Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Endocr J ; 70(7): 731-743, 2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37164685

RESUMEN

Glucocorticoids (GCs) are the important stress hormones and widely prescribed as drugs. Although stress has been suggested as a promoter of tumor progression, the direct influence of GCs on metastasis of tumor is not fully understood. Metastasis is a major cause of death in pancreatic cancer patients. In the present study, we investigated the effect of GCs on progression of pancreatic cancer and elucidated the underlying mechanism. It was found that GCs significantly promote cell adhesion, migration, and invasion of pancreatic cancer cells in vitro and their lung metastasis in vivo. Further mechanistic studies showed that GCs notably up-regulate the expression of a trans-membrane glycoprotein, mucin 1 (MUC1) and increase the activation of AKT. Inhibiting MUC1 expression not only attenuates the activation of AKT, but also significantly reduces the promoting effects of GCs on cell adhesion, migration, invasion, and lung metastasis of pancreatic cancer cells. Moreover, GCs not only significantly up-regulate expression of Rho-associated kinase 1/2 (ROCK1/2) and matrix metalloproteinase 3 and 7 (MMP3/7), but also activate ROCK2, which are also involved in the pro-migratory and pro-invasive effects of GCs in pancreatic cancer cells. Taken together, our findings reveal that GCs promote metastasis of pancreatic cancer cells through complex mechanism. MUC1-PI3K/AKT pathway, ROCK1/2 and MMP3/7 are involved in the promoting effect of GCs on cell migration, invasion and metastasis in pancreatic cancer cells. These results suggest the importance of reducing stress and GCs administration in patients with pancreatic cancer to avoid an increased risk of cancer metastasis.


Asunto(s)
Adhesión Celular , Movimiento Celular , Glucocorticoides , Neoplasias Pulmonares , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Pancreáticas , Glucocorticoides/farmacología , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pancreáticas/patología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Invasividad Neoplásica/patología , Quinasas Asociadas a rho/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo , Metaloproteinasa 3 de la Matriz/efectos de los fármacos , Metaloproteinasa 3 de la Matriz/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
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
4.
J Integr Neurosci ; 19(4): 651-662, 2020 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-33378839

RESUMEN

Recent studies have shown that Nogo-A and the Nogo-A receptor affect ß-amyloid metabolism and the downstream Rho GTP enzyme signaling pathway, which may affect the levels of ß-amyloid and tau. Nogo-A may play a key role in the pathogenesis of Alzheimer's disease. However, the underlying molecular mechanisms of Fasudil treatment in Alzheimer's disease are not yet clear. Our results have found that Fasudil treatment for two months substantially ameliorated behavioral deficits, diminished ß-amyloid plaque and tau protein pathology, and alleviated neuronal apoptosis in APP/PS1 transgenic mice. More importantly, two well-established markers for synaptic function, growth-associated protein 43 and synaptophysin, were upregulated after Fasudil treatment. Finally, the levels of Nogo-A, Nogo-A receptor complex NgR/p75NTR/LINGO-1 and the downstream Rho/Rho kinase signaling pathway were significantly reduced. These findings suggest that Fasudil exerts its neuroprotective function in Alzheimer's disease by inhibiting the Nogo-A/NgR1/RhoA signaling pathway.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/efectos de los fármacos , Apoptosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas tau/efectos de los fármacos , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nogo/efectos de los fármacos , Receptor Nogo 1/efectos de los fármacos , Quinasas Asociadas a rho/efectos de los fármacos
5.
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
6.
Biol Pharm Bull ; 43(3): 432-439, 2020 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-31875579

RESUMEN

Salvia przewalskii Maxim is a traditional Chinese herbal medicine and is known to have antibacterial, antiviral, anti-oxidant, anti-thrombotic and anti-depressant properties. However, the major active components of S. przewalskii and its anti-hypoxic effects are still unclear. This study probed the major active component and anti-hypoxic activity of S. przewalskii. The major active components of S. przewalskii were detected by HPLC. The anti-hypoxic effects of S. przewalskii were detected in mice and a rat model of hypoxic preconditioning. The results showed that there are eight active components, including sodium danshensu, rosmarinic acid, lithospermic acid, salvianolic acid B, dihydrotanshinone I, cryptotanshinone, tanshinone I and tanshinone IIA, and each component showed a certain anti-hypoxic effect. Moreover, S. przewalskii enhanced anti-hypoxia in mice, which was manifested as prolonged survival time in acute hypoxic preconditioning and the amelioration of acute hypoxia-induced changes in the activity of superoxide dismutase (SOD) and lactate dehydrogenase (LDH). In addition, S. przewalskii also repaired tissue damage in chronic hypoxia by downregulating hypoxia inducible factor-1α (HIF-1α), proliferating cell nuclear antigen (PCNA), Bcl-2, CDK4, CyclinD1 and P27Kip1 and inhibiting pro-inflammatory cytokines and the RhoA-Rho-associated protein kinase (ROCK) signalling pathway. Our findings provide new insight into the anti-hypoxic effect of S. przewalskii as a promising agent for high-altitude pulmonary hypertension treatment.


Asunto(s)
Hipoxia/tratamiento farmacológico , Extractos Vegetales/farmacología , Salvia/química , Quinasas Asociadas a rho/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo , Animales , Encéfalo/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Citocinas/efectos de los fármacos , Corazón/efectos de los fármacos , Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/inducido químicamente , Ratones , Modelos Animales , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
7.
Pathol Oncol Res ; 26(2): 947-954, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30915607

RESUMEN

Malignant glioma is the most common primary malignancy in the brain. It is aggressive, highly invasive, and destructive. Studies have shown that sevoflurane can affect the invasion and migration of a variety of malignant tumors. However, its effects on human glioma cells and related mechanisms are not clear. Cultured U251 and U87 cells were pretreated with sevoflurane. The effect of sevoflurane on cell proliferation, migration, apoptosis and invasion ability were evaluated by MTT, wound healing assay, cell apoptosis and transwell assays, respectively. miRNA-124-3p and ROCK1 signaling pathway genes expression in sevoflurane treated cell lines was measured by quantitative real-time PCR (qRT-PCR) and western blotting analysis. The potential target genes of miRNA were predicted by online software. Luciferase reporter assay was employed to validate the direct targeting of ROCK1 by miRNA-124-3p. In present studies, sevoflurane inhibits glioma cells proliferation, invasion and migration. Additionally, inversely correlation between miR-124-3p and ROCK1 expression in sevoflurane treated glioma cells was observed. Furthermore, sevoflurane inhibits glioma cells proliferation, migration and invasion through miR-124-3p/ROCK1 axis. Taken together, our study revealed that sevoflurane can inhibit glioma cell proliferation, invasion and migration. Its mechanism may be related to the upregulation of miR-124-3p, which suppresses ROCK1 signaling pathway. The results of the study will help to understand the pharmacological effects of inhaled general anesthetics more comprehensively and help to provide an experimental basis for selecting more reasonable anesthetics for cancer patients.


Asunto(s)
Anestésicos por Inhalación/farmacología , Glioma/patología , MicroARNs/efectos de los fármacos , Sevoflurano/farmacología , Quinasas Asociadas a rho/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , MicroARNs/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
8.
J Trauma Acute Care Surg ; 87(6): 1346-1353, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31464869

RESUMEN

BACKGROUND: Vascular hyporeactivity plays an important role in organ dysfunction induced by endotoxic shock. Given that cytokine, such as TNF-α, plays an important role in endotoxic shock, the aim of the present study is to investigate the role of Tumor Necrosis Factor (TNF)-α in vascular hyporeactivity following endotoxic shock and the mechanisms. METHODS: Lipopolysaccharide (LPS) (1 mg/kg) injection was used for replicating the endotoxic shock model in the rabbit. The changes in the level of TNF-α in plasma in the rabbits model and the contractile response of superior mesenteric arteries (SMA) to norepinephrine (NE) and Ca were observed. The mechanisms in TNF-α-induced vascular hyporeactivity were further explored. RESULTS: The levels of TNF-α in plasma were gradually increased after 1 hour of LPS administration and reached the peak at 6 hours. The contractile responses of SMA to NE were decreased at 1 hour of LPS and lowest at 6 hour. TNF-α (200 ng/mL) incubation decreased contractile response of SMA to NE significantly. Further studies found that calcium desensitization participated in the occurrence of TNF-α-induced vascular hyporeactivity, the changes were consistent with the changes of vascular reactivity, calcium sensitivities were decreased significantly at 1 hour, 2 hours, 4 hours, and 6 hours after LPS injection. TNF-α (200 ng/mL) incubation could significantly reduce the contractile response of SMA to Ca. The activity of Rho-kinase and the changes of myosin light chain 20 (MLC20) phosphorylation level were significantly decreased at 6 hours following LPS administration, and TNF-α (200 ng/mL) incubation led to a decrease of Rho-kinase and MLC20 phosphorylation. Arginine vasopressin significantly antagonized TNF-α (200 ng/mL)-induced the decrease of the vascular reactivity and calcium sensitivity. CONCLUSION: TNF-α is involved in vascular hyporeactivity after endotoxic shock. Calcium desensitization plays an important role in TNF-α-induced vascular hyporeactivity after endotoxic shock. Rho-kinase/MLC20 phosphorylation pathway takes part in the regulation of calcium desensitization and vascular hyporeactivity induced by TNF-α. Arginine vasopressin is beneficial to endotoxic shock in TNF-α-induced vascular hyporeactivity.


Asunto(s)
Choque Séptico/fisiopatología , Factor de Necrosis Tumoral alfa/fisiología , Vasoconstricción , Animales , Arginina Vasopresina/farmacología , Calcio/metabolismo , Femenino , Masculino , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Conejos , Factor de Necrosis Tumoral alfa/sangre , Vasoconstricción/efectos de los fármacos , Quinasas Asociadas a rho/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
9.
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
10.
J Hypertens ; 37(5): 1083-1095, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30418321

RESUMEN

OBJECTIVE: Primary aldosteronism is one of the most common cause of secondary hypertension. It is well known that the incidence of cardiovascular events is higher in patients with primary aldosteronism than in patients with essential hypertension. In a previous study, we showed that aldosterone-producing adenoma is associated with vascular function and structure. The aim of this study was to evaluate the effects of eplerenone on vascular function in the macrovasculature and microvasculature, arterial stiffness and Rho-associated kinase (ROCK) activity in patients with idiopathic hyperaldosteronism (IHA). METHODS: Vascular function, including reactive hyperemia index (RHI), flow-mediated vasodilation (FMD) and nitroglycerine-induced vasodilation (NID), arterial stiffness including brachial-ankle pulse wave velocity (baPWV) and brachial intima-media thickness (IMT) and ROCK activity in peripheral leukocytes were measured before and after 12 weeks of treatment with eplerenone in 50 patients with IHA. RESULTS: After 12 weeks, eplerenone decreased the aldosterone renin ratio but did not alter SBP and DBP. Eplerenone treatment increased log RHI from 0.56 ±â€Š0.25 to 0.69 ±â€Š0.25 (P < 0.01) and NID from 12.8 ±â€Š5.8 to 14.9 ±â€Š6.9% (P = 0.02) and it decreased baPWV from 1540 ±â€Š263 to 1505 ±â€Š281 (P = 0.04) and ROCK activity from 1.20 ±â€Š0.54 to 0.89 ±â€Š0.42 (P < 0.01), whereas there was no significant change in FMD (increase from 4.6 ±â€Š3.4 to 4.6 ±â€Š3.6%, P = 0.99) or brachial IMT (decrease from 0.28 ±â€Š0.07 to 0.28 ±â€Š0.04 mm, P = 0.14). CONCLUSION: Eplerenone improves microvascular endothelial function, vascular smooth muscle function, arterial stiffness and ROCK activity in patients with IHA. CLINICAL TRIAL REGISTRATION INFORMATION: URL for Clinical Trial: http://UMIN; Registration Number for Clinical Trial: UMIN000003409.


Asunto(s)
Antihipertensivos/farmacología , Eplerenona/farmacología , Hiperaldosteronismo/complicaciones , Hipertensión/tratamiento farmacológico , Quinasas Asociadas a rho/metabolismo , Adulto , Aldosterona/sangre , Índice Tobillo Braquial , Antihipertensivos/uso terapéutico , Presión Sanguínea , Arteria Braquial/diagnóstico por imagen , Endotelio Vascular/fisiopatología , Eplerenona/uso terapéutico , Femenino , Humanos , Hiperaldosteronismo/sangre , Hiperemia/fisiopatología , Hipertensión/etiología , Hipertensión/fisiopatología , Masculino , Microvasos/fisiopatología , Persona de Mediana Edad , Nitroglicerina/farmacología , Proyectos Piloto , Análisis de la Onda del Pulso , Renina/sangre , Ultrasonografía , Rigidez Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Quinasas Asociadas a rho/efectos de los fármacos
11.
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
12.
Kidney Blood Press Res ; 43(5): 1425-1436, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30212830

RESUMEN

BACKGROUND/AIMS: Endothelial-to-mesenchymal transition (EndMT) of glomerular endothelial cells (GEnCs) can induce albuminuria in diabetic nephropathy. Melatonin attenuates diabetic nephropathy, but its role and mechanism in EndMT of GEnCs in diabetic nephropathy remain unknown. METHODS: The effect of melatonin on EndMT induced by transforming growth factor (TGF)-ß2 in human renal GEnCs was determined by assaying the expression of endothelial marker cells (VE-cadherin and CD31) and mesenchymal cells (α-SMA and Snail), as well as monolayer permeability. The molecular mechanism of melatonin in these processes was focused on miR-497/ROCK signaling. Furthermore, the effect and mechanism of melatonin in EndMT were confirmed in glomeruli of rats with streptozotocin-induced diabetes. RESULTS: Melatonin increased expression of VE-cadherin and CD31 and inhibited α-SMA and Snail levels that were altered by TGF-ß2 in GEnCs. Melatonin treatment reduced expression and activity of ROCK1 and ROCK2, which suppressed TGF-ß2-induced hyperpermeability of GEnCs and EndMT of GEnCs. Melatonin reduced ROCK1 and ROCK2 expression and activity in TGF-ß2-stimulated GEnCs by enhancing expression of miR-497, which targets ROCK1 and ROCK2. Furthermore, we found that melatonin inhibited EndMT in glomeruli and albuminuria in rats with streptozotocin-induced diabetes. MiR-497 expression increased, whereas ROCK1 and ROCK2 expression and activity decreased in melatonin-treated diabetic rats. CONCLUSION: Melatonin attenuated EndMT of GEnCs via regulating miR-497/ROCK signaling in diabetic nephropathy. This study improves understanding of EndMT and the role of melatonin in diabetic nephropathy.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Nefropatías Diabéticas/patología , Células Endoteliales/patología , Melatonina/farmacología , Células Madre Mesenquimatosas/patología , MicroARNs/efectos de los fármacos , Quinasas Asociadas a rho/efectos de los fármacos , Animales , Línea Celular , Diabetes Mellitus Experimental , Nefropatías Diabéticas/etiología , Humanos , Glomérulos Renales/patología , Masculino , MicroARNs/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta2/farmacología , Quinasas Asociadas a rho/metabolismo
13.
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
14.
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
15.
Asian J Androl ; 20(4): 372-378, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29516877

RESUMEN

We evaluated whether LIM-kinase 2 inhibitor (LIMK2i) could improve erectile function by suppressing corporal fibrosis through the normalization of the Rho-associated coiled-coil protein kinase 1 (ROCK1)/LIMK2/Cofilin pathway in a rat model of cavernous nerve crush injury (CNCI). Sixty 11-week-old male Sprague-Dawley rats were divided equally into five groups: sham surgery (S), CNCI (I), and CNCI treated with low-dose (L), medium-dose (M), and high-dose (H) LIMK2i. The L, M, and H groups were treated with a daily intraperitoneal injection of LIMK2i (2.5, 5.0, and 10.0 mg kg-1 body weight, respectively) for 1 week after surgery. The erectile response was assessed using electrostimulation at 1 week, postoperatively. Penile tissues were processed for Masson's trichrome staining, double immunofluorescence, and Western blot assay. Erectile responses in the H group improved compared with the I group, while the M group showed only partial improvement. A significantly decreased smooth muscle/collagen ratio and an increased content of fibroblasts positive for phospho-LIMK2 were noted in the I group. The M and H groups revealed significant improvements in histological alterations and the dysregulated LIMK2/Cofilin pathway, except for LIMK2 phosphorylation in the M group. The inhibition of LIMK2 did not affect the ROCK1 protein expression. The content of fibroblasts positive for phospho-LIMK2 in the H group returned to the level found in the S group, whereas it did not in the M group. However, the L group did not exhibit such improvements. Our data suggest that the inhibition of LIMK2, particularly with administration of 10.0 mg kg-1 body weight LIMK2i, can improve corporal fibrosis and erectile function by normalizing the LIMK2/Cofilin pathway.


Asunto(s)
Disfunción Eréctil/tratamiento farmacológico , Quinasas Lim/antagonistas & inhibidores , Enfermedades del Pene/tratamiento farmacológico , Pene/inervación , Traumatismos de los Nervios Periféricos/complicaciones , Animales , Cofilina 1/efectos de los fármacos , Cofilina 1/metabolismo , Estimulación Eléctrica , Disfunción Eréctil/etiología , Fibroblastos/patología , Fibrosis/tratamiento farmacológico , Masculino , Enfermedades del Pene/complicaciones , Traumatismos de los Nervios Periféricos/patología , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Quinasas Asociadas a rho/efectos de los fármacos , Quinasas Asociadas a rho/genética
16.
J Clin Endocrinol Metab ; 102(6): 2100-2110, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28368445

RESUMEN

Context: Increased microparticle (MP) shedding by placental trophoblasts contributes to maternal vascular inflammatory response and endothelial dysfunction in preeclampsia. Vitamin D has beneficial effects in pregnancy; however, its effect on trophoblast MP release has not been investigated. Objective: To investigate if vitamin D could protect trophoblasts from oxidative stress-induced MP release. Design: Placental trophoblasts were isolated from uncomplicated and preeclamptic placentas. Effects of vitamin D on MP release induced by oxidative stress inducer CoCl2 were studied. Main Outcome Measures: Annexin V+ MPs were assessed by flow cytometry. Expression of caveolin-1, endothelial nitric oxide synthase (eNOS), procaspase-3, cleaved caspase-3, and Rho-associated coiled-coil protein kinase 1 (ROCK1) in trophoblasts and trophoblast-derived MPs were determined by Western blot. Results: Trophoblasts from preeclamptic pregnancies released significantly more MPs than cells from uncomplicated pregnancies (P < 0.01). CoCl2-induced increase in MP release was associated with upregulation of caveolin-1 and downregulation of eNOS expression in trophoblasts (P < 0.05), which could be attenuated by 1,25(OH)2D3. Moreover, 1,25(OH)2D3 could also inhibit CoCl2-induced procaspase-3 cleavage and ROCK1 activation in trophoblasts. Consistently, CoCl2-induced upregulation of procaspase-3, cleaved caspase-3, and ROCK1 expression in trophoblast-derived MPs were also reduced in cells treated with 1,25(OH)2D3. Conclusions: Placental trophoblasts from preeclamptic pregnancies released more MP than cells from uncomplicated pregnancies. Oxidative stress-induced increase in MP shedding is associated with upregulation of caveolin-1 and downregulation of eNOS expression in placental trophoblasts. Inhibition of caspase-3 cleavage and ROCK1 activation, together with upregulation of eNOS expression, could be the potential cellular/molecular mechanism(s) of vitamin D protective effects on placental trophoblasts.


Asunto(s)
Calcitriol/farmacología , Caspasa 3/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Trofoblastos/efectos de los fármacos , Vitaminas/farmacología , Quinasas Asociadas a rho/efectos de los fármacos , Adolescente , Adulto , Western Blotting , Estudios de Casos y Controles , Caspasa 3/metabolismo , Caveolina 1/efectos de los fármacos , Caveolina 1/metabolismo , Micropartículas Derivadas de Células/efectos de los fármacos , Micropartículas Derivadas de Células/metabolismo , Cobalto/farmacología , Femenino , Humanos , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Preeclampsia , Embarazo , Trofoblastos/metabolismo , Adulto Joven , Quinasas Asociadas a rho/metabolismo
17.
Am J Hypertens ; 30(4): 389-399, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28164209

RESUMEN

BACKGROUND: We investigated the influence of salt overconsumption on the functionality of the RhoA/Rho-associated kinase (ROCK) pathway and calcium regulation in arteries. METHODS: The aorta and small mesenteric arteries from rats fed a chow containing 2%, 4%, or 8% NaCl were evaluated in organ baths for the activity of the RhoA/ROCK pathway and intracellular calcium mobilization. Components of these pathways and intracellular calcium levels were also assessed in samples from 4% NaCl group. RESULTS: In arteries from animals fed regular chow, the ROCK inhibitor Y-27632 reduced the responses to phenylephrine, even when the smallest concentrations (1 and 3 µM) were tested. However, only higher concentrations of Y-27632 (10 and 50 µM) reduced phenylephrine-induced contraction in vessels from high-salt groups. Immunoblotting revealed augmented phosphorylation of the myosin phosphatase targeting subunit 1 and increased amounts of RhoA in the membrane fraction of aorta homogenates from the 4% NaCl group. Under calcium-free solution, vessels from NaCl groups presented reduced contractile responses to phenylephrine and caffeine, compared with the regular chow group. Moreover, decreased intracellular calcium at rest and after stimulation with ATP were found in aortic smooth muscle cells from 4% NaCl-fed rats, which also showed diminished levels of SERCA2 and SERCA3, but not of IP3 and ryanodine receptors, or STIM1 and Orai1 proteins. CONCLUSIONS: Arteries from rats subjected to high-salt intake are unable to properly regulate intracellular calcium levels and present augmented activity of the calcium sensitization pathway RhoA/ROCK. These changes may precede the development of vascular diseases induced by high-salt intake.


Asunto(s)
Aorta/efectos de los fármacos , Calcio/metabolismo , Arterias Mesentéricas/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Cloruro de Sodio Dietético/farmacología , Vasoconstricción/efectos de los fármacos , Proteínas de Unión al GTP rho/efectos de los fármacos , Quinasas Asociadas a rho/efectos de los fármacos , Amidas/farmacología , Animales , Aorta/citología , Aorta/metabolismo , Inhibidores Enzimáticos/farmacología , Receptores de Inositol 1,4,5-Trifosfato/efectos de los fármacos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteína ORAI1/efectos de los fármacos , Proteína ORAI1/metabolismo , Fenilefrina/farmacología , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 1/efectos de los fármacos , Proteína Fosfatasa 1/metabolismo , Piridinas/farmacología , Ratas , Ratas Wistar , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal/efectos de los fármacos , Molécula de Interacción Estromal 1/efectos de los fármacos , Molécula de Interacción Estromal 1/metabolismo , Vasoconstrictores/farmacología , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
18.
Ann Thorac Surg ; 103(2): 476-483, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27743639

RESUMEN

BACKGROUND: Rho-kinase, an intracellular serine/threonine kinase, is a key regulator of cytoskeletal dynamics. Recent studies have demonstrated that Rho-kinase is involved in the ischemia-reperfusion injury (IRI) pathogenesis of many organs; however, its involvement with lung IRI remains unclear. This study assessed the association of Rho-kinase with lung IRI and evaluated the protective effect of inhaled Rho-kinase inhibitors in lung IRI. METHODS: The study included isolated rat lung perfusion models, divided into three groups: sham, Rho-kinase inhibitor, and warm ischemia (n = 6 each). The lungs were exposed to 60 minutes of warm ischemia by perfusion cessation. At the onset of ischemia, nebulized fasudil, a novel Rho-kinase inhibitor, and saline were inhaled in the Rho-kinase inhibitor and warm ischemia groups, respectively. Perfusion was restarted after the ischemic period, and physiologic data were collected for 90 minutes. Lungs in the sham group were continuously perfused without ischemia or drug administrations. Protein expression in tissue specimens related to the Rho-kinase pathway was evaluated by Western blotting. RESULTS: Warm ischemia and subsequent reperfusion enhanced Rho-kinase activity, and this was suppressed by fasudil inhalation. Fasudil inhalation significantly attenuated IRI pathophysiology, including pulmonary vascular contraction, dynamic compliance, lung edema, and oxygenation. Molecular analysis showed that Rho-kinase suppressed myosin phosphatase and endothelial nitric oxide synthase activities, suggesting these are downstream targets of Rho-kinase during lung IRI pathogenesis. CONCLUSIONS: The present study suggests that Rho-kinase activation is involved in lung IRI pathogenesis and that inhaled Rho-kinase inhibitors may attenuate this pathogenesis.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Pulmón/patología , Pulmón/cirugía , Óxido Nítrico Sintasa de Tipo III/metabolismo , Daño por Reperfusión/prevención & control , Quinasas Asociadas a rho/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Administración por Inhalación , Animales , Biopsia con Aguja , Modelos Animales de Enfermedad , Inmunohistoquímica , Técnicas In Vitro , Masculino , Perfusión , Circulación Pulmonar/fisiología , Distribución Aleatoria , Ratas , Ratas Endogámicas Lew , Valores de Referencia , Sensibilidad y Especificidad , Resistencia Vascular/efectos de los fármacos , Quinasas Asociadas a rho/efectos de los fármacos
19.
Diabetes ; 66(2): 505-518, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27856608

RESUMEN

The angiotensin (ANG)-(1-7)/Mas receptor (MasR) pathway activates vascular repair-relevant functions of bone marrow progenitor cells. We tested the effects of ANG-(1-7) on mobilization and vasoreparative functions of progenitor cells that are impaired in diabetes. The study was performed in streptozotocin-induced diabetic (db/db) mice. Diabetes resulted in a decreased number of Lineage-Sca-1+c-Kit+ (LSK) cells in the circulation, which was normalized by ANG-(1-7). Diabetes-induced depletion of LSK cells in the bone marrow was reversed by ANG-(1-7). ρ-Kinase (ROCK) activity was increased specifically in bone marrow LSK cells by ANG-(1-7) in diabetes, and the beneficial effects of ANG-(1-7) were prevented by fasudil. ANG-(1-7) increased Slit3 levels in the bone marrow supernatants, which activated ROCK in LSK cells and sensitized them for stromal-derived factor-1α (SDF)-induced migration. Diabetes prevented the mobilization of LSK cells in response to ischemia and impaired the recovery of blood flow, both of which were reversed by ANG-(1-7) in both models of diabetes. Genetic ablation of MasR prevented ischemia-induced mobilization of LSK cells and impaired blood flow recovery, which was associated with decreased proliferation and migration of LSK cells in response to SDF or vascular endothelial growth factor. These results suggest that MasR is a promising target for the treatment of diabetic bone marrow mobilopathy and vascular disease.


Asunto(s)
Angiotensina I/farmacología , Vasos Sanguíneos/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Diabetes Mellitus Experimental/fisiopatología , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Células Madre/efectos de los fármacos , Vasodilatadores/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Vasos Sanguíneos/fisiopatología , Médula Ósea/efectos de los fármacos , Médula Ósea/fisiopatología , Linaje de la Célula , Quimiocina CXCL12/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Isquemia , Masculino , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Recuperación de la Función/efectos de los fármacos , Regeneración , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinasas Asociadas a rho/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...