Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Am J Cancer Res ; 13(5): 2030-2040, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37293174

RESUMEN

Therapeutic resistance to androgen-deprivation therapy is a major challenge for prostate cancer therapy. The present study aims to explore the effects of poly (ADP-ribose) polymerase (PARP) inhibitor olaparib and STL127705 on castration-resistant prostate cancer. Cell lines including PC-3 and enzalutamide-resistant LNCaP (erLNCaP) cells were treated with enzalutamide, enzalutamide plus olaparib, enzalutamide plus STL127705, or the combination of olaparib, STL127705, and enzalutamide. Cell viabilities and cell apoptosis were determined using the sulforhodamine B (SRB) assay and Annexin V/propidium iodide staining, respectively. Flow cytometry assay was applied to determine γH2AX intensity and the percentage of homologous recombination and non-homologous end-joining. Besides, a tumor-bearing animal model was established and treated with drugs as for cell lines. STL127705 and olaparib enhanced cytotoxicity of enzalutamide on erLNCaP and PC-3 cells. Furthermore, STL127705 and olaparib promoted enzalutamide-induced cell apoptosis and enhanced γH2AX intensity. In vitro study also showed that the combination of STL127705, olaparib, and enzalutamide inhibited homologous recombination and non-homologous end-joining repair systems in PC-3 cells. In vivo study demonstrated that the combination of STL127705, olaparib, and enzalutamide exhibited a significant anti-tumor effect. STL127705 combined with olaparib have a potential therapeutic effect on castration-resistant prostate cancer through inhibiting homologous recombination and non-homologous end-joining repair.

2.
Life Sci ; 317: 121491, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36758669

RESUMEN

AIMS: Diabetes mellitus (DM) has become a global problem, causing a huge economic burden. The purpose of this study is to find a new potential method and mechanism for the treatment of DM. MAIN METHODS: The oxidation, glycation and insulin resistance cell models were built to screen the potential anti-diabetic chemicals. Then the DM mice were induced by the combination of high-fat diet (HFD) and intraperitoneal injection of streptozotocin (50 mg/kg) for five days. The alfuzosin (1.2 mg/kg) was administered by intraperitoneal injection once daily for sequential 12 weeks. Fasting blood glucose, blood lipid, oxidative stress and key markers of glucose metabolism were detected. PGK1/AKT/GLUT4 pathway related proteins were analyzed by Western blot. KEY FINDINGS: Alfuzosin ameliorated oxidative stress, glycative stress and insulin resistance in HepG2 cells. Further, in a high-fat diet/streptozotocin (HFD/STZ)-induced diabetic mouse model, alfuzosin reduced fasting blood glucose, improved insulin sensitivity. Mechanically, alfuzosin activated PGK1 directly to stimulate the protein kinase B (AKT) signaling pathway, thus facilitating glucose uptake as well as improving insulin resistance. SIGNIFICANCE: The present finding has shed a new light on the treatment of DM and provides validation for PGK1 as a therapeutic target for DM.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Animales , Ratones , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Hipoglucemiantes/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estreptozocina
3.
Biochim Biophys Acta Gen Subj ; 1867(3): 130303, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36627088

RESUMEN

Globally, diabetes mellitus has been a major epidemic bringing metabolic and endocrine disorders. Currently, 1 in 11 adults suffers from diabetes mellitus, among the patients >90% contract type 2 diabetes mellitus (T2DM). Therefore, it is urgent to develop new drugs that effectively prevent and treat type 2 diabetes through new targets. With high-throughput screening, we found that sulfathiazole decreased the blood glucose and improved glucose metabolism in T2DM mice. Notably, we discovered that sulfathiazole treated T2DM by activating CYP19A1 protein to synthesize estrogen. Collectively, sulfathiazole along with CYP19A1 target bring new promise for the better therapy of T2DM.


Asunto(s)
Aromatasa , Diabetes Mellitus Tipo 2 , Sulfatiazoles , Animales , Ratones , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Estrógenos , Sulfatiazoles/uso terapéutico , Aromatasa/efectos de los fármacos
4.
Eur J Pharmacol ; 938: 175432, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36460132

RESUMEN

Obesity, a global epidemic chronic metabolic disease, urgently demands novel therapies. As an antimalarial drug, quinacrine has not been reported for its anti-obesity effect to our knowledge. This study aimed to explore the ability of quinacrine to attenuate obesity. In an in vitro adipogenic model, quinacrine exhibited an outstanding suppression on adipogenesis of 3T3-L1 cells, mainly by activating the AMPK (Adenosine 5'-monophosphate (AMP)-activated protein kinase) signaling pathway to regulate preadipocytes differentiation and lipid accumulation. In addition, C57BL/6N female mice were fed with high-fat diet and high-fructose water for 14 weeks to establish an obesity model, followed by oral administration of quinacrine or orlistat. After 9 weeks of treatment, quinacrine significantly reduced the body weight and energy intake, ameliorated the impaired glucose tolerance and restored the homeostasis of serum lipids. Also, quinacrine improved lipid profile and optimized the expression of AMPK signaling pathway related proteins in livers and adipose tissues of obese mice. Quinacrine reverses obesity through activating AMPK phosphorylation to down-regulate adipogenesis, along with lowering the risk of type 2 diabetes and atherosclerosis. It should be a novel application for the treatment of obesity and its associated diseases.


Asunto(s)
Fármacos Antiobesidad , Diabetes Mellitus Tipo 2 , Femenino , Ratones , Animales , Adipogénesis , Proteínas Quinasas Activadas por AMP/metabolismo , Quinacrina/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Adipocitos , Ratones Endogámicos C57BL , Células 3T3-L1 , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/metabolismo , Dieta Alta en Grasa/efectos adversos , Fármacos Antiobesidad/farmacología , Transducción de Señal , Lípidos
5.
J Diabetes Metab Disord ; 21(2): 1731-1741, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36404863

RESUMEN

Background: The global prevalence of type 2 diabetes mellitus (T2DM) raises the rates of its complications, such as diabetic nephropathy and cardiovascular diseases. To conquer the complications, new strategies to reverse the deterioration of T2DM are urgently needed. In this project, we aimed to examine the hypoglycemic effect of primaquine and explore its specific target. Methods: In vitro T2DM insulin resistance model was built in HepG2 cells to screen the potential anti-diabetic chemicals. On the other hand, the potential protein targets were explored by molecular docking. Accordingly, we chose C57BL/6 N mice to establish T2DM model to verify the effect of the chemicals on anti-hyperglycemia and diabetic complications. Results: By targeting the Keratin 7 (K7) to activate EGFR/Akt glucose metabolism signaling pathway, primaquine poses a potent hypoglycemic effect. The level of acetyl-CoA is enhanced markedly, supporting that primaquine upregulates the aerobic glycolysis. Moreover, primaquine ameliorates kidney function by reducing the secretion of urinary proteins and creatinine, especially for the urea nitrogen which is significantly decreased compared to no-treatment T2DM mice. Notably, primaquine restores the level of plasma low-density lipoprotein cholesterol (LDL-C) nearly to normal, minimizing the incidence of cardiovascular diseases. Conclusions: We find that primaquine may reverse the dysregulated metabolism to prevent diabetic complications by stimulating EGFR/Akt signaling axis, shedding new light on the therapy of T2DM. Graphical abstract: Insulin resistance is characterized by reduced p-Akt and glucose metabolism, dominated by anaerobic glycolysis. Primaquine activates the complex made of K7 and EGFR, further stimulating Akt phosphorylation. Then, p-Akt promotes the aerobic glucose metabolism and upregulates Ac-CoA to mobilize TCA cycle, improving insulin sensitivity. Supplementary Information: The online version contains supplementary material available at 10.1007/s40200-022-01135-8.

6.
Life Sci ; 304: 120725, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-35751919

RESUMEN

AIMS: Diabetes mellitus (DM) is a major global health threat characterized by insulin resistance. A new tactic to ameliorate insulin resistance, thereby reversing the exacerbation of DM, is urgently needed. The work is aiming to provide a new strategy for DM treatment as well as to identify new targets. MAIN METHODS: C57BL/6 N mice were raised with high-fat diet (HFD) and infused with streptozotocin (STZ) to induce diabetes. The blood glucose, serum insulin, blood lipid and oxidative stress were detected. In vitro insulin resistance model experiment has been made to examine the molecular mechanisms underlying anti-diabetic effect of potential active chemicals in human hepatocellular carcinoma cells (HepG2). KEY FINDINGS: Acyclovir, an antiviral nucleotide analog, alleviates insulin resistance by reducing blood lipids as well as oxidative stress and elevating insulin sensitivity on diabetic mice, which is in accord with results in the insulin resistance model of HepG2 cells. Mechanically, acyclovir stimulates pyruvate kinase M1 (PKM1) directly to activate adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK)/Sirtuin1 (SIRT1) signaling pathway, thus improving insulin resistance. SIGNIFICANCE: The present study supports that acyclovir should be translated to remedy DM, and PKM1 might be a valuable target to develop new medicines.


Asunto(s)
Diabetes Mellitus Experimental , Resistencia a la Insulina , Proteínas Quinasas Activadas por AMP/metabolismo , Aciclovir , Animales , Diabetes Mellitus Experimental/metabolismo , Dieta Alta en Grasa/efectos adversos , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Piruvato Quinasa
7.
J Cancer ; 13(15): 3652-3659, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36606191

RESUMEN

Background: miR-143 is known to be downregulated in various cancer cells and tumors and generally plays a tumor-suppressor role. miR-143. However, the role of miR-143 in the mediation of the sensitivity of prostate cancer cells to abiraterone acetate remains unrevealed. Methods: The expression levels of miRNAs were determined by miRNA microarray and quantitative real-time PCR (qRT-PCR). The protein levels were assessed by Western blot assay. Cell viability and apoptosis were respectively measured by Cell Counting Kit-8 (CCK-8) assay and flow cytometry. Results: We identified that miR-143 was significantly downregulated in PC3-AbiR cells compared to PC3 cells. Overexpression of miR-143 promoted PC-AbiR sensitivity to abiraterone acetate in vitro and in vivo. We also revealed that miR-143 upregulation inhibited p-JNK (c-Jun N-terminal kinases) and increased p-Bcl2 (B-cell lymphoma 2), contributing to abiraterone acetate-induced apoptosis in PC3-AbiR cells. Finally, we showed that the combination of miR-143 and abiraterone acetate exerted the most profound tumor inhibition effect and prolonged the mice survival rate in PC3-AbiR tumor-bearing mice. Conclusion: Upregulation of miR-143 may serve as a new strategy to enhance the therapeutical effect of abiraterone acetate on prostate cancer patients who are resistant to abiraterone acetate.

8.
J BUON ; 26(4): 1549-1555, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34565018

RESUMEN

PURPOSE: To study the effect of triptolide (TP) on radiosensitivity of human glioma U251 cells and its mechanism, so as to provide new ideas and methods for the radiotherapy of glioma. METHODS: U251 cells were treated with 10, 50, 100 nmol/L TP at different concentrations and irradiated with 0, 2, 4, 6, 8 Gy X-ray. The radiosensitivity of cells in each group were detected by MTT. U251 cells were then divided into control group, 10 nmol/L TP group, 4 Gy radiation group, 10 nmol/L TP + 4 Gy radiation group. The formation ability of U251 cells in each group was detected by colony formation assay. Flow cytometry was used to detect cell cycle and apoptosis in each group. Western blot was used to detect the changes of PI3K/Akt signal pathway in each group. RESULTS: When 10, 50, 100 nmol/L TP were combined with 2, 4, 6, 8 Gy X-ray, the proliferation inhibition rate of U251 cells in each group increased significantly (p<0.05); compared with 10 nmol/L TP alone group and 4 Gy radiation alone group, the colony formation ability rate of U251 cells in 10 nmol/L TP + 4Gy radiation combined group decreased significantly (p<0.05), the cell cycle was blocked in G1 phase, and the apoptosis rate was significantly reduced (p<0.05). The level of p-pi3k and p-Akt decreased significantly (p<0.05). CONCLUSION: Triptolide could significantly increase the radiosensitivity of human glioma U251 cells and play a role by inhibiting the PI3K/Akt signal pathway.


Asunto(s)
Diterpenos/farmacología , Glioma/radioterapia , Fenantrenos/farmacología , Tolerancia a Radiación/efectos de los fármacos , Compuestos Epoxi/farmacología , Humanos , Células Tumorales Cultivadas
9.
J Mol Neurosci ; 71(12): 2514-2525, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33786723

RESUMEN

Accumulating evidence suggests that neuronal apoptosis plays a critical role in early brain injury (EBI) after subarachnoid hemorrhage (SAH), and the inhibition of apoptosis can induce neuroprotective effects in SAH animal models. c-Abl has been reported to promote neuronal apoptosis in Alzheimer's disease and cerebral ischemia, but its role in SAH had not been illuminated until now. In the present study, the effect of c-Abl on neuronal apoptosis induced by SAH was investigated. c-Abl protein levels and neuronal apoptosis were markedly increased 24 h after SAH, and the inhibition of endogenous c-Abl reduced neuronal apoptosis and mortality and ameliorated neurological deficits. Furthermore, c-Abl inhibition decreased the expression of cleaved caspase-3 (CC-3) after SAH. These results demonstrate the proapoptotic effect of c-Abl in EBI after SAH. Additionally, c-Abl inhibition further enhanced the SAH-induced phosphorylation of Akt and glycogen synthase kinase (GSK)3ß. LY294002 abrogated the beneficial effects of targeting c-Abl and exacerbated neuronal apoptosis after SAH. SAH decreased LRP-1 levels and downregulated LRP-1 by RAP, and LRP-1 small interfering RNA (siRNA) induced a dramatic decrease in Akt/GSK3ß activation in the presence of c-Abl siRNA. This is the first report showing that the c-Abl tyrosine kinase may play a key role in SAH-induced neuronal apoptosis by regulating the LRP-1-dependent Akt/GSK3ß survival pathway. Thus, c-Abl has the potential to be a novel target for EBI therapy after SAH.


Asunto(s)
Apoptosis , Proteínas Proto-Oncogénicas c-abl/metabolismo , Transducción de Señal , Hemorragia Subaracnoidea/metabolismo , Animales , Caspasa 3/metabolismo , Cromonas/farmacología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Morfolinas/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar
10.
BMC Cancer ; 19(1): 441, 2019 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-31088402

RESUMEN

BACKGROUND: Glioblastoma is a disease with high heterogeneity that has long been difficult for doctors to identify and treat. ARHI is a remarkable tumor suppressor gene in human ovarian cancer and many other cancers. We found over-expression of ARHI can also inhibit cancer cell proliferation, decrease tumorigenicity, and induce autophagic cell death in human glioma and inhibition of the late stage of autophagy can further enhance the antitumor effect of ARHI through inducing apoptosis in vitro or vivo. METHODS: Using MTT assay to detect cell viability. The colony formation assay was used to measure single cell clonogenicity. Autophagy associated morphological changes were tested by transmission electron microscopy. Flow cytometry and TUNEL staining were used to measure the apoptosis rate. Autophagy inhibitor chloroquine (CQ) was used to study the effects of inhibition at late stage of autophagy on ARHI-induced autophagy and apoptosis. Protein expression were detected by Western blot, immunofluorescence and immunohistochemical analyses. LN229-derived xenografts were established to observe the effect of ARHI in vivo. RESULTS: ARHI induced autophagic death in glioma cells, and blocking late-stage autophagy markedly enhanced the antiproliferative activites of ARHI. In our research, we observed the inhibition of RAS-AKT-mTOR signaling in ARHI-glioma cells and blockade of autophagy flux at late stage by CQ enhanced the cytotoxicity of ARHI, caused accumulation of autophagic vacuoles and robust apoptosis. As a result, the inhibition of RAS augmented autophagy of glioma cells. CONCLUSION: ARHI may also be a functional tumor suppressor in glioma. And chloroquine (CQ) used as an auxiliary medicine in glioma chemotherapy can enhance the antitumor effect of ARHI, and this study provides a novel mechanistic basis and strategy for glioma therapy.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Proteínas ras/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Autofagia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Cloroquina/farmacología , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas ras/genética , Proteínas de Unión al GTP rho/genética
11.
Cell Physiol Biochem ; 51(4): 1566-1583, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30497066

RESUMEN

BACKGROUND/AIMS: Glioblastoma multiforme (GBM) is the most devastating and widespread primary central nervous system tumour in adults, with poor survival rate and high mortality rates. Existing treatments do not provide substantial benefits to patients; therefore, novel treatment strategies are required. Peiminine, a natural bioactive compound extracted from the traditional Chinese medicine Fritillaria thunbergii, has many pharmacological effects, especially anticancer activities. However, its anticancer effects on GBM and the underlying mechanism have not been demonstrated. This study was conducted to investigate the potential antitumour effects of peiminine in human GBM cells and to explore the related molecular signalling mechanisms in vitro and in vivo Methods: Cell viability and proliferation were detected with MTT and colony formation assays. Morphological changes associated with autophagy were assessed by transmission electron microscopy (TEM). The cell cycle rate was measured by flow cytometry. To detect changes in related genes and signalling pathways in vitro and in vivo, RNA-seq, Western blotting and immunohistochemical analyses were employed. RESULTS: Peiminine significantly inhibited the proliferation and colony formation of GBM cells and resulted in changes in many tumour-related genes and transcriptional products. The potential anti-GBM role of peiminine might involve cell cycle arrest and autophagic flux blocking via changes in expression of the cyclin D1/CDK network, p62 and LC3. Changes in Changes in flow cytometry results and TEM findings were also observed. Molecular alterations included downregulation of the expression of not only phospho-Akt and phospho-GSK3ß but also phospho-AMPK and phospho-ULK1. Furthermore, overexpression of AKT and inhibition of AKT reversed and augmented peiminine-induced cell cycle arrest in GBM cells, respectively. The cellular activation of AMPK reversed the changes in the levels of protein markers of autophagic flux. These results demonstrated that peiminine mediates cell cycle arrest by suppressing AktGSk3ß signalling and blocks autophagic flux by depressing AMPK-ULK1 signalling in GBM cells. Finally, peiminine inhibited the growth of U251 gliomas in vivo. CONCLUSION: Peiminine inhibits glioblastoma in vitro and in vivo via arresting the cell cycle and blocking autophagic flux, suggesting new avenues for GBM therapy.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Autofagia/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Cevanas/uso terapéutico , Glioblastoma/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Cevanas/farmacología , Femenino , Fritillaria/química , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal/efectos de los fármacos
12.
Cell Death Dis ; 9(10): 1032, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30302016

RESUMEN

Glioblastoma is the most common and aggressive primary brain tumor in adults. New drug design and development is still a major challenge for glioma treatment. Increasing evidence has shown that nitazoxanide, an antiprotozoal drug, has a novel antitumor role in various tumors and exhibits multiple molecular functions, especially autophagic regulation. However, whether nitazoxanide-associated autophagy has an antineoplastic effect in glioma remains unclear. Here, we aimed to explore the underlying molecular mechanism of nitazoxanide in glioblastoma. Our results showed that nitazoxanide suppressed cell growth and induced cell cycle arrest in glioblastoma by upregulating ING1 expression with a favorable toxicity profile. Nitazoxanide inhibited autophagy through blockage of late-stage lysosome acidification, resulting in decreased cleavage of ING1. A combination with chloroquine or Torin1 enhanced or impaired the chemotherapeutic effect of nitazoxanide in glioblastoma cells. Taken together, these findings indicate that nitazoxanide as an autophagy inhibitor induces cell cycle arrest in glioblastoma via upregulated ING1 due to increased transcription and decreased post-translational degradation by late-stage autophagic inhibition.


Asunto(s)
Antiprotozoarios/farmacología , Autofagia/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Proteína Inhibidora del Crecimiento 1/metabolismo , Tiazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cloroquina/farmacología , Glioma/tratamiento farmacológico , Glioma/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Naftiridinas/farmacología , Nitrocompuestos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
13.
Oncol Rep ; 40(2): 867-876, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29989652

RESUMEN

Oxymatrine (OM), a natural quinolizidine alkaloid extracted from the traditional Chinese herb Sophora flavescens, has been revealed to produce antitumor activities in various cancer cell lines, including glioblastoma lines, in vitro. However, the mechanisms by which OM exerts its antitumor effect against glioma are poorly understood. The aim of this study was to investigate the role of OM in the proliferation, apoptosis and invasion of glioma cells and to reveal the underlying mechanisms. The effects of OM on U251MG cells in vitro were determined using a Cell Counting Kit­8 (CCK­8) assay, flow cytometric analysis, Annexin V­FITC/PI staining, DAPI staining, a terminal deoxynucleotidyl transferase­mediated dUTP nick end­labeling (TUNEL) assay, a Transwell assay and western blotting. Our data indicated that OM inhibited proliferation, arrested the cell cycle at the G0/G1 phase, decreased the expression levels of G1 cell cycle regulatory proteins (cyclin D1, CDK4 and CDK6), inhibited invasion and induced apoptosis in glioma cells. Additional investigations revealed that the expression levels of p­STAT3 and key proteins in the EGFR/PI3K/Akt/mTOR signaling pathway, such as p­EGFR, p­Akt and p­mTOR, were markedly decreased after OM treatment, while the total STAT3, EGFR, Akt and mTOR levels were not affected. These findings indicated that the EGFR/PI3K/Akt/mTOR signaling pathway and STAT3 suppression may be a potential mechanism of the OM­mediated antitumor effect in glioblastoma cells and that EGFR may be a target of OM. Hence, OM may be a promising drug and may offer a novel therapeutic strategy for malignant gliomas in the future.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos/farmacología , Glioblastoma/tratamiento farmacológico , Quinolizinas/farmacología , Transducción de Señal/efectos de los fármacos , Sophora/química , Alcaloides/uso terapéutico , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Glioblastoma/patología , Humanos , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolizinas/uso terapéutico , Factor de Transcripción STAT3/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
14.
Cell Physiol Biochem ; 44(4): 1381-1395, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29186708

RESUMEN

BACKGROUND/AIMS: Glioma is the most devastating cancer in the brain and has a poor prognosis in adults. Therefore, there is a critical need for novel therapeutic strategies for the management of glioma patients. Isogambogenic acid, an active compound extracted from the Chinese herb Garcinia hanburyi, induces autophagic cell death. METHODS: Cell viability was detected with MTT assays. Cell proliferation was assessed using the colony formation assay. Morphological changes associated with autophagy and apoptosis were tested by TEM and Hoechst staining, respectively. The apoptosis rate was measured by flow cytometry. Western blot, immunofluorescence and immunohistochemical analyses were used to detect protein expression. U87-derived xenografts were established for the examination of the effect of isogambogenic acid on glioma growth in vivo. RESULTS: Isogambogenic acid induced autophagic death in U87 and U251 cells, and blocking late-stage autophagy markedly enhanced the antiproliferative activities of isogambogenic acid. Moreover, we observed the activation of AMPK-mTOR signalling in isogambogenic acid-treated glioma cells. Furthermore, the activation of AMPK or the inhibition of mTOR augmented isogambogenic acid-induced autophagy. Inhibition of autophagy attenuated apoptosis in isogambogenic acid-treated glioma cells. Finally, isogambogenic acid inhibited the growth of U87 glioma in vivo. CONCLUSION: Isogambogenic acid inhibits the growth of glioma via activation of the AMPK-mTOR signalling pathway, which may provide evidence for future clinical applications in glioma therapy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/toxicidad , Proliferación Celular/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Xantonas/toxicidad , Adenina/análogos & derivados , Adenina/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal/efectos de los fármacos , Trasplante Heterólogo , Xantonas/química , Xantonas/uso terapéutico
15.
Plant Cell ; 29(9): 2214-2232, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28814646

RESUMEN

Plants use both cell surface-resident pattern recognition receptors (PRRs) and intracellular nucleotide binding leucine-rich repeat (NLR) receptors to detect various pathogens. Plant PRRs typically recognize conserved pathogen-associated molecular patterns (PAMPs) to provide broad-spectrum resistance. By contrast, plant NLRs generally detect pathogen strain-specific effectors and confer race-specific resistance. Here, we demonstrate that the tomato (Solanum lycopersicum) NLR Sw-5b confers broad-spectrum resistance against American-type tospoviruses by recognizing a conserved 21-amino acid peptide region within viral movement protein NSm (NSm21). Sw-5b NB-ARC-LRR domains directly associate with NSm21 in vitro and in planta. Domain swap, site-directed mutagenesis and structure modeling analyses identified four polymorphic sites in the Sw-5b LRR domain that are critical for the recognition of NSm21 Furthermore, recognition of NSm21 by Sw-5b likely disturbs the residues adjacent to R927 in the LRR domain to weaken the intramolecular interaction between LRR and NB-ARC domains, thus translating recognition of NSm21 into activation of Sw-5b. Natural variation analysis of Sw-5b homologs from wild tomato species of South America revealed that the four polymorphic sites in the Sw-5b LRR domain were positively selected during evolution and are all necessary to confer resistance to tospovirus. The results described here provide a new example of a plant NLR mediating broad-spectrum resistance through recognition of a small conserved PAMP-like region within the pathogen effector.


Asunto(s)
Secuencia Conservada , Resistencia a la Enfermedad , Epítopos/metabolismo , Enfermedades de las Plantas/inmunología , Proteínas de Plantas/metabolismo , Solanum lycopersicum/inmunología , Solanum lycopersicum/virología , Tospovirus/fisiología , Secuencia de Aminoácidos , Muerte Celular , Modelos Moleculares , Péptidos/química , Enfermedades de las Plantas/virología , Proteínas de Plantas/química , Polimorfismo Genético , Unión Proteica , Dominios Proteicos
16.
New Phytol ; 212(1): 161-75, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27558751

RESUMEN

The tomato resistance protein Sw-5b differs from the classical coiled-coil nucleotide-binding leucine-rich repeat (CC-NB-LRR) resistance proteins by having an extra N-terminal domain (NTD). To understand how NTD, CC and NB-LRR regulate autoinhibition and activation of Sw-5b, we dissected the function(s) of each domain. When viral elicitor was absent, Sw-5b LRR suppressed the central NB-ARC to maintain autoinhibition of the NB-LRR segment. The CC and NTD domains independently and additively enhanced the autoinhibition of NB-LRR. When viral elicitor was present, the NB-LRR segment of Sw-5b was specifically activated to trigger a hypersensitive response. Surprisingly, Sw-5b CC suppressed the activation of NB-LRR, whereas the extra NTD of Sw-5b became a positive regulator and fully activated the resistance protein, probably by relieving the inhibitory effects of the CC. In infection assays of transgenic plants, the NB-LRR segment alone was insufficient to confer resistance against Tomato spotted wilt tospovirus; the layers of NTD and CC regulation on NB-LRR were required for Sw-5b to confer resistance. Based on these findings, we propose that, to counter the negative regulation of the CC on NB-LRR, Sw-5b evolved an extra NTD to coordinate with the CC, thus developing a multilayered regulatory mechanism to control autoinhibition and activation.


Asunto(s)
Nicotiana/genética , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Proteínas/química , Proteínas/metabolismo , Secuencia Conservada , Proteínas Repetidas Ricas en Leucina , Modelos Moleculares , Plantas Modificadas Genéticamente , Dominios Proteicos , Relación Estructura-Actividad , Nicotiana/virología
17.
PLoS Pathog ; 12(2): e1005443, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26863622

RESUMEN

Plant viruses move through plasmodesmata to infect new cells. The plant endoplasmic reticulum (ER) is interconnected among cells via the ER desmotubule in the plasmodesma across the cell wall, forming a continuous ER network throughout the entire plant. This ER continuity is unique to plants and has been postulated to serve as a platform for the intercellular trafficking of macromolecules. In the present study, the contribution of the plant ER membrane transport system to the intercellular trafficking of the NSm movement protein and Tomato spotted wilt tospovirus (TSWV) is investigated. We showed that TSWV NSm is physically associated with the ER membrane in Nicotiana benthamiana plants. An NSm-GFP fusion protein transiently expressed in single leaf cells was trafficked into neighboring cells. Mutations in NSm that impaired its association with the ER or caused its mis-localization to other subcellular sites inhibited cell-to-cell trafficking. Pharmacological disruption of the ER network severely inhibited NSm-GFP trafficking but not GFP diffusion. In the Arabidopsis thaliana mutant rhd3 with an impaired ER network, NSm-GFP trafficking was significantly reduced, whereas GFP diffusion was not affected. We also showed that the ER-to-Golgi secretion pathway and the cytoskeleton transport systems were not involved in the intercellular trafficking of TSWV NSm. Importantly, TSWV cell-to-cell spread was delayed in the ER-defective rhd3 mutant, and this reduced viral infection was not due to reduced replication. On the basis of robust biochemical, cellular and genetic analysis, we established that the ER membrane transport system serves as an important direct route for intercellular trafficking of NSm and TSWV.


Asunto(s)
Retículo Endoplásmico/metabolismo , Enfermedades de las Plantas/virología , Proteínas de Movimiento Viral en Plantas/metabolismo , Plasmodesmos/metabolismo , Solanum lycopersicum/virología , Tospovirus , Hojas de la Planta/metabolismo , Hojas de la Planta/virología , Plantas Modificadas Genéticamente , Transporte de Proteínas/fisiología , Nicotiana/virología
18.
J Comput Chem ; 36(11): 844-52, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25760852

RESUMEN

To probe the kinetic performance of microsolvated α-nucleophile, the G2(+)M calculations were carried out for the gas-phase S(N)2 reactions of monohydrated and dihydrated α-oxy-nucleophiles XO(-)(H2O)(n = 1,2) (X = HO, CH3O, F, Cl, Br), and α-sulfur-nucleophile, HSS(-)(H2O)(n = 1,2), toward CH3Cl. We compared the reactivities of hydrated α-nucleophiles to those of hydrated normal nucleophiles. Our calculations show that the α-effect of monohydrated and dihydrated α-oxy-nucleophiles will become weaker than those of unhydrated ones if we apply a plot of activation barrier as a function of anion basicity. Whereas the enhanced reactivity of monohydrated and dihydrated ROO(-) (R = H, Me) could be observed if compared them with the specific normal nucleophiles, RO(-) (R = H, Me). This phenomena can not be seen in the comparisons of XO(-)(H2O)(n = 1,2) (X = F, Cl, Br) with ClC2H4O(-)(H2O)(n = 1,2), a normal nucleophile with similar gas basicity to XO(-)(H2O)(n = 1,2). These results have been carefully analyzed by natural bond orbital theory and activation strain model. Meanwhile, the relationships between activation barriers with reaction energies and the ionization energies of α-nucleophile are also discussed.


Asunto(s)
Gases , Técnicas Químicas Combinatorias , Nitrocompuestos/química , Azufre , Termodinámica , Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA