Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Molecules ; 29(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38893528

RESUMEN

The existing kinase inhibitors for hepatocellular carcinoma (HCC) have conferred survival benefits but are hampered by adverse effects and drug resistance, necessitating the development of novel agents targeting distinct pathways. To discover potent new anti-HCC compounds, we leveraged scaffold hopping from Sorafenib and introduced morpholine/piperidine moieties to develop ureido-substituted 4-phenylthiazole analogs with optimized physicochemical properties and binding interactions. Notably, compound 27 exhibited potent cytotoxicity against HepG2 cells (IC50 = 0.62 ± 0.34 µM), significantly exceeding Sorafenib (IC50 = 1.62 ± 0.27 µM). Mechanistic investigations revealed that compound 27 potently inhibited HCC cell migration and colony formation, and it induced G2/M arrest and early-stage apoptosis. Kinase profiling revealed IGF1R as a key target, which compound 27 potently inhibited (76.84% at 10 µM). Molecular modeling substantiated compound 27's strong binding to IGF1R via multiple hydrogen bonds. Computational predictions indicate favorable drug-like properties for compound 27. These findings provide a promising drug candidate for the treatment of HCC patients.


Asunto(s)
Antineoplásicos , Apoptosis , Proliferación Celular , Inhibidores de Proteínas Quinasas , Receptor IGF Tipo 1 , Tiazoles , Humanos , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Proliferación Celular/efectos de los fármacos , Células Hep G2 , Tiazoles/química , Tiazoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Movimiento Celular/efectos de los fármacos , Relación Estructura-Actividad , Simulación del Acoplamiento Molecular , Receptores de Somatomedina/antagonistas & inhibidores , Receptores de Somatomedina/metabolismo , Estructura Molecular , Línea Celular Tumoral , Sorafenib/farmacología , Sorafenib/química , Modelos Moleculares
2.
BMC Cancer ; 15: 389, 2015 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-25958313

RESUMEN

BACKGROUND: Emerging data have suggested that cell surface GRP78 is a multifunctional receptor and has been linked to proliferative and antiapoptotic signaling cascades. Activated α2-macroglobin (α2M*) is a natural circulating ligand of cell surface GRP78. Association of cell surface GRP78 with α2M* is involved in the regulation of cell proliferation, survival and apoptosis in human cancers. METHODS: The invasion and metastasis of HCC cells were examined using transwell and wound healing assay; Cell surface expression of GRP78 was detected by in cell western assay. Translocation of GRP78 from cytosol to cell surface was observed by transfection of GRP78-EGFP plus TRIRC-WGA staining. The levels of Src, phosphor-Src, FAK, phospho-FAK, EGFR, phospho-EGFR, phospho-Cortactin, phospho-Paxillin were determined by western blot. Cell surface expression of GRP78 in HCC tissue samples was observed by immunofluorescence. The distribution of Paxillin and Cortactin in HCC cells was also observed by immunofluorescence. The interaction between GRP78 and Src were detected by far-western blot, co-immunoprecipitation and GST pulldown. GRP78 mRNA was detected by RT-PCR. RESULTS: In the current study, we showed that association of cell surface GRP78 with α2M* stimulated the invasion and metastasis of HCC. Cell surface GRP78 could interact directly with c-Src, promoted the phosphorylation of c-Src at Y416. Inhibition of the tyrosine kinase activity of c-Src with PP2 reverted the stimulatory effect caused by association of cell surface GRP78 with α2M*. Moreover, association of cell surface GRP78 with α2M* facilitates the interaction between EGFR and c-Src and consequently phosphorylated EGFR at Y1101 and Y845, promoting the invasion and metastasis of HCCs. However, inhibition of the tyrosine kinase of c-Src do not affect the interaction between EGFR and Src. CONCLUSION: c-Src plays a critical role in the invasion and metastasis of HCC induced by association of cell surface GRP78 with α2M*. Cell surface GRP78 directly binds and phosphorylates c-Src. As a consequence, c-Src phosphorylated EGFR, promoting the invasion and metastasis of HCCs.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas de Choque Térmico/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , alfa-Macroglobulinas/metabolismo , Familia-src Quinasas/metabolismo , Proteína Tirosina Quinasa CSK , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Membrana Celular/metabolismo , Chaperón BiP del Retículo Endoplásmico , Receptores ErbB/metabolismo , Expresión Génica , Proteínas de Choque Térmico/genética , Humanos , Neoplasias Hepáticas/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Unión Proteica , Transporte de Proteínas , Transducción de Señal
3.
J Nanobiotechnology ; 12: 43, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25358257

RESUMEN

BACKGROUND: Active targeting endocytosis mediated by the specific interaction between folic acid and its receptor has been a hotspot in biological therapy of many human cancers. Various studies have demonstrated that folate and its conjugates could facilitate the chemotherapeutic drug delivery into folate receptor (FR)-positive tumor cells in vitro and in vivo. In order to utilize FA-FR binding specificity to achieve targeted delivery of drugs into tumor cells, we prepared Gefitinib loaded folate decorated bovine serum albumin conjugated carboxymethyl-ß-cyclodextrin nanoparticles for enhancing drug delivery in cancer cells. On this context, the aim of our study was to develop a novel nano-delivery system for promoting tumor-targeting drug delivery in folate receptor-positive Hela cells. RESULTS: We prepared folic acid (FA)-decorated bovine serum albumin (BSA) conjugated carboxymethyl-ß-cyclodextrin (CM-ß-CD) nanoparticles (FA-BSA-CM-ß-CD NPs) capable of entrapping a hydrophobic Gefitinib. It was observed that nanoparticles are monodisperse and spherical nanospheres with an average diameter of 90.2 nm and negative surface charge of -18.6 mV. FA-BSA-CM-ß-CD NPs could greatly facilitate Gefitinib uptake and enhance the toxicity to folate receptor-positive Hela cells. Under the reaction between FA and FR, Gefitinib loaded FA-BSA-CM-ß-CD NPs induced apoptosis of Hela cells through elevating the expression of caspase-3 and inhibited autophagy through decreasing the expressing of LC3. It also confirmed that clathrin-mediated endocytosis and macropinocytosis exerted great influence on the internalization of both NPs. CONCLUSIONS: These results demonstrated that FA may be an effective targeting molecule and FA-BSA-CM-ß-CD NPs provided a new strategy for the treatment of human cancer cells which over-expressed folate receptors.


Asunto(s)
Autofagia/efectos de los fármacos , Ácido Fólico/química , Nanopartículas/administración & dosificación , Nanopartículas/química , Quinazolinas/administración & dosificación , Adenosina Trifosfato/metabolismo , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Receptores de Folato Anclados a GPI/metabolismo , Gefitinib , Células HeLa/efectos de los fármacos , Células HeLa/metabolismo , Humanos , Terapia Molecular Dirigida , Albúmina Sérica Bovina/química , Espectroscopía Infrarroja por Transformada de Fourier , beta-Ciclodextrinas/química
4.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(3): 304-312, 2024 Jun 01.
Artículo en Inglés, Zh | MEDLINE | ID: mdl-39049649

RESUMEN

OBJECTIVES: This study aims to investigate the influence of glucose regulated protein (GRP) 78 on osteoblast differentiation in periodontal ligament fibroblasts (PDLFs) under cyclic mechanical stretch and determine the underlying mechanism. METHODS: FlexCell 5000 cell mechanical device was applied to simulate the stress environment of orthodontic teeth. GRP78High and GRP78Low subpopulation were obtained by flow sorting. Gene transfection was performed to knockdown GRP78 and c-Src expression and overexpress c-Src. Western blot analysis was used to detect the protein expression of Runt-related gene 2 (RUNX2), Osterix, osteocalcin (OCN), and osteopontin (OPN). Immunoprecipitation assay was used to determine the interaction of GRP78 with c-Src. The formation of cellular mineralized nodules was determined by alizarin red staining. RESULTS: GRP78 was heterogeneously expressed in PDLFs, and GRP78High and GRP78Low subpopulations were obtained by flow sorting. The osteogenic differentiation ability and phosphorylation level of c-Src kinase in the GRP78High subpopulation were significantly increased compared with those in GRP78Low subpopulation after cyclic mechanical stretch (P<0.05). GRP78 interacted with c-Src in PDLFs. The overexpression c-Src group showed significantly increased osteogenic differentiation ability than the vector group (P<0.05), and the sic-Src group showed significantly decreased osteogenic differentiation ability (P<0.05) after cyclic mechanical stretch. CONCLUSIONS: GRP78 upregulates c-Src expression by interacting with c-Src kinase and promotes osteogenic differentiation under cyclic mechanical stretch in PDLFs.


Asunto(s)
Diferenciación Celular , Proteínas de Choque Térmico , Osteoblastos , Ligamento Periodontal , Proteínas Proto-Oncogénicas pp60(c-src) , Transducción de Señal , Estrés Mecánico , Humanos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteína Tirosina Quinasa CSK/metabolismo , Chaperón BiP del Retículo Endoplásmico/metabolismo , Fibroblastos/metabolismo , Proteínas de Choque Térmico/metabolismo , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteogénesis , Osteopontina/metabolismo , Ligamento Periodontal/metabolismo , Ligamento Periodontal/citología , Fosforilación , Familia-src Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo
5.
Sci Rep ; 14(1): 18107, 2024 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-39103425

RESUMEN

The limitations associated with conventional cancer treatment modalities, particularly for breast cancer, underscore the imperative for developing safer and more productive drug delivery systems. A promising strategy that has emerged is the combination of chemotherapy with gas therapy. We synthesized curcumin-loaded amorphous calcium carbonate nanoparticles (Cur-CaCO3) via a gas diffusion reaction in the present study. Subsequently, a "one-step" ethanol injection method was employed to fabricate lipid-coated calcium carbonate nanoparticles (Cur-CaCO3@LA-Lip) loaded with L-arginine, aimed at harnessing the synergistic effects of chemotherapy and nitric oxide to enhance antitumor efficacy. Transmission electron microscopy analysis revealed that Cur-CaCO3@LA-Lip nanoparticles were subspherical with a distinct lipid layer encapsulating the periphery. Fourier transform infrared spectroscopy, X-ray powder diffraction, and differential scanning calorimetry results confirmed the successful synthesis of Cur-CaCO3@LA-Lip. The nanoparticles exhibited significant drug loading capacities of 8.89% for curcumin and 3.1% for L-arginine. In vitro and in vivo assessments demonstrated that Cur-CaCO3@LA-Lip nanoparticles facilitated sustained release of curcumin and exhibited high cellular uptake, substantial tumor accumulation, and excellent biocompatibility. Additionally, the nanoparticles showed robust cytotoxicity and potent antitumor efficacy, suggesting their potential as a formidable candidate for breast cancer therapy.


Asunto(s)
Neoplasias de la Mama , Curcumina , Nanopartículas , Óxido Nítrico , Curcumina/farmacología , Curcumina/administración & dosificación , Curcumina/química , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Nanopartículas/química , Animales , Humanos , Óxido Nítrico/administración & dosificación , Óxido Nítrico/metabolismo , Óxido Nítrico/química , Ratones , Lípidos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Carbonato de Calcio/química , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Portadores de Fármacos/química , Arginina/química
6.
Int J Biol Sci ; 18(2): 491-506, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35002505

RESUMEN

Oral squamous cell carcinoma (OSCC) refers to the malignant tumor of the head and neck with a highest morbidity. It exhibits a poor prognosis and unsatisfactory treatment partially attributed to delayed diagnosis. As indicated from existing reports, the protein histidine phosphatase LHPP acts as a vital factor in tumorigenesis in liver, lung, bladder, breast and pancreatic tumor tissues. Thus far, the functional mechanism of LHPP in OSCC remains unclear. DGE analysis, OSCC cell lines and OSCC cases were found that LHPP was down-regulated in OSCC tissues and cells compared with that in normal oral mucosa tissues and cells, and was closely related to OSCC differentiation. Cell counting Kit 8 test, EdU proliferation test, scratches test, invasion test, monoclonal formation test, mouse xenograft tumor model, HE staining and immunohistochemistry showed that LHPP inhibited OSCC growth, proliferation and migration in vivo and in vitro. GO and KEGG enrichment analysis, LHPP transcription factor analysis and flow cytometry found that LHPP promotes the apoptosis of OSCC by decreasing the transcriptional activity of p-PI3K and p-Akt. Finally, our results suggested that LHPP inhibited the progression of OSCC through the PI3K/AKT signaling pathway, indicating that LHPP may be a new target for the treatment of OSCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Pirofosfatasa Inorgánica/biosíntesis , Neoplasias de la Boca/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Femenino , Genes Supresores de Tumor , Humanos , Pirofosfatasa Inorgánica/genética , Pirofosfatasa Inorgánica/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
ACS Omega ; 7(44): 39760-39771, 2022 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-36385800

RESUMEN

Oral cancer is one of the most common malignancies of the head and neck, and approximately 90% of oral cancers are oral squamous cell carcinomas (OSCCs). The purinergic P2Y2 receptor is upregulated in breast cancer, pancreatic cancer, colorectal cancer, and liver cancer, but its role in OSCC is still unclear. Here, we examined the effects of P2Y2 on the invasion and migration of oral cancer cells (SCC15 and CAL27). The BALB/c mouse model was used to observe the involvement of P2Y2 with tumors in vivo. P2Y2, Src, and EGFR are highly expressed in OSCC tissues and cell lines. Stimulation with ATP significantly enhanced cell invasion and migration in oral cancer cells, and enhanced the activity of Src and EGFR protein kinases, which is mediated by the PI3K/AKT signaling pathway. P2Y2 knockdown attenuated the above ATP-driven events in vitro and in vivo. The PI3K/AKT signaling pathway was blocked by Src or EGFR inhibitor. Extracellular ATP activates the PI3K/AKT pathway through the P2Y2-Src-EGFR axis to promote OSCC invasion and migration, and thus, P2Y2 may be a potential novel target for antimetastasis therapy.

8.
Int J Biochem Cell Biol ; 153: 106316, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36280040

RESUMEN

Notum, which belongs to the α/ß hydrolase family, is a deacylated extracellular protein that regulates the Wnt signaling pathway. Studies have found that Notum participates in the progression of colorectal cancer and hepatocellular carcinoma, but its role in oral squamous cell carcinoma (OSCC) is currently unclear. This study aimed to explore the role of Notum in regulating OSCC and further reveal the underlying mechanisms. Various approaches including bioinformatics analysis, enzyme-linked immunosorbent assay and immunohistochemical staining were used to detect the expression of Notum in OSCC cells and tissues. Cell counting kit-8 assay, clone formation assay, wound healing assay, transwell assay and in-gel zymography assay were explored to evaluate the regulation of Notum in OSCC proliferation and migration. Hoechst 33342/PI assay, cell immunofluorescence, flow cytometry and in vivo tumorigenesis experiment were applied for OSCC apoptosis. Real-time quantitative polymerase chain reaction analysis was performed for mRNA level while western blotting was conducted to detect protein expression. The results showed that Notum was highly expressed in OSCC tissues and cells, and Notum promoted the proliferation and migration of OSCC cells while it inhibited their apoptosis. Furthermore, signaling pathway analysis showed that Notum led to potential pro-survival of OSCC through crosstalk between sonic hedgehog (Shh) and Wnt/ß-catenin signaling via phosphorylation of glycogen synthase kinase-3 beta. These results will help to elucidate the mechanism and also provide new ideas for targeted treatment of OSCC.


Asunto(s)
Esterasas , Neoplasias de la Boca , Carcinoma de Células Escamosas de Cabeza y Cuello , Humanos , beta Catenina/genética , beta Catenina/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Esterasas/genética , Esterasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Vía de Señalización Wnt/genética
9.
Shanghai Kou Qiang Yi Xue ; 30(6): 573-578, 2021 Dec.
Artículo en Zh | MEDLINE | ID: mdl-35587009

RESUMEN

PURPOSE: To investigate the effect of Met kinase inhibitor BMS-777607 on proliferation and apoptosis of tongue squamous cell carcinoma cell line CAL27. METHODS: The effect of BMS-777607 on proliferation of CAL27 was detected by MTT method, clone formation assay and EdU cell imaging. Morphological changes of apoptosis of CAL27 cells induced by BMS-777607 were observed by Heochst33342 staining. JC-1 staining was used to detect the changes of mitochondrial membrane potential of CAL27 cells treated with BMS-777607. Western blot was used to detect the effect of BMS-777607 on the expression of proliferation protein Akt, p-Akt and apoptosis-related proteins Bcl-2, Cleaved caspase-3, Bax and Parp in CAL27 cells. The data were analyzed using SPSS 22.0 software package. RESULTS: BMS-777607 inhibited proliferation and promoted apoptosis of CAL27 cells in a concentration-dependent manner(P<0.05). It also inhibited the expression of Bcl-2 and p-Akt and promoted the expression of Bax, Cleaved caspase-3 and Parp protein (P<0.05). CONCLUSIONS: BMS-777607 can inhibit proliferation and promote apoptosis of CAL27 cells.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de la Lengua , Aminopiridinas , Apoptosis , Proteínas Reguladoras de la Apoptosis , Carcinoma de Células Escamosas/metabolismo , Caspasa 3/farmacología , Caspasa 3/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Humanos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/uso terapéutico , Piridonas , Lengua , Neoplasias de la Lengua/tratamiento farmacológico , Proteína X Asociada a bcl-2/farmacología
10.
Front Pharmacol ; 12: 615889, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33716742

RESUMEN

The multireceptor tyrosine kinase inhibitor sorafenib is a Food and Drug Administration-approved first-line drug for the treatment of advanced liver cancer that can reportedly extend overall survival in patients with advanced hepatocellular carcinoma (HCC). Primary and acquired resistance to sorafenib are gradually increasing however, leading to failure of HCC treatment with sorafenib. It is therefore crucial to study the potential mechanism of sorafenib resistance. The results of the current study indicate that neurite outgrowth inhibitor protein B receptor (NgBR) is overexpressed in cultured sorafenib-resistant cells, and that its expression is negatively correlated with the sensitivity of liver cancer cells to sorafenib. Artesunate can inhibit the expression of NgBR, and it may block sorafenib resistance. Herein we report that sorafenib treatment in combination with artesunate overcomes HCC resistance to sorafenib alone in a cell culture model.

11.
Aging (Albany NY) ; 13(3): 4242-4257, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33495407

RESUMEN

Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumors worldwide, and its prognosis is still not optimistic. Oxaliplatin is a type of platinum chemotherapeutic agent, but its treatment effects on OSCC and molecular mechanisms have not been fully elucidated. Parthanatos, a unique form of cell death, plays an important role in a variety of physiological and pathological processes. This study aims to investigate whether oxaliplatin inhibits OSCC by inducing parthanatos. Our results showed that oxaliplatin inhibited the proliferation and migration of OSCC cells in vitro, and also inhibited the tumorigenesis in vivo. Further experiments proved that oxaliplatin induced parthanatos in OSCC cells, characterized by depolarization of the mitochondrial membrane potential, up-regulation of PARP1, AIF and MIF in the nucleus, as well as the nuclear translocation of AIF. Meanwhile, PARP1 inhibitor rucaparib and siRNA against PARP1 attenuated oxaliplatin-induced parthanatos in OSCC cells. In addition, we found that oxaliplatin caused oxidative stress in OSCC cells, and antioxidant NAC not only relieved oxaliplatin-induced overproduction of reactive oxygen species (ROS) but also reversed parthanatos caused by oxaliplatin. In conclusion, our results indicate that oxaliplatin inhibits OSCC by activating PARP1-mediated parthanatos through increasing the production of ROS.


Asunto(s)
Antineoplásicos/farmacología , Mitocondrias/efectos de los fármacos , Neoplasias de la Boca/metabolismo , Oxaliplatino/farmacología , Parthanatos/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Línea Celular Tumoral , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Neoplasias de la Boca/patología , Trasplante de Neoplasias , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
BMC Cancer ; 10: 20, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20082722

RESUMEN

BACKGROUND: Glucose regulated protein 78 (Grp78) is involved in the invasion and metastasis in many human cancers including gastric cancer, breast cancer, prostate cancer. But the role of Grp78 in the invasion of human hepatocellular carcinoma has not been reported. In this article, we examined if Grp78 was associated with the invasion of hepatocellular carcinoma and explored the possible underlying mechanism. METHODS: The Grp78 and FAK expression levels in 44 patients with hepatocellular carcinoma were examined using immunohistochemistry. Grp78 overexpressing SMMC7721 cells were established by pcDNA3.1 (+)-Grp78 transfection and screened by G418. Grp78 and FAK levels in Grp78 overexpressing cells were down-regulated by siRNA transfection. The invasion status of tumor cells was evaluated by transwell assay in vitro, and chick embryo metastasis model in vivo. Cell spreading was determined by cell spreading assay, and quantitatively measured by Orisis software HUG. Grp78, pY397 FAK, pY576/577 FAK and FAK levels were detected by western blot. RhoA activity was detected by GST pulldown assay. The distribution of actin cytoskeleton was observed by fluorescent staining. RESULTS: Grp78 expression levels in 44 patients with hepatocellular carcinoma were negatively correlated with tumor grading, and positively correlated with portal invasion and intra-hepatic invasion. Overexpression of Grp78 in SMMC7721 cells promoted the invasion of cancer cells in vitro and in vivo, and this increase in tumor cell invasion was blocked by Grp78 siRNA knockdown. Our results also revealed that overexpression of Grp78 in SMMC7721 cells accelerated the process of cell spreading and promoted lamellipodia formation. Further analysis showed that overexpression of Grp78 in SMMC7721 cells increased pY397 and pY576/577 levels of FAK. Grp78 siRNA knockdown decreased FAK activation and activity. Our results also revealed that Grp78 overexpression in SMMC7721 cells decreased RhoA-GTP level, and Grp78 siRNA knockdown rescued RhoA-GTP level in Grp78 overexpressing cells, indicating Grp78 inhibited RhoA activity in hepatocellular carcinoma cells. Furthermore, overexpression of Grp78 in SMMC7721 cells increased phospho-p190RhoGAP level. FAK siRNA knockdown in Grp78 overexpressing cells reversed phospho-p190RhoGAP level. These data suggested that Grp78 inhibited RhoA activity by up-regulated phospho-p190RhoGAP level and Grp78 mediated p190RhoGAP phosphorylation is FAK dependent. CONCLUSION: Grp78 promoted the invasion of hepatocellular carcinoma both in vitro and in vivo. Overexpression of Grp78 in hepatocellular carcinoma cells enhanced the activation and activity of FAK which negatively regulated Rock kinase activity by promoting the phosphorylation of p190RhoGAP.


Asunto(s)
Carcinoma Hepatocelular/patología , Proteínas de Choque Térmico/metabolismo , Neoplasias Hepáticas/patología , Actinas/química , Adulto , Anciano , Animales , Línea Celular Tumoral , Embrión de Pollo , Citoesqueleto/metabolismo , Chaperón BiP del Retículo Endoplásmico , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo
13.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 38(5): 495-501, 2020 Oct 01.
Artículo en Zh | MEDLINE | ID: mdl-33085231

RESUMEN

OBJECTIVE: This study aims to study the effect of the enhancer of zeste homolog 2 (EZH2) inhibitor GSK126 on the proliferation and apoptosis of human tongue squamous cell carcinoma cells in vitro and explore its related mechanisms in order to obtain insights into the clinical treatment of tongue squamous cell carcinoma. METHODS: Different concentrations of GSK126 were applied to CAL-27 cells of tongue squamous cell carcinoma, and the effects of drugs on cell proliferation were detected through methyl thiazolyl tetrazolium (MTT) assay, colony formation assay, and 5-ethynyl-2'-deoxyuridine (EdU) fluorescence staining. Hoechst33342 fluorescence staining and the JC-1 method were used in observing apoptosis. The expression levels of extracellular regulated protein kinases (ERK), phospho-extracellular regulated protein kinases (p-ERK), Bax, Bcl-2, and Cleaved caspase-9 in Cal-27 cells were detected through Western blot. RESULTS: GSK126 inhibited CAL-27 cell proliferation and promoted apoptosis. GSK126 down-regulated the expression of p-ERK and Bcl-2 and increased the expression of Bax and Cleaved caspase-9 (P<0.05). CONCLUSIONS: GSK126 can inhibit the proliferation of CAL-27 cells in tongue squamous cell carcinoma and promote its apoptosis, and the related mechanism may be associated with the inhibition of the MEK/ERK signaling pathway and activation of the Bax/Bcl-2 pathway.


Asunto(s)
Apoptosis , Carcinoma de Células Escamosas , Neoplasias de la Lengua , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Indoles , Piridonas , Neoplasias de la Lengua/tratamiento farmacológico
14.
Clin Exp Pharmacol Physiol ; 35(1): 97-102, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18047635

RESUMEN

1. The aims of the present study were to determine whether delta-opioid receptor stimulation enhanced proliferation of and to investigate the role of the extracellular signal-regulated kinase (ERK) pathway in ventricular myocytes from neonatal rats. 2. At concentratins ranging from 10 nmol/L to 10 micromol/L, [D-Ala2,D-Leu5]enkephalin (DADLE) concentration-dependently promoted myocardial growth and DNA synthesis and altered the cytoskeleton. 3. At 1 micromol/L, DADLE also increased the expression and phosphorylation of ERK. 4. These effects of 1 micromol/L DADLE were abolished by 10 micromol/L naltrindole, a selective delta-opioid receptor antagonist, 10 nmol/L U0126, a selective ERK antagonist, 1 micromol/L staurosporine, an inhibitor of protein kinase (PK) C, and 100 micromol/L Rp-adenosine 3',5'-cyclic monophosphorothioate triethylammonium salt hydrate (Rp-cAMPS), an inhibitor of PKA. 5. In conclusion, delta-opioid receptor stimulation enhances the proliferation and development of the ventricular myocytes of neonatal rats. The ERK pathway and related signalling mechanisms, namely PKC and PKA, are involved.


Asunto(s)
Analgésicos Opioides/farmacología , Proliferación Celular/efectos de los fármacos , Leucina Encefalina-2-Alanina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Receptores Opioides delta/agonistas , Transducción de Señal/efectos de los fármacos , Animales , Animales Recién Nacidos , Butadienos/farmacología , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citoesqueleto/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Nitrilos/farmacología , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/metabolismo , Estaurosporina/farmacología , Tionucleótidos/farmacología , Factores de Tiempo
15.
Oncotarget ; 8(5): 7502-7520, 2017 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-27447746

RESUMEN

Here, BTG1 overexpression inhibited proliferation, induced differentiation, autophagy, and apoptosis in colorectal cancer cells (p<0.05). BTG1 overexpression reduced mitochondrial membrane potential and caused senescence in HCT-116 transfectants (p<0.05). BTG1-induced G2 arrest might be related to Cyclin B1 and Cdc25B hypoexpression in HCT-15 transfectants, while G1 arrest in HCT-116 transfectants overexpressing p21 and p27. BTG1 overexpression decreased the expression of Bcl-2, Bcl-xL, XIAP, Akt1 or survivin and increased the expression of Bax or p53 in colorectal cancer cells. BTG1-induced autophagy was dependent on Beclin-1 expression. BTG1 overexpression might weaken ß-catenin pathway in colorectal cancer cells. The chemosensitivity of BTG1 transfectants to paclitaxel, cisplatin, MG132 or SAHA was positively correlated with its apoptotic induction. There was a lower expression level of BTG1 in cancer than matched non-neoplastic mucosa by RT-PCR (p<0.05), while versa for Western blot and immunohistochemical data (p<0.05). BTG1 overexpression significantly suppressed the growth of HCT-15 and HCT-116 via inhibiting proliferation, inducing apoptosis and autophagy in nude mice. Up-regulated BTG1 expression plays an important role in colorectal carcinogenesis as a potential biomarker. BTG1 expression might reverse aggressive phenotypes, so it might be employed as a target of gene therapy for colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Terapia Genética/métodos , Proteínas de Neoplasias/genética , Animales , Antineoplásicos/farmacología , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , beta Catenina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Curr Pharm Biotechnol ; 17(13): 1117-1125, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27587021

RESUMEN

Physalis pubescens L. (P. pubescens) is an edible plant used in folk medicine in China. There is traditional, but not scientific, evidence for the anti-tumour effects of P. pubescens. This study aimed to identify whether, or not, antioxidants rich in phenols and flavonoids from fruits and calyxes of P. pubescens can be the candidates for further development of an anti-hepatoma fraction, and if such biological effects coupled with reactive oxygen species (ROS) changes, can provide a direction for subsequent biological action. The effects of calyx-origin (or fruit-origin) total phenol and flavonoid (CTPF or FTPF) from P. pubescens on Malhavu cell viability were evaluated by using a counting-kit-8 (CCK-8) method. Morphological characterisation of cells was undertaken and the structures were photographed (200 × magnification) using Hoechst 3348 staining after exposure to different concentrations of CTPF or FTPF. Induced-apoptosis activity was determined using flow cytometry (FC) after Annexin VFITC/ PI staining. The corresponding ROS changes in Malhavu cells were observed and quantified by the uploading of 2', 7'-dichlorofluorescin diacetate (DCFH-DA). Anti-oxidation was evaluated by a cellular oxidation-stress model and chemical assessments for DPPH, hydroxyl radial, super-oxide radicals, and reducing power. Result shows that CTPF led to significant anti-proliferation in a time- and dosedependent manner. However, FTPF promoted cell viability at 100-1000 µg/mL with a dose-response manner in 24 h. With the extension of exposure time to 48 h, the cell viability did not increase with the growth of FTPF. Morphological characterisation and FC assay both demonstrated that CTPF, and not FTPF possessed induced-apoptotic activity. CTPF potentially induced cell apoptosis by promoting oxidative stress. FTPF indicated pro-oxidation at a concentration of 10 µg/mL and anti-oxidation capabilities at higher concentrations. ROS scavenging assay by oxidation-stress model indicated that CTPF (10 - 400 µg/mL) had ROS inhibitory capacity (R2 = 0.5156, p < 0.0001). FTPF (10 - 100 µg/mL) boosted the level of ROS (p < 0.0001) and inhibited the generation of ROS at 100-400 µg/mL (R2 = 0.5951, p < 0.0001). CTPF is a potential candidate requiring further exploration for the development of antihepatoma ingredients. The down-regulation of cell viability was related to production and reduction of cellular ROS.


Asunto(s)
Antioxidantes/farmacología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Physalis/química , Extractos Vegetales/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Flavonoides/farmacología , Humanos , Neoplasias Hepáticas/patología , Estrés Oxidativo/efectos de los fármacos , Fenoles/farmacología , Especies Reactivas de Oxígeno/metabolismo
17.
Oncotarget ; 7(34): 54596-54615, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27409347

RESUMEN

ING5 can interact with p53, thereby inhibiting cell growth and inducing apoptosis. We found that ING5 overexpression not only inhibited proliferation, migration, and invasion, but also induced G2 arrest, differentiation, autophagy, apoptosis, glycolysis and mitochondrial respiration in lung cancer cells. ING5 transfection up-regulated the expression of Cdc2, ATG13, ATG14, Beclin-1, LC-3B, AIF, cytochrome c, Akt1/2/3, ADFP, PFK-1 and PDPc, while down-regulated the expression of Bcl-2, XIAP, survivin,ß-catenin and HXK1. ING5 transfection desensitized cells to the chemotherapy of MG132, paclitaxel, and SAHA, which paralleled with apoptotic alteration. ING5 overexpression suppressed the xenograft tumor growth by inhibiting proliferation and inducing apoptosis. ING5 expression level was significantly higher in normal tissue than that in lung cancer at both protein and mRNA levels. Nuclear ING5 expression was positively correlated with ki-67 expression and cytoplasmic ING5 expression. Cytoplasmic ING5 expression was positively associated with lymph node metastasis, and negatively with age, lymphatic invasion or CPP32 expression. ING5 expression was different in histological classification: squamous cell carcinoma > adenocarcinoma > large cell carcinoma > small cell carcinoma. Taken together, our data suggested that ING5 downregulation might involved in carcinogenesis, growth, and invasion of lung cancer and could be considered as a promising marker to gauge the aggressiveness of lung cancer. It might be employed as a potential target for gene therapy of lung cancer.


Asunto(s)
Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Células A549 , Anciano , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Femenino , Humanos , Ácidos Hidroxámicos/farmacología , Estimación de Kaplan-Meier , Leupeptinas/farmacología , Neoplasias Pulmonares/clasificación , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Paclitaxel/farmacología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Oncol Lett ; 10(2): 653-660, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26622549

RESUMEN

microRNA-34a (miRNA/miR-34a) functions as a tumor suppressor gene in gastric cancer and may be involved in system-wide regulatory networks. To clarify the expression of all predicted target genes of this miRNA, a comprehensive and systematic analysis of miR-34a-target genes in gastric cancer was conducted in the present study. In the initial analysis, the potential functions, pathways and networks of gastric cancer-associated molecules and miR-34a targets were identified. In the final integrative analysis of gastric cancer-associated miR-34a targets, 30 hub genes were identified using overlap calculations, indicating that miR-34a may be significant in the development and progression of gastric cancer through the Smad signaling pathway, the cell cycle, the mitogen-activated protein kinase signaling pathway, apoptosis, the Notch signaling pathway and other pathways. The present study provides a bioinformatic analysis of miR-34a-targets in gastric cancer, describes numerous target genes and novel coregulatory networks, and may provide an opportunity to identify a critical regulatory network for predicting the molecular mechanisms of miR-34a in the development and progression of gastric cancer.

19.
Int J Nanomedicine ; 10: 245-56, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25565817

RESUMEN

Nanoparticles (NPs) which target specific agents could effectively recognize the target cells and increase the stability of chemical agents by encapsulation. As such, NPs have been widely used in cancer treatment research. Recently, over 90% of treatment failure cases in patients with metastatic cancer were attributed to resistance to chemotherapy. Surface-exposed glucose-regulated protein of 78 kDa (GRP78) is expressed highly on many tumor cell surfaces in many human cancers and is related to the regulation of invasion and metastasis. Herein, we report that NPs conjugated with antibody against GRP78 (mAb GRP78-NPs) inhibit the adhesion, invasion, and metastasis of hepatocellular carcinoma (HCC) and promote drug delivery of 5-fluorouracil into GRP78 high-expressed human hepatocellular carcinoma cells. Our new findings suggest that mAb GRP78-NPs could enhance drug accumulation by effectively transporting NPs into cell surface GRP78-overexpressed human hepatocellular carcinoma cells and then inhibit cell proliferation and viability and induce cell apoptosis by regulating caspase-3. In brief, mAb GRP78-NPs effectively inhibit cancer cell invasion and enhance antitumor efficiency by targeted drug delivery.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Proteínas de Choque Térmico/metabolismo , Nanopartículas/química , Invasividad Neoplásica/prevención & control , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Fluorouracilo/química , Fluorouracilo/farmacología , Proteínas de Choque Térmico/genética , Humanos , Neoplasias Hepáticas/tratamiento farmacológico
20.
Oncotarget ; 6(32): 33658-74, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26378040

RESUMEN

5-FU is a common first-line chemotherapeutic drug for the treatment of hepatocellular carcinoma. However the development of acquired resistance to 5-FU confines its clinical usages. Although this phenomenon has been the subject of intense investigation, the exact mechanism of acquired resistance to 5-FU remains elusive. Here, we report that over-expression of GRP78 contributes to acquired resistance to 5-FU in HCC by up-regulating the c-Src/LSF/TS axis. Moreover, we found that the resistance to 5-FU conferred by GRP78 is mediated by its ATPase domain. The ATPase domain differentially increased the expression of LSF, TS and promoted the phosphorylation of ERK and Akt. We further identified that GRP78 interacts physically with c-Src through its ATPase domain and promotes the phosphorylation of c-Src, which in turn increases the expression of LSF in the nucleus. Together, GRP78 confers the resistance to 5-FU by up-regulating the c-Src/LSF/TS axis via its ATPase domain.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Fluorouracilo/farmacología , Proteínas de Choque Térmico/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Timidilato Sintasa/metabolismo , Factores de Transcripción/metabolismo , Familia-src Quinasas/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Proteína Tirosina Quinasa CSK , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Resistencia a Antineoplásicos , Chaperón BiP del Retículo Endoplásmico , Femenino , Proteínas de Choque Térmico/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Transducción de Señal , Timidilato Sintasa/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA