Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Intervalo de año de publicación
1.
Eur J Pharm Sci ; 202: 106894, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39245357

RESUMEN

M2-like tumor-associated macrophages (M2-TAMs) are closely correlated with metastasis and poor clinical outcomes in lung squamous cell carcinoma (LUSC). Previous studies have demonstrated that STAT6 is an important signaling molecule involved in the polarization of M2-TAMs, EMT is the main way for TAMs to promote tumor progression. However, little attention has been paid to the effect of STAT6 inhibition on LUSC, and it is difficult to achieve an ideal gene silencing effect in immune cells using traditional gene transfection methods. Here, we investigated the optimal concentration of 12-myristic 13-acetate (PMA), lipopolysaccharide (LPS) for the induction of THP-1 into M1-TAMs and M2-TAMs. The expression of pSTAT6 and STAT6 was confirmed in three types of macrophages, and it was demonstrated that pSTAT6 can be used as a specific target of M2-TAMs derived from THP-1. Ultrasound-mediated nanobubble destruction (UMND) is a non-invasive and safe gene delivery technology. We also synthesized PLGA-PEI nanobubbles (NBs) to load and deliver STAT6 small interfering RNA (siRNA) into M2-TAMs via UMND. The results show that the NBs could effectively load with siRNA and had good biocompatibility. We found that UMND enhanced the transfection efficiency of siRNA, as well as the silencing effect of pSTAT6 and the inhibition of M2-TAMs. Simultaneously, when STAT6 siRNA entered M2-TAMs by UMND, proliferation, migration, invasion and EMT in LUSC cells could be inhibited via the transforming growth factor-ß1 (TGF-ß1) pathway. Therefore, our results confirm that UMND is an ideal siRNA delivery strategy, revealing its potential to inhibit M2-TAMs polarization and ultimately treat LUSC.

2.
Oncol Res ; 32(4): 679-690, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38560575

RESUMEN

Liver cancer is a prevalent malignant cancer, ranking third in terms of mortality rate. Metastasis and recurrence primarily contribute to the high mortality rate of liver cancer. Hepatocellular carcinoma (HCC) has low expression of focal adhesion kinase (FAK), which increases the risk of metastasis and recurrence. Nevertheless, the efficacy of FAK phosphorylation inhibitors is currently limited. Thus, investigating the mechanisms by which FAK affects HCC metastasis to develop targeted therapies for FAK may present a novel strategy to inhibit HCC metastasis. This study examined the correlation between FAK expression and the prognosis of HCC. Additionally, we explored the impact of FAK degradation on HCC metastasis through wound healing experiments, transwell invasion experiments, and a xenograft tumor model. The expression of proteins related to epithelial-mesenchymal transition (EMT) was measured to elucidate the underlying mechanisms. The results showed that FAK PROTAC can degrade FAK, inhibit the migration and invasion of HCC cells in vitro, and notably decrease the lung metastasis of HCC in vivo. Increased expression of E-cadherin and decreased expression of vimentin indicated that EMT was inhibited. Consequently, degradation of FAK through FAK PROTAC effectively suppressed liver cancer metastasis, holding significant clinical implications for treating liver cancer and developing innovative anti-neoplastic drugs.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Línea Celular Tumoral , Pronóstico , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Movimiento Celular , Invasividad Neoplásica/genética , Metástasis de la Neoplasia
3.
Environ Toxicol ; 39(2): 991-1000, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37994395

RESUMEN

Breast cancer is the most common cancer in the world, with metastasis being one of the leading causes of death among patients. The acidic environment of breast cancer tissue promotes tumor cell invasion and migration by inducing epithelial-mesenchymal transformation (EMT) in tumor cells, but the exact mechanisms are not yet fully understood. This study investigated the expression of acid-sensitive ion channel 1a (ASIC1a) in breast cancer tissue samples and explored the mechanisms by which ASIC1a mediates the promotion of EMT in breast cancer cells in an acidic microenvironment through in vivo and in vitro experiments. The results showed that first, the expression of ASIC1a was significantly upregulated in breast cancer tissue and was correlated with the TNM (tumor node metastasis) staging of breast cancer. Furthermore, ASIC1a expression was higher in tumors with lymph node metastasis than in those without. Second, the acidic microenvironment promoted [Ca2+ ]i influx via ASIC1a activation and regulated the expression of ß-catenin, Vimentin, and E-cadherin, thus promoting EMT in breast cancer cells. Inhibition of ASIC1a activation with PcTx-1 could suppress EMT in breast cancer cells. Finally, in vivo studies also showed that inhibition of ASIC1a could reduce breast cancer metastasis, invasion, and EMT. This study suggests that ASIC1a expression is associated with breast cancer staging and metastasis. Therefore, ASIC1a may become a new breast cancer biomarker, and the elucidation of the mechanism by which ASIC1a promotes EMT in breast cancer under acidic microenvironments provides evidence for the use of ASIC1a as a molecular target for breast cancer treatment.


Asunto(s)
Neoplasias de la Mama , beta Catenina , Humanos , Femenino , beta Catenina/metabolismo , Neoplasias de la Mama/metabolismo , Biomarcadores de Tumor , Vía de Señalización Wnt , Canales Iónicos/metabolismo , Transición Epitelial-Mesenquimal , Línea Celular Tumoral , Movimiento Celular , Microambiente Tumoral
4.
J Nanobiotechnology ; 21(1): 208, 2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37408047

RESUMEN

BACKGROUND: The immune checkpoint inhibitor (ICI) anti-PD-L1 monoclonal antibody can inhibit the progress of hepatocellular carcinoma (HCC). Epithelial-mesenchymal transformation (EMT) can promote tumor migration and the formation of immune-suppression microenvironment, which affects the therapeutic effect of ICI. Yin-yang-1 (YY1) is an important transcription factor regulating proliferation, migration and EMT of tumor cells. This work proposed a drug-development strategy that combined the regulation of YY1-mediated tumor progression with ICIs for the treatment of HCC. METHODS: We first studied the proteins that regulated YY1 expression by using pull-down, co-immunoprecipitation, and duo-link assay. The active compound regulating YY1 content was screened by virtual screening and cell-function assay. Isorhamnetin (ISO) and anti-PD-L1 antibody dual-functional mesoporous silica nanoparticles (HMSN-ISO@ProA-PD-L1 Ab) were prepared as an antitumor drug to play a synergistic anti-tumor role. RESULTS: YY1 can specifically bind with the deubiquitination enzyme USP7. USP7 can prevent YY1 from ubiquitin-dependent degradation and stabilize YY1 expression, which can promote the proliferation, migration and EMT of HCC cells. Isorhamnetin (ISO) were screened out, which can target USP7 and promote YY1 ubiquitin-dependent degradation. The cell experiments revealed that the HMSN-ISO@ProA-PD-L1 Ab nanoparticles can specifically target tumor cells and play a role in the controlled release of ISO. HMSN-ISO@ProA-PD-L1 Ab nanoparticles inhibited the growth of Hepa1-6 transplanted tumors and the effect was better than that of PD-L1 Ab treatment group and ISO treatment group. HMSN-ISO@ProA-PD-L1 Ab nanoparticles also exerted a promising effect on reducing MDSC content in the tumor microenvironment and promoting T-cell infiltration in tumors. CONCLUSIONS: The isorhamnetin and anti-PD-L1 antibody dual-functional nanoparticles can improve tumor immune microenvironment and inhibit YY1-mediated tumor progression. This study demonstrated the possibility of HCC treatment strategies based on inhibiting USP7-mediated YY1 deubiquitination combined with anti-PD-L1 monoclonal Ab.


Asunto(s)
Carcinoma Hepatocelular , Neuropatía Hereditaria Motora y Sensorial , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Microambiente Tumoral , Peptidasa Específica de Ubiquitina 7 , Ubiquitinas/farmacología , Línea Celular Tumoral , Factor de Transcripción YY1/metabolismo
5.
Int J Mol Sci ; 24(14)2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37511343

RESUMEN

NCOA7 is a nuclear receptor coactivator that is downregulated in a variety of cancers. However, the expression and prognostic significance of NCOA7 in clear cell renal cell carcinoma (ccRCC) remain unknown. The expression of NCOA7 in ccRCC tissues was analyzed using bioinformatics analysis, Western blotting, and immunohistochemistry. Kaplan-Meier analysis, the receiver operating characteristic (ROC) curve, and clinicopathological correlation analysis were used to assess the predictive power of NCOA7. Overexpression function tests were conducted in cells and mouse models to clarify the function and mechanism of NCOA7 in inhibiting the progression of ccRCC. NCOA7 expression was downregulated in all three subtypes of renal cell carcinoma, and only had significant prognostic value for patients with ccRCC. NCOA7 overexpression inhibited the proliferation, invasion, and metastasis of ccRCC cells in vivo and in vitro. Mechanistically, NCOA7 inhibited the MAPK/ERK pathway to regulate epithelial-mesenchymal transformation (EMT) and apoptosis, thereby inhibiting the progression of ccRCC. NCOA7 inhibits tumor growth and metastasis of ccRCC through the MAPK/ERK pathway, thus indicating its potential as a prognostic marker and therapeutic target for ccRCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Animales , Ratones , Carcinoma , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Sistema de Señalización de MAP Quinasas , Transducción de Señal , Humanos
6.
Transl Cancer Res ; 12(2): 236-246, 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36915593

RESUMEN

Background: Metabolic reprogramming and epithelial-mesenchymal transformation (EMT) play an important role in lung cancer. In recent studies, metabolic enzymes such as Fructose-1,6-bisphosphatase 1 (FBP1) have shown potential functions beyond regulating metabolism. Methods: Western blot assay was performed to detect glycolysis-related and EMT-related protein expression levels. The glucose uptake kit and adenosine triphosphate (ATP) detection kit were used to detect glucose uptake rate and ATP content. Transwell assay was used to determine the invasiveness of lung adenocarcinoma cells. Wound healing assay was used to determine the metastatic ability of lung adenocarcinoma cells. Methyl thiazolyl tetrazolium (MTT) assay and EdU staining were performed to investigate the effect of FBP1 overexpression on lung adenocarcinoma proliferation. Results: Overexpression of FBP1 down-regulated glycolysis-related protein levels and inhibited glucose uptake and ATP production, while knockdown of FBP1 had the opposite effect. Overexpression of FBP1 reversed EMT and inhibited Slug expression. Meanwhile, overexpression of FBP1 impaired the invasion, metastasis and proliferation ability of lung adenocarcinoma cells. In contrast, FBP1 knockdown promoted the EMT process, up-regulated Slug expression and enhanced the invasion, metastasis and proliferation of lung adenocarcinoma cells. Conclusions: Therefore, FBP1 can be used as one of the potential clinical targets through inhibiting glycolysis, cell invasion and proliferation by inhibiting Slug mediated EMT processes.

7.
Transl Cancer Res ; 11(8): 2834-2842, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36093546

RESUMEN

Background: Colon cancer is the third leading cause of tumor-related deaths in the world. Inhibition of autophagy in the treatment of malignant tumors has attracted extensive attention. However, the association between inhibition of autophagy by 3-methyladenine (3-MA) and epithelial mesenchymal transformation (EMT) in colon cancer cells has not yet been fully elucidated. Methods: In this study, colon cancer cell lines (LOVO and SW620) were treated with 3-MA. Wound healing assays and transwell assays were used to detect the effect of inhibition of autophagy on the migration and invasion of colon cancer cells. The expression of EMT-associated markers, Twist1, E-cadherin, and vimentin, in colon cancer cells with and without 3-MA treatment was detected by Western blotting, immunohistochemistry, immunofluorescence staining, and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results: Our data showed that inhibition of autophagy by 3-MA significantly enhanced the migration and invasion of colon cancer cells. At the molecular level, inhibition of autophagy upregulated the expression of Twist1 and vimentin, downregulated the expression of E-cadherin, and induced the EMT of colon cancer cells. Conclusions: Inhibition of autophagy by 3-MA upregulated the expression of Twist1 in colon cancer cells and promoted cancer cell migration and invasion through EMT. Inhibition of autophagy may have adverse effects on colon cancer.

8.
J Transl Med ; 20(1): 191, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35509067

RESUMEN

BACKGROUND: Triosephosphate isomerase 1 (TPI1), as a key glycolytic enzyme, is upregulated in multiple cancers. However, expression profile and regulatory mechanism of TPI1 in breast cancer (BRCA) remain mysterious. METHODS: Western blotting and immunohistochemistry (IHC) assays were used to investigate the expression of TPI1 in BRCA specimens and cell lines. TPI1 correlation with the clinicopathological characteristics and prognosis of 362 BRCA patients was analyzed using a tissue microarray. Overexpression and knockdown function experiments in cells and mice models were performed to elucidate the function and mechanisms of TPI1-induced BRCA progression. Related molecular mechanisms were clarified using co-IP, IF, mass spectrometric analysis, and ubiquitination assay. RESULTS: We have found TPI1 is highly expressed in BRCA tissue and cell lines, acting as an independent indicator for prognosis in BRCA patients. TPI1 promotes BRCA cell glycolysis, proliferation and metastasis in vitro and in vivo. Mechanistically, TPI1 activates phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway to regulate epithelial-mesenchymal transformation (EMT) and aerobic glycolysis, which is positively mediated by cell division cycle associated 5 (CDCA5). Moreover, TPI1 interacts with sequestosome-1 (SQSTM1)/P62, and P62 decreases the protein expression of TPI1 by promoting its ubiquitination in MDA-MB-231 cells. CONCLUSIONS: TPI1 promotes BRCA progression by stabilizing CDCA5, which then activates the PI3K/AKT/mTOR pathway. P62 promotes ubiquitin-dependent proteasome degradation of TPI1. Collectively, TPI1 promotes tumor development and progression, which may serve as a therapeutic target for BRCA.


Asunto(s)
Neoplasias de la Mama , Fosfatidilinositol 3-Quinasas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Neoplasias de la Mama/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Mamíferos/metabolismo , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
9.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-940455

RESUMEN

ObjectiveTo investigate the inhibitory effect of Astragalus polysaccharide (APS) on epithelial-mesenchymal transition (EMT) induced by transforming growth factor-β1 (TGF-β1) in cisplatin (DDP)-resistant lung adenocarcinoma cell line A549/DDP cells transplanted into nude mice and the molecular mechanism in improving DDP resistance. MethodBALB/c nude mice were randomly divided into a blank group, a model group, a DDP group, and a combination group (APS combined with DDP). A549/DDP cells were infected with TGF-β1-overexpressed lentiviral vector and the negative control. The infected cells were inoculated subcutaneously in nude mice. The A549/DDP cells with TGF-β1 gene overexpression were inoculated into all groups except the control group with negative TGF-β1 gene overexpression. The drug intervention was performed eight days after cell inoculation. The mice in the combination group received intragastric administration of APS (0.3 g·kg-1·d-1) and intraperitoneal injection of cisplatin (0.003 5 g·kg-1), and those in the cisplatin group received intraperitoneal injection of cisplatin (0.003 5 g·kg-1). After 32 days of cell inoculation, the nude mice were killed and the tumor tissues and lungs were collected. The tumor weight was recorded and the inhibition rate was calculated. The number of metastatic nodules of the lung tumor on the whole slide was counted under the microscope. Immunohistochemistry, Western blot, and real-time fluorescence-based quantitative polymerase chain reaction (Real-time PCR) were used to detect the protein and gene expression of EMT molecular markers α-catenin and N-cadherin, and tumor drug resistance markers human lung resistance protein (LRP), multidrug resistance-associated protein (MRP), and P-glycoprotein (P-gp) in the transplanted tumor. ResultCompared with the blank group, the model group showed increased tumor weight and metastatic nodules of the lung tumor (P<0.05), decreased protein and mRNA expression of α-catenin (P<0.05), and elevated protein and mRNA expression of N-cadherin, LRP, MRP, and P-gp (P<0.05). Compared with the model group and the cisplatin group, the combination group showed reduced tumor weight and metastatic nodules of the lung tumor (P<0.05), increased protein and mRNA expression of α-catenin (P<0.05), and decreased protein and mRNA expression of N-cadherin, LRP, MRP, and P-gp (P<0.05). ConclusionAPS can inhibit the growth and metastasis of the transplanted tumor of lung adenocarcinoma and improve cisplatin resistance, which may be related to the inhibition of EMT of tumor cells.

10.
Small ; 18(5): e2106498, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34921576

RESUMEN

Cell morphology and migration depend critically on the adhesions on the extracellular matrix (ECM), determined by the transmembrane protein integrins. The epithelial to mesenchymal transition (EMT) is a prominent transformation process in which adherent cells acquire a mesenchymal phenotype and a promoted migration. EMT plays important roles in embryonic development and cancer metastasis, and its hallmarks include the acquisition of front-back cell polarity and loss of cell-cell contact. However, how integrins dynamically regulate cell-ECM adhesions and cellular behaviors during EMT is still unclear. Using single-particle tracking of ß1-integrins labeled with quantum dots, the temporal-spatial on-membrane dynamics of integrins in the EMT of MCF10A cells is revealed. ß1-integrins exhibit significantly enhanced dynamics, which temporally behave more diffusive and less immobilized, and spatially become distributed asymmetrically with front regions being more dynamic. These dynamic alterations are shown to arise from microtubule remodeling in EMT. The results shed new light on the EMT mechanism from the cell-ECM adhesion perspective, and suggest that the enhanced integrin diffusion may represent as a new hallmark of EMT.


Asunto(s)
Transición Epitelial-Mesenquimal , Integrinas , Movimiento Celular , Células Epiteliales , Transición Epitelial-Mesenquimal/genética , Matriz Extracelular/metabolismo , Integrinas/metabolismo , Transducción de Señal
11.
Ann Transl Med ; 9(17): 1374, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34733926

RESUMEN

BACKGROUND: Gallbladder carcinoma (GBC) remains a highly lethal disease worldwide. MiR-552 family members promote the malignant progression of a variety of digestive system tumors, but the role of miR-552-3p in GBC has not been elucidated. miR-552-3p was predicted to target the 3'-untranslated region (3'UTR) of the mRNA for the tumor suppressor gene "repulsive guidance molecule BMP co-receptor a" (RGMA). The aim of the present study was to clarify the roles and mechanisms of miR-552-3p targeting RGMA in the malignant progression of GBC. METHODS: In vitro: expression of miR-552-3p was detected by real-time quantitative PCR (qRT-PCR) in tumor and non-tumor adjacent tissues (NATs). Lentivirus-miR-552-3p was employed to knockdown this miRNA in GBC cell lines. Stem cell-related transcription factors and markers were assessed by qRT-PCR. Cell Counting Kit-8 (CCK-8), sphere formation and transwell assays were used to determine the malignant phenotypes of GBC cells. Targeting the 3'UTR of RGMA by miR-552-3p was verified by integrated analysis including bioinformatics prediction, luciferase assays, measures of changes of gene expression and rescue experiments. In vivo: mouse models of subcutaneous tumors and lung metastases were established to observe the effect of miR-552-3p on tumorigenesis and organ metastasis, respectively. RESULTS: MiR-552-3p was abnormally highly expressed in GBC tissues and cancer stem cells. Interference with miR-552-3p in SGC-996 and GBC-SD cells significantly inhibited GBC stem cell expansion. Reciprocally, miR-552-3p promoted GBC cell proliferation, migration and invasion both in vitro and in vivo; hence, interference with this miRNA impeded the malignant progression of GBC. Furthermore, the important tumor suppressor gene RGMA was identified as a target of miR-552-3p. The effects of miR-552-3p on cell proliferation and metastasis were abrogated or enhanced by gain or loss of RGMA function, respectively. Mechanistically, miR-552-3p promoted GBC progression by reactivating the Akt/ß-catenin pathway and epithelial-mesenchymal transformation (EMT). Clinically, miR-552-3p correlated with multi-malignant characteristics of GBC and acted as a prognostic marker for GBC outcome. CONCLUSIONS: MiR-552-3p promotes the malignant progression of GBC by inhibiting the mRNA of the tumor suppressor gene RGMA, resulting in reactivation of the Akt/ß-catenin signaling pathway.

12.
Biochem Biophys Res Commun ; 581: 81-88, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34656852

RESUMEN

BACKGROUND: Pulmonary fibrosis (PF) has attracted more and more attention due to its irreversibility and high mortality rate. Currently, there is no effective treatment option is available to reverse the disease. Caspase recruitment domain-containing membrane-associated guanylate kinase protein (CARMA3) has been recognized as a proinflammatory molecule involved in many lung diseases, such as Allergic airway inflammation and lung cancer. Bleomycin (Bleo), as an alkaline sugar peptide antibiotics, is often used as a first-line anti-tumor agent. Its toxic effect is to induce pulmonary fibrosis (PF) and its clinical symptoms, so it has been widely used in the construction of pulmonary fibrosis model. METHODS: Wild type mice (WT, n = 20) and CARMA3 knockout mice (CARMA3-KO, n = 20) were generated and injected with bleomycin or saline via trachea. The severity of fibrosis was evaluated by fibrosis markers and lung histological morphology. Furthermore, the amount of alveolar epithelial cells and inflammation in lung tissue were examined. Finally, epithelial-mesenchymal transition was further investigated. RESULTS: We found CARMA3 expression in the mice alveolar epithelial cells. And compared with WT mice, CARMA3-KO mice showed reduced deposition of collagen fibers, inflammation and destruction of alveolar epithelial cells in lung tissue. In addition, after bleomycin induction, the expressions of proinflammatory factors and collagen-related factors in CARMA3-KO mice were much lower than those in WT mice. The epithelial-mesenchymal transformation phenotype was also improved in CARMA3-KO mice compared to WT mice. CONCLUSION: Our Results shows that CARMA3 plays an important role in the pathogenesis of bleomycin-induced pulmonary fibrosis. CARMA3 could alleviate the fibrosis by improving inflammation, deposition of collagen and damage of alveolar epithelial cells, which revealed that CARMA3 may be a potential target for pulmonary fibrosis.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Bleomicina/administración & dosificación , Proteínas Adaptadoras de Señalización CARD/genética , Fibronectinas/genética , Pulmón/metabolismo , Fibrosis Pulmonar/genética , Actinas/genética , Actinas/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/patología , Animales , Acuaporina 5/genética , Acuaporina 5/metabolismo , Proteínas Adaptadoras de Señalización CARD/deficiencia , Cadherinas/genética , Cadherinas/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Transición Epitelial-Mesenquimal/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Proteína C Asociada a Surfactante Pulmonar/genética , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Transducción de Señal , Vimentina/genética , Vimentina/metabolismo
13.
Tissue Cell ; 73: 101598, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34333382

RESUMEN

Colorectal cancer (CRC) is one of the most common tumors that has a high incidence worldwide. Targeted therapy for CRC has received much attention recently. It is still necessary to develop novel and promising therapeutic targets to improve the prognosis. SYNPO2, also known as synapsopoprotein 2 or myopod, encodes actin binding proteins and has been characterized as a tumor suppressor for aggressive cancers. SYNPO2 has been reported to inhibit the activity of YAP/TAZ. However, whether SYNPO2 could regulate the progression of CRC through the YAP/YAZ signaling pathway remains unclear. Herein, it was found that the expression of SYNPO2 was low in hypoxia-exposed CRC cells, consistent with the data from TCGA database. SYNPO2 inhibited the growth of CRC cells upon hypoxia treatment and promoted the cell apoptosis. Additionally, SYNPO2 inhibited the migration and epithelial-mesenchymal transformation (EMT) CRC cell upon hypoxia treatment. Mechanically, the results demonstrated that SYNPO2 suppressed hypoxia-induced progression of CRC by regulating YAP-Kruppel like factor 5 (KLF5) axis. Therefore, SYNPO2 can serve as a promising therapeutic target for CRC treatment.


Asunto(s)
Movimiento Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas de Microfilamentos/metabolismo , Transducción de Señal , Hipoxia Tumoral , Proteínas Señalizadoras YAP/metabolismo , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Hipoxia Tumoral/genética
14.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-906087

RESUMEN

Objective:To study the effect of icaritin on the proliferation, apoptosis, migration and invasion of human epithelial ovarian cancer A2780 cells and the inhibitory mechanism of icaritin against cell invasion and migration via the regulation of epithelial-mesenchymal transformation (EMT)-related molecule expression. Method:A2780 cells were divided into the blank control group and low-, medium-, and high-dose (5, 10, 20 μmol·L<sup>-1</sup>) icaritin groups and received the corresponding inventions for 48 h. Cell proliferation and viability were detected using the cell counting kit-8 (CCK-8). The cellular proliferation inhibition and apoptosis rates were assayed by flow cytometry. The cell invasion and migration were observed in Scratch test and transwell test, followed by the calculation of wound healing rate and migration rate. The protein and mRNA expression levels of EMT-related molecules including E-cadherin, N-cadherin, and Vimentin and tumor invasion and migration-related molecule matrix metalloproteinase-9 (MMP-9) were measured by Western blot and real-time polymerase chain reaction (Real-time PCR). Result:As revealed by CCK-8 assay and flow cytometry, compared with the blank control group, the icaritin groups all exhibited elevated proliferation inhibition rate (<italic>P</italic><0.01) and apoptosis rate (<italic>P</italic><0.05). According to the Scratch test and transwell test, compared with the blank control group, the icaritin groups displayed weakened invasion and migration ability and decreased number and rate of cell invasion and migration (<italic>P</italic><0.05). Western blot and Real-time PCR results showed that the protein and mRNA expression levels of N-cadherin, MMP-9 and Vimentin in each icaritin group were down-regulated as compared with those in the blank control group, while the expression of E-cadherin was up-regulated. Conclusion:Icaritin inhibits the proliferation and promotes the apoptosis of human ovarian cancer A2780 cells, and it inhibits the invasion and migration of A2780 cells possibly by regulating the expression of EMT-related molecules.

15.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-906176

RESUMEN

Objective:To explore the molecular mechanism of spleen-strengthening traditional Chinese medicine (TCM) Weichang'an granule in inhibiting the invasion and metastasis of human gastric cancer MKN45 cells. Method:MKN45 cells were cultured <italic>in vitro</italic> and incubated with different concentrations(600, 900, 1 200, 1 500 mg·L<sup>-1</sup>)of Weichang'an granule for 24, 48, 72 h. Cell counting kit-8 (CCK-8) assay was used to detect its effect on the cell proliferation. Western blot was used to detect the expression of RUN and FYVE domain containing 3(RUFY3) in normal gastric mucosa cells and different gastric cancer cell lines. The expression of RUFY3 in the gastric cancer cells after Weichang'an granule intervention (600, 900, 1 200 mg·L<sup>-1</sup>) was detected by Western blot. Lentivirus transfection technique was used to achieve the stable and silenced expression of RUFY3 in gastric cancer MKN45 cells. Transwell assay was used to evaluate the influence of Weichang'an granule and silenced RUFY3 on the metastasis and invasion ability of MKN45. E-cadherin,N-cadherin,Vimentin,Zinc-finger transcription factor (SNAIL1 and SNAIL2) protein expression levels were detected by Western blot. Result:RUFY3 expression in human gastric cancer cells was significantly higher than that in normal gastric mucosa.The protein expression of RUFY3 in MKN45 cells of silenced RUFY3 group was significantly lower than that in Lentivirus negative group (<italic>P</italic><0.01). Weichang'an granule inhibited the expression of RUFY3 in human MKN45 gastric cancer cells (<italic>P</italic><0.05, <italic>P</italic><0.01) in a concentration-dependent manner. As compared with the blank group, both Weichang'an granule and silenced RUFY3 inhibited the metastasis and invasion ability of MKN45 (<italic>P</italic><0.01). After Weichang'an granule and silenced RUFY3 treatment, the protein expression of epithelial marker gene E-cadherin was up-regulated (<italic>P</italic><0.01), the protein expression of Vimentin and N-cadherin decreased, but with no statistical difference,while SNAIL1 and SNAIL2 were both significantly down-regulated (<italic>P</italic><0.01). Conclusion:By targeting RUFY3 to regulate epithelial mesenchymal transformation, the spleen-strengthening TCM compound Weichang'an granule can inhibit the invasion and metastasis of gastric cancer.

16.
Technol Cancer Res Treat ; 19: 1533033820926592, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32462986

RESUMEN

Gastric cancer is one of the most common gastrointestinal malignancy with high mortality in East Asia. Investigation of pathogenic mechanisms of gastric cancer is crucial to develop novel therapeutic strategies and identify new therapeutic candidates. Brain-type glycogen phosphorylase is a glycogen phosphorylase involved in glycogen metabolism, which participates in multiple physiological and pathological processes. Overexpression of brain-type glycogen phosphorylase has been reported in various types of cancer, such as colorectal cancer and non-small cell lung cancer, however, the potential role of brain-type glycogen phosphorylase in gastric cancer remains unclear. Herein, we observed brain-type glycogen phosphorylase expression was significantly elevated in human gastric cancer tissues and positively correlated with the clinical-pathological features including tumor size, lymph node involvement, and tumor, node, metastasis stage of patients with gastric cancer. We further reported brain-type glycogen phosphorylase depletion suppressed the growth of gastric cancer, weakened the epithelial-mesenchymal transformation, and reduced the migration and invasion ability in cell models. We further confirmed brain-type glycogen phosphorylase depletion inhibited tumor growth and lung metastasis in mice. Importantly, we found brain-type glycogen phosphorylase regulated the progression of gastric cancer via Wnt/ß-catenin pathway, shedding lights on brain-type glycogen phosphorylase as a promising therapeutic target for drug design and development targeting gastric cancer.


Asunto(s)
Encéfalo/enzimología , Regulación Neoplásica de la Expresión Génica , Glucógeno Fosforilasa/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Gástricas/patología , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Glucógeno Fosforilasa/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Células Tumorales Cultivadas , Proteína Wnt1/genética , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
17.
Respir Physiol Neurobiol ; 276: 103402, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32006666

RESUMEN

BACKGROUND: Epithelial-mesenchymal transformation (EMT) is a central mechanism for the occurrence and development of pulmonary fibrosis. Therefore, to identify the key target molecules regulating the EMT process is considered as an important direction for the prevention and treatment of pulmonary fibrosis. Transglutaminase 2 (TG2) has been recently found to play an important role in the regulation of inflammation and the generation of extracellular matrix. Here, our study focuses on the roles of TG2 in pulmonary fibrosis and EMT. METHODS: at first, the expression of TG2 and the EMT-related markers like E-cadherin, Vimentin, and α-SMA were detected with Western Blotting, immunohistochemistry and other methods in the mice with pulmonary fibrosis induced by bleomycin. Further, MLE 12 cells were used to study the effects on EMT of the inhibition of TG2 in vitro. Finally, GK921, an inhibitor against TG2, was used to show its function in both prevention and treatment of pulmonary fibrosis induced by bleomycin in mice. RESULTS: bleomycin succeeded to induce pulmonary fibrosis in mice, with increased TG2 expression, EMT and Akt activation. Knock-down of TG2 by siRNA technique in MLE 12 cell (a mouse alveolar epithelial cell line) and GK921 (an inhibitor of TG2) all inhibited the EMT process, however SC79, an activator of Akt rescued above inhibition. Finally, GK921 alleviated pulmonary fibrosis in mice induced by bleomycin. CONCLUSION: Blocking TG2 reduces bleomycin-induced pulmonary fibrosis in mice via inhibiting EMT.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Proteínas de Unión al GTP/genética , Fibrosis Pulmonar/genética , Transglutaminasas/genética , Acetatos/farmacología , Actinas/efectos de los fármacos , Actinas/metabolismo , Animales , Antibióticos Antineoplásicos/toxicidad , Benzopiranos/farmacología , Bleomicina/toxicidad , Cadherinas/efectos de los fármacos , Cadherinas/metabolismo , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteínas de Unión al GTP/antagonistas & inhibidores , Proteínas de Unión al GTP/metabolismo , Técnicas de Silenciamiento del Gen , Técnicas In Vitro , Ratones , Proteína Glutamina Gamma Glutamiltransferasa 2 , Fibrosis Pulmonar/metabolismo , Pirazinas/farmacología , Mucosa Respiratoria/citología , Transglutaminasas/antagonistas & inhibidores , Transglutaminasas/metabolismo , Vimentina/efectos de los fármacos , Vimentina/metabolismo
18.
J Cancer ; 10(25): 6439-6456, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31772677

RESUMEN

Ethnopharmacology relevance: Jiedu Sangen Decoction (JSD), an empirical prescription of Traditional Chinese Medicine (TCM), has been reported to inhibit invasion and metastasis of colon cancer in our previous study. The aim of this study was to investigate the mechanism of JSD-triggered inhibition of invasion and metastasis in colon cancer. Methods: In vitro, AKT1 knockdown (si-AKT1) or overexpression (oe-AKT1) cells were successfully constructed both in SW480 and SW620 cell lines. Si-AKT1 and oe-AKT1 cells were then treated with or without JSD. Cell invasion, metastasis potential and expression of epithelial-mesenchymal transformation (EMT)-related and AKT1/GSK-3ß proteins were then observed by wound healing, transwell, and western blot assays. In vivo, liver metastasis model mice were developed by inoculating SW480 cells. After JSD diet intervention, living fluorescence imaging and weight measurements were carried out to investigate JSD induced inhibition effects on liver metastasis of colon cancer. Immunohistochemistry and western blot assays were performed to observe tissue features and detect protein expression. Results: Invasion and metastasis potential, as well as EMT of colon cancer, can be markedly inhibited by JSD treatment or AKT1 knockdown, while enhanced by AKT1 overexpression. JSD-induced inhibition effects were significantly weakened when AKT1 was knocked down, while clearly enhanced when AKT1 was overexpressed. Additionally, JSD could lead to an increase in expression of E-cadherin, and a decrease in expression of N-cadherin, Vimentin, p-AKT1, AKT1, p- GSK-3ß, Snail, Slug, and Twist in colon cancer cells. Conclusion: JSD reverses EMT and inhibits invasion and metastasis of colon cancer through the AKT/GSK-3ß signaling pathway.

19.
Bull Exp Biol Med ; 166(3): 377-382, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30627897

RESUMEN

The effects of laminins 332 and 411 (LM-332 and LM-411) on the epithelial-mesenchymal transformation of colorectal cancer cells (lines HT-29, HCT-116, and RKO) with different metastatic potential were studied. Culturing of RKO cells on both laminins was associated with modification of the cell shape, which became more spindle-like or stellate, and with higher expression of EMT-associated transcription factors SNAI1 and ZEB1. In addition, culturing on LM-332 led to a decrease in the expression of laminin α5 chain (LAMA5), while culturing on LM-411 led to an increase in the expression of a cell-cell junction component (DSP). Culturing of HT-29 cells on LM-332 was associated with the formation of more close contacts between the cells and by a higher expression of epithelial markers (CDH1 and DSP genes) and a decrease in SNAI1 expression. Culturing of HCT-116 cells on both laminins led to a decrease in FN1 expression, on LM-332 - to an increase in laminin α4 chain (LAMA4) expression, and on LM-411 - to a lesser expression of LAMA4 and transcription factors SNAI2 and ZEB1. These data indicated that colorectal cancer cell adhesion to laminins contributed to the probability of epithelial-mesenchymal transformation of cells. The direction of this transformation seemed to depend on the initial characteristics of the cells.


Asunto(s)
Materiales Biocompatibles Revestidos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Laminina/farmacología , Plásticos/farmacología , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Desmoplaquinas/genética , Desmoplaquinas/metabolismo , Transición Epitelial-Mesenquimal/genética , Fibronectinas/genética , Fibronectinas/metabolismo , Perfilación de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Laminina/genética , Laminina/metabolismo , Especificidad de Órganos , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Propiedades de Superficie , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
20.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-843413

RESUMEN

Objective • To establish the transforming growth factor-β2 (TGF-β2) induced epithelial-mesenchymal transition (EMT) model of retinal pigment epithelium cells, and investigate the effect and mechanism of lutein on EMT. Methods • ARPE-19 cells were cultured and divided into 4 groups including control group, TGF-β2 group, TGF-β2+lutein group and lutein group. The mRNA levels of α-smooth muscle actin (α-SMA), fibronectin (FN) and E-cadherin were analyzed by real-time PCR. The protein expression of α-SMA, FN and occludin were assayed by Western blotting. Immunofluorescence was used to detect the change of α-SMA. Meanwhile, Western blotting was performed to detect the expression levels of pSmad3 in the TGF/Smad signaling pathway. Results • TGF-β2 induced EMT was inhibited by lutein. Lutein decreased the mRNA and protein levels of the mesenchymal markers α-SMA and FN, and increased the expression of the epithelial markers E-cadherin and occludin (all P<0.05). Immunofluorescence showed that lutein can inhibit the conversion of epithelial cells into myofibroblasts. Lutein significantly downregulated the high expression of pSmad3 in TGF-β2 treated ARPE-19 cells (P=0.001). Conclusion • Lutein inhibits TGF-β2 induced EMT by downregulating the expression of pSmad3 in TGF-β/Smad signaling pathway, indicating it may attenuate subretinal fibrosis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA