Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Commun Signal ; 22(1): 378, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39061097

RESUMEN

Artesunate (ART), a natural product isolated from traditional Chinese plant Artemisia annua, has not been extensively explored for its anti-melanoma properties. In our study, we found that ART inhibited melanoma cell proliferation and induced melanoma cell ferroptosis. Mechanistic study revealed that ART directly targets Ido1, thereby suppressing Hic1-mediated transcription suppression of Hmox1, resulting in melanoma cell ferroptosis. In CD8+ T cells, ART does not cause cell ferroptosis due to the low expression of Hmox1. It also targets Ido1, elevating tryptophan levels, which inhibits NFATc1-mediated PD1 transcription, consequently activating CD8+ T cells. Our study uncovered a potent and synergistic anti-melanoma efficacy arising from ART-induced melanoma cell ferroptosis and concurrently enhancing CD8+ T cell-mediated immune response both in vivo and in vitro through directly targeting Ido1. Our study provides a novel mechanistic basis for the utilization of ART as an Ido1 inhibitor and application in clinical melanoma treatment.


Asunto(s)
Artesunato , Ferroptosis , Indolamina-Pirrol 2,3,-Dioxigenasa , Melanoma , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Ferroptosis/efectos de los fármacos , Animales , Artesunato/farmacología , Artesunato/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Línea Celular Tumoral , Humanos , Ratones Endogámicos C57BL , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética
2.
Hereditas ; 159(1): 45, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36451247

RESUMEN

BACKGROUND: Coronavirus disease 2019 (COVID-19) caused a series of biological changes in cancer patients which have rendered the original treatment ineffective and increased the difficulty of clinical treatment. However, the clinical treatment for cancer patients infected with COVID-19 is currently unavailable. Since bioinformatics is an effective method to understand undiscovered biological functions, pharmacological targets, and therapeutic mechanisms. The aim of this study was to investigate the influence of COVID-19 infection in cancer patients and to search the potential treatments. METHODS: Firstly, we obtained the COVID-19-associated genes from seven databases and analyzed the cancer pathogenic genes from Gene Expression Omnibus (GEO) databases, respectively. The Cancer/COVID-19-associated genes were shown by Venn analyses. Moreover, we demonstrated the signaling pathways and biological functions of pathogenic genes in Cancer/COVID-19. RESULTS: We identified that Go-Ichi-Ni-San complex subunit 1 (GINS1) is the potential therapeutic target in Cancer/COVID-19 by GEPIA. The high expression of GINS1 was not only promoting the development of cancers but also affecting their prognosis. Furthermore, eight potential compounds of Cancer/COVID-19 were identified from CMap and molecular docking analysis. CONCLUSION: We revealed the GINS1 is a potential therapeutic target in cancer patients infected with COVID-19 for the first time, as COVID-19 will be a severe and prolonged pandemic. However, the findings have not been verified actually cancer patients infected with COVID-19, and further studies are needed to demonstrate the functions of GINS1 and the clinical treatment of the compounds.


Asunto(s)
COVID-19 , Neoplasias , Humanos , Biología Computacional , COVID-19/genética , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Pandemias , Proteínas de Unión al ADN
3.
J Cell Mol Med ; 24(10): 5695-5706, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32297701

RESUMEN

Diabetic retinopathy (DR) is one of the most prominent microvascular complications of diabetes, which remains the leading cause of legal blindness in the world. Arctiin, a bioactive compound from Arctium lappa L., has been reported to have antidiabetic activity. In this study, we investigated the effect of arctiin on a human retinal capillary endothelial cell (HRCEC) line and how arctiin inhibits cell proliferation in high glucose (HG)-induced HRCECs. Results showed that arctiin decreased HG-induced HRCECs proliferation in a dose-dependent manner by inducing cell cycle arrest at the G0/G1 phase. Tube formation assay and immunofluorescence staining indicated that arctiin abrogated tube formation induced by HG-induced HRCECs in a dose-dependent manner via down-regulation of VEGF expression. Mechanistic study indicated that perturbation of the ROCK1/PTEN/PI3K/Akt signalling pathway plays a vital role in the arctiin-mediated anti-proliferative effect. Furthermore, pre-incubation of HRCECs with Y-27632 attenuated arctiin-induced cell cycle arrest, cell proliferation and tube formation inhibition. Y-27632 also reversed the activation of PTEN, the inactivation/dephosphorylation of PI3K/Akt and down-regulation of VEGF. Taken together, the results demonstrated that arctiin inhibits the proliferation of HG-induced HRCECs through the activation of ROCK1 and PTEN and inactivation of PI3K and Akt, resulting in down-regulation of VEGF, which inhibits endothelial cell proliferation.


Asunto(s)
Células Endoteliales/metabolismo , Glucosa/metabolismo , Glucósidos/genética , Vasos Retinianos/citología , Vasos Retinianos/metabolismo , Transducción de Señal , Biomarcadores , Puntos de Control del Ciclo Celular , Proliferación Celular , Retinopatía Diabética/etiología , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Células Endoteliales/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Furanos/metabolismo , Glucosa/farmacología , Glucósidos/metabolismo , Humanos , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
4.
J Cell Mol Med ; 24(1): 618-631, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31724308

RESUMEN

Proliferation and metastasis are significantly malignant characteristics of human lung cancer, but the underlying molecular mechanisms are poorly understood. Chromobox 4 (CBX4), a member of the Polycomb group (PcG) family of epigenetic regulatory factors, enhances cellular proliferation and promotes cancer cell migration. However, the effect of CBX4 in the progression of lung cancer is not fully understood. We found that CBX4 is highly expressed in lung tumours compared with adjacent normal tissues. Overexpression of CBX4 significantly promotes cell proliferation and migration in human lung cancer cell lines. The knockdown of CBX4 obviously suppresses the cell growth and migration of human lung cancer cells in vitro. Also, the proliferation and metastasis in vivo are blocked by CBX4 knockdown. Furthermore, CBX4 knockdown effectively arrests cell cycle at the G0/G1 phase through suppressing the expression of CDK2 and Cyclin E and decreases the formation of filopodia through suppressing MMP2, MMP9 and CXCR4. Additionally, CBX4 promotes proliferation and metastasis via regulating the expression of BMI-1 which is a significant regulator of proliferation and migration in lung cancer cells. Taken together, these data suggest that CBX4 is not only a novel prognostic marker but also may be a potential therapeutic target in lung cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Ligasas/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/patología , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Femenino , Humanos , Ligasas/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Complejo Represivo Polycomb 1/genética , Proteínas del Grupo Polycomb/genética , Pronóstico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Appl Environ Microbiol ; 86(3)2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31704682

RESUMEN

Staphylococcus aureus infection poses a serious threat to public health, and antibiotic resistance has complicated the clinical treatment and limited the solutions available to solve this problem. Cold atmospheric plasma (CAP) is a promising strategy for microorganism inactivation. However, the mechanisms of microbial inactivation or resistance remain unclear. In this study, we treated S. aureus strains with a self-assembled CAP device and found that CAP can kill S. aureus in an exposure time-dependent manner. In addition, the liquid environment can influence the survival rate of S. aureus post-CAP treatment. The S. aureus cells can be completely inactivated in normal saline and phosphate-buffered saline but not in tryptic soy broth culture medium. Scanning and transmission electron microscopy revealed that the CAP-treated S. aureus cells maintained integrated morphological structures, similar to the wild-type strain. Importantly, the CAP-treated S. aureus cells exhibited a reduced pigment phenotype. Deletion of the staphyloxanthin biosynthetic genes crtM and crtN deprived the pigmentation ability of S. aureus Newman. Both the Newman-ΔcrtM and Newman-ΔcrtN mutants presented high sensitivity to CAP treatment, whereas Newman-ΔcrtO exhibited a survival rate comparable to wild-type Newman after CAP treatment. Our data demonstrated that the yellow pigment intermediates of the staphyloxanthin biosynthetic pathway are responsible for the protection of S. aureus from CAP inactivation. The key enzymes, such as CrtM and CrtN, of the golden staphyloxanthin biosynthetic pathway could be important targets for the design of novel sterilization strategies against S. aureus infections.IMPORTANCEStaphylococcus aureus is an important pathogen that can be widely distributed in the community and clinical settings. The emergence of S. aureus with multiple-antibiotic resistance has complicated staphylococcal infection control. The development of alternative strategies with powerful bactericidal effects is urgently needed. Cold atmospheric plasma (CAP) is a promising strategy for microorganism inactivation. Nevertheless, the underlying mechanisms of microbial inactivation or resistance are not completely illustrated. In this study, we validated the bactericidal effects of CAP on S. aureus, including antibiotic-resistant strains. We also found that the golden staphyloxanthin, as well as its yellow pigment intermediates, protected S. aureus against CAP, and blocking the staphyloxanthin synthesis pathway at the early steps could strengthen the sensitivity of S. aureus to CAP treatment. These data provide insights into the germicidal mechanism of CAP from the aspect of bacteria and suggest new targets against S. aureus infections.


Asunto(s)
Antibacterianos/farmacología , Viabilidad Microbiana/efectos de los fármacos , Gases em Plasma/metabolismo , Staphylococcus aureus/fisiología , Xantófilas/metabolismo , Vías Biosintéticas , Staphylococcus aureus/efectos de los fármacos , Factores de Tiempo
6.
Hereditas ; 157(1): 32, 2020 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-32703314

RESUMEN

BACKGROUND: Non-small cell lung cancer (NSCLC) is the major type of lung cancer with high morbidity and poor prognosis. Erlotinib, an inhibitor of epidermal growth factor receptor (EGFR), has been clinically applied for NSCLC treatment. Nevertheless, the erlotinib acquired resistance of NSCLC occurs inevitably in recent years. METHODS: Through analyzing two microarray datasets, erlotinib resistant NSCLC cells microarray (GSE80344) and NSCLC tissue microarray (GSE19188), the differentially expressed genes (DEGs) were screened via R language. DEGs were then functionally annotated by Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, which up-regulated more than 2-folds in both datasets were further functionally analyzed by Oncomine, GeneMANIA, R2, Coremine, and FunRich. RESULTS: We found that matrix metalloproteinase 1 (MMP1) may confer the erlotinib therapeutic resistance in NSCLC. MMP1 highly expressed in erlotinib-resistant cells and NSCLC tissues, and it associated with poor overall survival. In addition, MMP1 may be associated with COPS5 and be involve in an increasing transcription factors HOXA9 and PBX1 in erlotinib resistance. CONCLUSIONS: Generally, these results demonstrated that MMP1 may play a crucial role in erlotinib resistance in NSCLC, and MMP1 could be a prognostic biomarker for erlotinib treatment.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Biología Computacional , Resistencia a Antineoplásicos/genética , Clorhidrato de Erlotinib/farmacología , Metaloproteinasa 1 de la Matriz/genética , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Línea Celular Tumoral , Biología Computacional/métodos , Clorhidrato de Erlotinib/uso terapéutico , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Neoplasias Pulmonares/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Mutación , Pronóstico , Inhibidores de Proteínas Quinasas/uso terapéutico , Reproducibilidad de los Resultados , Transcriptoma
7.
Immunol Res ; 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38691319

RESUMEN

Recent advances in cancer immunotherapy, especially immune checkpoint inhibitors (ICIs), have revolutionized the clinical outcome of many cancer patients. Despite the fact that impressive progress has been made in recent decades, the response rate remains unsatisfactory, and many patients do not benefit from ICIs. Herein, we summarized advanced studies and the latest insights on immune inhibitory factors in the tumor microenvironment. Our in-depth discussion and updated landscape of tumor immunosuppressive microenvironment may provide new strategies for reversing tumor immune evasion, enhancing the efficacy of ICIs therapy, and ultimately achieving a better clinical outcome.

8.
Neoplasia ; 48: 100966, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38237304

RESUMEN

Although immune checkpoint therapy has significantly improved the prognosis of patients with melanoma, urgent attention still needs to be paid to the low patient response rates and the challenges of precisely identifying patients before treatment. Therefore, it is crucial to investigate novel immunosuppressive mechanisms and targets in the tumor microenvironment in order to reverse tumor immune escape. In this study, we found that the cell cycle checkpoint Aurora kinase B (AURKB) suppressed the anti-tumor immune response, and its inhibitor, Tozasertib, effectively activated T lymphocyte cytokine release in vitro and anti-tumor immunity in vivo. Tozasertib significantly inhibited melanoma xenograft tumor growth by decreasing the number of inhibitory CD4+ Treg cells in the tumors, which, in turn, activated CD8+ T cells. Single-cell analysis revealed that AURKB suppressed anti-tumor immunity by increasing MIF-CD74/CXCR4 signaling between tumor cells and lymphocytes. Our study suggests that AURKB is a newly identified anti-tumor immunity suppressor, whose inhibitors may be developed as novel anti-tumor immunity drugs and may have synergistic anti-melanoma effects with immune checkpoint therapies.


Asunto(s)
Antineoplásicos , Melanoma , Humanos , Melanoma/patología , Linfocitos T Reguladores , Linfocitos T CD8-positivos , Antineoplásicos/uso terapéutico , Microambiente Tumoral
9.
FEBS J ; 290(24): 5720-5743, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37581853

RESUMEN

Src homolog and collagen homolog binding protein 1 (SHCBP1) binds to the SH2 domain of SHC-transforming protein 1 (SHC1) and is involved in midbody organization and cytokinesis completion. SHCBP1 has been reported to be a cancer driver gene, promoting cancer progression. However, the functional role and underlying mechanism of SHCBP1 in regulating lung adenocarcinoma (LUAD) cell proliferation and migration are incompletely understood. Here, we discovered that SHCBP1 is overexpressed in LUAD tissues and is associated with a poor prognosis. SHCBP1 knockdown inhibited LUAD cell proliferation and migration by arresting the cell cycle and preventing epithelial-mesenchymal transition (EMT) via decreasing cyclin-dependent kinase 1 (CDK1) expression. Mechanistically, CDK1 overexpression reversed SHCBP1 knockdown-induced inhibition of proliferation and migration, confirming CDK1 as a key downstream target of SHCBP1. In addition, we proposed that rucaparib may be a small-molecule inhibitor of SHCBP1 and validated both in vitro and in vivo that rucaparib inhibits cell proliferation and migration via suppression of the SHCBP1/CDK1 pathway in LUAD. Our study elucidates a newly identified role of SHCBP1 in promoting cell proliferation and migration in LUAD, and suggests rucaparib as a potential inhibitor for LUAD treatment.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Proteína Quinasa CDC2/genética , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Proliferación Celular , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Movimiento Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proteínas Adaptadoras de la Señalización Shc
10.
Microb Biotechnol ; 15(6): 1910-1921, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35290715

RESUMEN

Pseudomonas aeruginosa is an important opportunistic human pathogen, which raises a worldwide concern for its increasing resistance. Nonthermal plasma, which is also called cold atmospheric plasma (CAP), is an alternative therapeutic approach for clinical infectious diseases. However, the bacterial factors that affect CAP treatment remain unclear. The sterilization effect of a portable CAP device on different P. aeruginosa strains was investigated in this study. Results revealed that CAP can directly or indirectly kill P. aeruginosa in a time-dependent manner. Scanning electron microscopy and transmission electron microscope showed negligible surface changes between CAP-treated and untreated P. aeruginosa cells. However, cell leakage occurred during the CAP process with increased bacterial lactate dehydrogenase release. More importantly, pigmentation of the P. aeruginosa culture was remarkably reduced after CAP treatment. Further mechanical exploration was performed by utilizing mutants with loss of functional genes involved in pyocyanin biosynthesis, including P. aeruginosa PAO1 strain-derived phzA1::Tn, phzA2::Tn, ΔphzA1/ΔphzA2, phzM::Tn and phzS::Tn, as well as corresponding gene deletion mutants based on clinical PA1 isolate. The results indicated that pyocyanin and its intermediate 5-methyl phenazine-1-carboxylic acid (5-Me-PCA) play important roles in P. aeruginosa resistance to CAP treatment. The unique enzymes, such as PhzM in the pyocyanin biosynthetic pathway, could be novel targets for the therapeutic strategy design to control the growing P. aeruginosa infections.


Asunto(s)
Pseudomonas aeruginosa , Piocianina , Proteínas Bacterianas/genética , Vías Biosintéticas , Humanos , Pseudomonas aeruginosa/genética , Piocianina/metabolismo
11.
Carbohydr Polym ; 294: 119805, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-35868765

RESUMEN

Uncontrolled hemorrhage poses a severe life-threatening situation. However, traditional hemostats still have various limitations. It is urgent to develop a material with excellent biocompatibility and hemostatic ability. Evidence has shown that carboxymethyl chitosan (CMCS) has hemostatic properties and good compatibility. Herein, we develop an expandable hemostatic sponge by modifying CMCS with cellulose nanofibrils (CNFs) through the CO-NH cross-linking method. We verified its potential as a hemostatic agent both in vivo and in vitro. The results demonstrated that the prepared carboxymethyl chitosan/cellulose nanofiber composite (CNF-CMCS) sponges could absorb blood, quickly expand to exert pressure in the wound, and exhibit an excellent coagulation ability. The CNF-CMCS sponges significantly decreased the bleeding time and blood loss in several hemorrhage models and possessed a significant advantage in treating the deep penetrating injury hemorrhage. Therefore, the sponges provide a unique application prospect and potential as a penetrating trauma hemostatic agent.


Asunto(s)
Quitosano , Hemostáticos , Nanofibras , Carboximetilcelulosa de Sodio/farmacología , Celulosa/farmacología , Quitosano/farmacología , Quitosano/uso terapéutico , Hemorragia/tratamiento farmacológico , Hemostasis , Hemostáticos/farmacología , Hemostáticos/uso terapéutico , Humanos
12.
J Adv Res ; 31: 165-175, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34194840

RESUMEN

Background: The treatment of patients with Staphylococcus aureus infections mainly relies on antistaphylococcal regimens that are established with effective antibiotics. In antibiotic therapy or while living in nature, pathogens often face the sub-inhibitory concentrations (sub-MICs) of antibiotics due to drug pharmacokinetics, diffusion barriers, waste emission, resistant organism formation, and farming application. Different categories of antibiotics at sub-MICs have diverse effects on the physiological and chemical properties of microorganisms. These effects can result in virulence alterations. However, the mechanisms underlying the actions of antibiotics at sub-MICs on S. aureus virulence are obscure. Aim of review: In this review, we focus on the effects of sub-MICs of antibiotics on S. aureus virulence from the aspects of cell morphological change, virulence factor expression, bacterial adherence and invasion, staphylococcal biofilm formation, and small-colony variant (SCV) production. The possible mechanisms of antibiotic-induced S. aureus virulence alterations are also addressed. Key scientific concepts of review: Five main aspects of bacterial virulence can be changed in S. aureus exposure to the sub-MIC levels of antibiotics, resulting in deformed bacterial cells to stimulate abnormal host immune responses, abnormally expressed virulence factors to alter disease development, changed bacterial adhesion and invasion abilities to affect colonization and diffusion, altered biofilm formation to potentate material-related infections, and increased SCV formation to achieve persistent infection and recurrence. These advanced findings expand our knowledge to rethink the molecular signaling roles of antibiotics beyond their actions as antimicrobial agents.


Asunto(s)
Antibacterianos/administración & dosificación , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/patogenicidad , Adhesión Bacteriana , Biopelículas , Humanos , Pruebas de Sensibilidad Microbiana/métodos , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Virulencia , Factores de Virulencia
13.
Int J Oncol ; 55(4): 833-844, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31485605

RESUMEN

Rho­associated protein kinase 1 (ROCK1), a member of the ROCK family, serves an important function in cell migration and invasion in neoplasms. ROCK1 has been found to be overexpressed in several types of cancers. However, the role of ROCK1 in non­small­cell lung cancer (NSCLC) is poorly understood. In the present study, ROCK1 was found to be overexpressed in NSCLC cells and tissues, and it was associated with poor survival of NSCLC patients. Subsequently, ROCK1 knockdown NSCLC cell lines were established using shRNA. ROCK1 knockdown significantly reduced the migration and invasion ability in the cell monolayer scratching and Transwell assays. ROCK1 knockdown was also found to markedly inhibit cell adhesion ability. Moreover, the phosphorylation of focal adhesion kinase (FAK) was inhibited by ROCK1 knockdown, reducing NSCLC cell migration and invasion ability. This mechanistic study revealed that ROCK1 significantly enhanced cell migration and invasion by inhibiting the phosphatase and tensin homolog (PTEN)/phosphoinositide 3­kinase (PI3K)/FAK pathway. More importantly, the interruption of the PTEN/PI3K/FAK pathway markedly rescued the inhibition of cell migration and invasion mediated by ROCK1 knockdown. Taken together, these results suggest a novel role for ROCK1 in cell migration and invasion by inhibiting cell adhesion ability, and indicate that ROCK1 may be of value as a therapeutic target for the treatment of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Transducción de Señal , Regulación hacia Arriba , Quinasas Asociadas a rho/genética , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Análisis de Supervivencia , Análisis de Matrices Tisulares
14.
Int J Oncol ; 55(2): 415-424, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31173174

RESUMEN

IR­783, a near­infrared heptamethine cyanine dye, has been reported to possess cancer targeting and anticancer effects; Ηowever, the molecular mechanism by which IR­783 exhibits anti­breast cancer activity is unclear. In the present study, the inhibitory effects of IR­783 on the proliferation and migration of breast cancer cells were investigated. Our results revealed that IR­783 inhibited MDA­MB­231 and MCF­7 cell proliferation in a dose­ and time­dependent manner by inducing cell cycle arrest at the G0/G1 phase. In addition, a Transwell assay demonstrated that IR­783 treatment suppressed the migratory ability of MDA­MB­231 and MCF­7 cells. Furthermore, IR­783 treatment decreased the expression levels of matrix metalloproteinase (MMP)­2 and MMP­9 in MDA­MB­231 cells. Furthermore, IR­783 induced MDA­MB­231 and MCF­7 cell mitochondrial fission, and also decreased the levels of ATP. This was accompanied with a decrease in polymerized filamentous actin, which is the fundamental component of filopodia at the cell surface. Collectively, the results of the present study demonstrated that IR­783 inhibited the proliferation and migration of MDA­MB­231 and MCF­7 cells by inducing mitochondrial fission and subsequently decreasing ATP levels, resulting in cell cycle arrest and filopodia formation suppression. These findings suggest that IR­783 may be developed into an effective novel drug for treating breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colorantes Fluorescentes/farmacología , Indoles/farmacología , Dinámicas Mitocondriales/efectos de los fármacos , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Indoles/uso terapéutico , Rayos Infrarrojos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA