Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nanotechnol Sci Appl ; 17: 167-188, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39280996

RESUMEN

Purpose: The use of nanotechnology in medicine has gained attention in developing drug delivery systems. GO has the potential to deliver microRNA (miRNA) mimics or antisense structures. MiRNAs regulate gene expression and their dysregulation is implicated in diseases, including cancer. This study aims to observe changes in morphology, viability, mRNA expression of mTOR/PI3K/Akt and PTEN genes in U87, U118, U251, A172 and T98 glioblastoma cells and xenograft models after GO self-assembly with mimic miRNA-7. Methods: Colloidal suspension of graphene oxide (GO) was used for obtaining the GO-mimic miRNA-7 nanosystems by self-assembly method. The ultrastructure, size distribution and ATR-FTIR and UV-Vis spectrum were analyzed. The Zeta potential was measured to verify the stability of obtained nanosystem. The entrapment efficiency, loading capacity and released kinetics of mimic miRNA-7 form GO-mimic miRNA-7 nanosystems were analyzed. The transfection efficiency into the glioblastoma cell lines U87, U118, U251, A172 and T98 of mimic miRNA-7 delivered by GO nanosystems was measure by confocal microscopy and flow cytometry. The changes at mRNA expression level of mTOR, PI3K, AKT1 and PTEN genes was measured by qPCR analysis. The xenograft model of U87 and A172 tumour tissue was performed to analyze the effect at tumor size and volume after GO- mimic miRNA-7 nanosystem administration. Results: The ultrastructure of GO-mimic miRNA-7 nanosystems showed high affinity of mimic miRNA into the GO. The results of transfection efficiency, cell morphology and viability showed that GO -miRNA-7 effectively deliver mimics miRNA-7 into U87, U118, U251, A172 and T98 glioblastoma cells. This approach can reverse miRNA-7 expression's downstream effects and target the mTOR PI3K/Akt pathway observed at gene expression level, reducing xenograft tumour size and volume. Conclusion: The findings of the study could have significant implications for the development of advanced and precise GO based nanosystems specifically designed for miRNA therapy in cancer treatment.

2.
Am J Cancer Res ; 11(9): 4308-4328, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659889

RESUMEN

Metastasis is the primary cause of death in lung cancer, one of the most prevalent and deadly neoplasms. The tumour-associated macrophages (TAMs) are crucial mediators to induce epithelial-mesenchymal transition (EMT) and promote lung metastasis via release of the cytokines. Matrine, a naturally occurring alkaloid, has been found with a variety of pharmacological effects, such as anti-cancer. In this study, an in vitro co-culture cell systems and a Lewis-bearing mouse model were employed to assay the potential effects of matrine on macrophages polarization, and its regulatory effects on EMT of Lewis lung cancer cells (LLCs). Our results clearly demonstrated that matrine inhibited M2-like RAW264.7 polarization, reducing the production of anti-inflammatory cytokines (IL-4, IL-10, and Arg-1), and M2 surface markers (CD206) were induced by LLCs via mTOR/PI3k/Akt signaling pathway, while it had no significant effect on M1 macrophages polarization. In vitro assays suggested that matrine partially blocked the metastasis of LLCs, and inhibited EMT induced by M2-like macrophages, which was evidenced by up-regulating the expression of E-cadherin and down-regulating the expression of N-cadherin, vimentin, and Snail. In vivo studies revealed that matrine decreased the ratio of CD206+/F4/80+, promoted the expression of CD4+ and CD8+ T cells, and inhibited the expression of Th2 in tumor and spleen tissues. Cell co-culture experiments revealed that Matrine promoted T-cell proliferation, which was impaired by tumour-derived CD11b+ myeloid cells. Collectively, our findings suggest that suppression of M2-like macrophages polarization of TAMs is a potential mechanism underlying the anti-metastasis effects of matrine in lung cancer.

3.
Expert Opin Ther Pat ; 31(11): 965-975, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34098816

RESUMEN

INTRODUCTION: The mammalian target of rapamycin (mTOR) kinase is a central component in the PI3K/Akt/mTOR pathway and plays a crucial role in tumor biology, making it one appealing therapeutic target. In the past decade, the mTORi (mTOR inhibitor) development field has made great progress, with more agents entering key trials and the proposal of third-generation mTORi concept. Yet to achieve significant clinical success, combined efforts from multiple disciplines are ever needed. AREAS COVERED: This review focuses on the progress of mTORi development with anticancer potential from the perspective of the patent literature proposed between 2011 and 2020. EXPERT OPINION: The highly complex regulatory mechanism network of mTOR proposes huge challenges to the development of clinically efficient mTORis. While in-depth biological research and fundamental medchemistry research are of importance to provide guidelines for improving mTORis, new technologies to pre-diagnose applicable populations is another key to provide precise personal cancer treatment. New mTOR agents are ever needed to tackle the common problems of side effects and drug resistance.


Asunto(s)
Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Desarrollo de Medicamentos , Resistencia a Antineoplásicos , Humanos , Neoplasias/enzimología , Patentes como Asunto , Inhibidores de Proteínas Quinasas/efectos adversos , Serina-Treonina Quinasas TOR/metabolismo
4.
Cell Rep ; 35(1): 108940, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33784499

RESUMEN

SARS-CoV-2 has currently precipitated the COVID-19 global health crisis. We developed a medium-throughput drug-screening system and identified a small-molecule library of 34 of 430 protein kinase inhibitors that were capable of inhibiting the SARS-CoV-2 cytopathic effect in human epithelial cells. These drug inhibitors are in various stages of clinical trials. We detected key proteins involved in cellular signaling pathways mTOR-PI3K-AKT, ABL-BCR/MAPK, and DNA-damage response that are critical for SARS-CoV-2 infection. A drug-protein interaction-based secondary screen confirmed compounds, such as the ATR kinase inhibitor berzosertib and torin2 with anti-SARS-CoV-2 activity. Berzosertib exhibited potent antiviral activity against SARS-CoV-2 in multiple cell types and blocked replication at the post-entry step. Berzosertib inhibited replication of SARS-CoV-1 and the Middle East respiratory syndrome coronavirus (MERS-CoV) as well. Our study highlights key promising kinase inhibitors to constrain coronavirus replication as a host-directed therapy in the treatment of COVID-19 and beyond as well as provides an important mechanism of host-pathogen interactions.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Daño del ADN , Isoxazoles/farmacología , Pirazinas/farmacología , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos , Células A549 , Animales , COVID-19/metabolismo , COVID-19/patología , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Células HEK293 , Células HeLa , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/metabolismo , Células Vero
5.
Biomed Pharmacother ; 137: 111335, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33581648

RESUMEN

Liver cancer is a critical clinical condition with augmented malignancy, rapid progression, and poor prognosis. Liver cancer often initiates as fibrosis, develops as cirrhosis, and results in cancer. For centuries, medicinal plants have been incorporated in various liver-associated complications, and recently, research has recognized that many bioactive compounds from medicinal plants may interact with targets related to liver disorders. Phyllanthin from the Phyllanthus species is one such compound extensively used by folklore practitioners for various health benefits. However, most practices continue to be unrecognized scientifically. Hence, in this work, we investigated the protective role of phyllanthin on diethylnitrosamine (DEN) induced liver carcinoma in Wistar Albino rats and the anti-tumor potential on human hepatocellular carcinoma (HCC) HepG2 cells. The DEN-challenged liver cancer in experimental rats caused increased liver weight, 8-OHD, hepatic tissue injury marker, lipid peroxidation, and tumor markers levels. Remarkably, phyllanthin counteracted the DEN effect by ameliorating all the liver function enzymes, oxidative DNA damage, and tumor-specific markers by enhanced anti-oxidant capacity and induced caspase-dependent apoptosis through the mTOR/ PI3K signaling pathway. MTT assay demonstrated that phyllanthin inhibited the HepG2 cell growth in a dose-dependent manner. Fascinatingly, phyllanthin did not demonstrate any substantial effect on the normal cell line, HL7702. In addition, HepG2 cells were found in the late apoptotic stage upon treatment with phyllanthin as depicted by acridine orange/ethidium bromide staining. Overall, this work offers scientific justification that phyllanthin can be claimed to be a safe candidate with potential chemotherapeutic activity against HCC.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Lignanos/farmacología , Neoplasias Hepáticas/prevención & control , 8-Hidroxi-2'-Desoxicoguanosina/metabolismo , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Antioxidantes/uso terapéutico , Apoptosis/genética , Biomarcadores de Tumor/metabolismo , Peso Corporal/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dietilnitrosamina/toxicidad , Modelos Animales de Enfermedad , Células Hep G2 , Humanos , Lignanos/uso terapéutico , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/genética , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Ratas Wistar , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
6.
Anat Rec (Hoboken) ; 304(3): 647-661, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32589351

RESUMEN

Methotrexate (MTX) is a widely used chemotherapeutic agent; nevertheless, the nephrotoxicity associated with its use has limited its clinical use. Rebamipide (REB) is a gastro-protective agent with diverse promising biological activities. Here, we investigated the renoprotective effects of REB against MTX-induced nephrotoxicity in rats. Male Wistar rats were allocated into four groups: the normal control group, the REB group (100 mg kg-1 day-1 , PO, for 12 days), the MTX group (which received a single injection of 20 mg/kg, ip), and the REB + MTX group (which received 100 mg kg-1 day-1 REB for 7 days before and 5 days after being injected with 20 mg/kg MTX). Interestingly, MTX triggered kidney injury, characterized by renal dysfunction along with histopathological alterations. Moreover, increased reactive oxygen species level and inflammatory response were detected in the kidney of MTX-treated rats. However, REB prevented MTX-induced oxidative kidney injury and boosted an antioxidant balance. Mechanistically, REB markedly activated the NRF-2 protein and upregulated the expression of both SIRT-1 and FOXO-3 genes. Additionally, REB administration strongly inhibited the inflammatory response by downregulating both NF-κB-p65 and TLR-4. Finally, the coadministration of REB and MTX activated the mTOR/PI3K/AKT signaling pathway. Simultaneously, REB treatment attenuated the reduction in glomerular size, the widening of the capsular spaces, and the tubular cell damage due to MTX administration. Taken together, these results indicate the potential of REB as adjuvant therapy to prevent nephrotoxicity in patients receiving MTX treatment.


Asunto(s)
Alanina/análogos & derivados , Antioxidantes/uso terapéutico , Inflamación/metabolismo , Enfermedades Renales/tratamiento farmacológico , Riñón/efectos de los fármacos , Metotrexato/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Quinolonas/farmacología , Alanina/farmacología , Alanina/uso terapéutico , Animales , Antimetabolitos Antineoplásicos/efectos adversos , Antioxidantes/farmacología , Riñón/metabolismo , Enfermedades Renales/metabolismo , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Quinolonas/uso terapéutico , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Reprod Sci ; 27(2): 681-689, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31942708

RESUMEN

The activation of dormant primordial follicles and ovarian angiogenesis has been attempted as a new treatment strategy for age-related ovarian aging. This study examined whether visfatin rescues age-related fertility decline in female mice aged 18 months, and whether this effect relates to the mTOR/PI3K signaling pathways for activation of primordial follicles and ovarian angiogenesis. Female mice were intraperitoneally injected with 0.1 ml of 500 ng/ml or 1000 ng/ml of visfatin three times at intervals of 2 days, and both ovaries were provided for H&E staining. In another experiment, the mice were superovulated with pregnant mare's serum gonadotropin and human chorionic gonadotropin, and were mated with males. After 18 h, zygotes were collected and cultured for 4 days, and numbers and embryo developmental competency of zygotes retrieved were evaluated. The expression of mTOR/PI3K signaling pathway regulated genes (4EBP1, S6K1, and RPS6) and angiogenic factors (VEGF, visfatin, and SDF-1α) in the ovary were examined. As well, visfatin-treated mice were mated with male mice for 2 weeks, and the pregnancy outcome was monitored up to 3 weeks. Visfatin significantly increased the total numbers of follicles compared with control. Numbers of zygotes retrieved, blastocyst formation rate, and pregnancy rate were significantly increased at 500 ng/ml of visfatin (2.83%, 40.0%, and 80%, respectively) compared with control (0, 0, and no pregnancy). Ovarian expressions of S6K1, RPS6, VEGF, visfatin, and SDF-1α were significantly stimulated at 500 ng/ml of visfatin. These results show that visfatin treatment of an optimal dose rescues age-related decline in fertility, possibly by stimulating mTOR/PI3K signaling.


Asunto(s)
Envejecimiento/fisiología , Citocinas/fisiología , Fertilidad/fisiología , Neovascularización Fisiológica , Nicotinamida Fosforribosiltransferasa/fisiología , Ovario/fisiología , Animales , Femenino , Ratones Endogámicos C57BL , Oocitos/fisiología , Ovario/enzimología , Embarazo
8.
Handb Exp Pharmacol ; 249: 13-30, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28233182

RESUMEN

Multiple signaling pathways regulate cell proliferation and survival and are therefore important for maintaining homeostasis of development. The balance between cell growth and death is achieved through orchestrated signal transduction pathways mediated by complex functional interactions between signaling axes, among which, PI3K/Akt and Ras/MAPK as well as JAK/STAT play a dominant role in promoting cell proliferation, differentiation, and survival. In clinical cancer therapies, drug resistance is the major challenge that occurs in almost all targeted therapeutic strategies. Recent advances in research have suggested that the intrinsic pro-survival signaling crosstalk is the driving force in acquired resistance to a targeted therapy, which may be abolished by interfering with the cross-reacting network.


Asunto(s)
Proliferación Celular , Supervivencia Celular , Neoplasias/tratamiento farmacológico , Transducción de Señal , Ciclo Celular , Resistencia a Antineoplásicos , Humanos , Quinasas Janus/fisiología , Sistema de Señalización de MAP Quinasas , Terapia Molecular Dirigida , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Factores de Transcripción STAT/fisiología , Serina-Treonina Quinasas TOR/fisiología
9.
J Nutr Biochem ; 41: 124-136, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28092744

RESUMEN

Quercetin, a bioflavonoid contained in several vegetables daily consumed, has been studied for long time for its antiinflammatory and anticancer properties. Quercetin interacts with multiple cancer-related pathways such as PI3K/AKT, Wnt/ß-catenin and STAT3. These pathways are hyperactivated in primary effusion lymphoma (PEL), an aggressive B cell lymphoma whose pathogenesis is strictly linked to the oncogenic virus Kaposis' Sarcoma-associated Herpesvirus (KSHV). In this study, we found that quercetin inhibited PI3K/AKT/mTOR and STAT3 pathways in PEL cells, and as a consequence, it down-regulated the expression of the prosurvival cellular proteins such as c-FLIP, cyclin D1 and cMyc. It also reduced the release of IL-6 and IL-10 cytokines, leading to PEL cell death. Moreover, quercetin induced a prosurvival autophagy in these cells and increased the cytotoxic effect of bortezomib, a proteasomal inhibitor, against them. Interestingly, quercetin decreased also the expression of latent and lytic KSHV proteins involved in PEL tumorigenesis and up-regulated the surface expression of HLA-DR and calreticulin, rendering the dying cells more likely detectable by the immune system. The results obtained in this study indicate that quercetin, which does not exert any cytotoxicity against normal B cells, may represent a good candidate for the treatment of this aggressive B cell lymphoma, especially in combination with autophagy inhibitors or with bortezomib.


Asunto(s)
Antineoplásicos Fitogénicos/metabolismo , Apoptosis , Autofagia , Regulación hacia Abajo , Linfoma de Efusión Primaria/metabolismo , Quercetina/metabolismo , Transducción de Señal , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/efectos adversos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Bortezomib/agonistas , Bortezomib/farmacología , Línea Celular Tumoral , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Agonismo de Drogas , Humanos , Interleucinas/antagonistas & inhibidores , Interleucinas/metabolismo , Linfoma de Efusión Primaria/tratamiento farmacológico , Linfoma de Efusión Primaria/inmunología , Linfoma de Efusión Primaria/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quercetina/efectos adversos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
10.
J Mol Cell Biol ; 7(2): 132-42, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25801958

RESUMEN

Histone deacetylase 1 (HDAC1) is an important epigenetic controller involved in transcriptional regulation through modification of chromatin structure. Genetic and epigenetic changes and deregulation of signal transduction pathways have been implicated in the development of breast cancer. Downregulation of estrogen receptor α (ERα) expression is one of the mechanisms behind the acquisition of endocrine resistance. Sustained and increased hormone and growth factor receptor signaling in breast cancer cells contribute to resistance to endocrine therapy. Both HDACs and the PI3K/mTOR signaling pathway are becoming promising targets in breast cancer, reversing also acquired hormone resistance. Here we show how mitogens, activating the PI3K/mTOR pathway, trigger the phosphorylation of HDAC1 in breast cancer cells, which is completely dependent on the activity of the p70 S6 kinase (S6K1). Our findings show that S6K1, overexpressed in many breast cancers, controls HDAC1-dependent transcriptional regulation of ERα levels upon mitogenic stimuli, controlling HDAC1 recruitment to the ERα promoter. Furthermore, cell treatment with both mTOR and HDACs inhibitors shows an additive effect in inhibiting breast cancer proliferation. This confirms the novel cross-talk between the HDAC1 and PI3K pathways with clinical implications towards the treatment of this malignant disease.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Histona Desacetilasa 1/metabolismo , Procesamiento Proteico-Postraduccional , Neoplasias de la Mama/genética , Supervivencia Celular , Receptor alfa de Estrógeno/genética , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Mitógenos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA