiation, whereas stimulating either depolymerization or polymerization of actin attenuated NMDAR function. These findings indicate that Tat activates a RhoA/ROCK signaling pathway resulting in actin remodeling and subsequent reduction of NMDAR function. Adaptation of NMDAR function may be a mechanism to protect neurons from excessive Ca(2+) influx and could reveal targets for the treatment of HIV-associated neurocognitive disorders.


Asunto(s)
Actinas/fisiología , Calcio/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/metabolismo , N-Metilaspartato/farmacología , Neuronas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Animales , ADN/genética , Hipocampo/citología , Neuronas/efectos de los fármacos , Cultivo Primario de Células , Ratas , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rhoA/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA