Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Zhongguo Zhong Yao Za Zhi ; 49(1): 185-196, 2024 Jan.
Article in Chinese | MEDLINE | ID: mdl-38403351

ABSTRACT

This study investigated the effect of trametenolic acid(TA) on the migration and invasion of human hepatocellular carcinoma HepG2.2.15 cells by using Ras homolog gene family member C(RhoC) as the target and probed into the mechanism, aiming to provide a basis for the utilization of TA. The methyl thiazolyl tetrazolium(MTT) assay was employed to examine the proliferation of HepG2.2.15 cells exposed to TA, and scratch and Transwell assays to examine the cell migration and invasion. The pull down assay was employed to determine the impact of TA on RhoC GTPase activity. Western blot was employed to measure the effect of TA on the transport of RhoC from cytoplasm to cell membrane and the expression of RhoC/Rho-associated kinase 1(ROCK1)/myosin light chain(MLC)/matrix metalloprotease 2(MMP2)/MMP9 pathway-related proteins. RhoC was over-expressed by transient transfection of pcDNA3.1-RhoC. The changes of F-actin in the cytoskeleton were detected by Laser confocal microscopy. In addition, the changes of cell migration and invasion, expression of proteins in the RhoC/ROCK1/MLC/MMP2/MMP9 pathway, and RhoC GTPase activity were detected. The subcutaneously transplanted tumor model of BALB/c nude mice and the low-, medium-, and high-dose(40, 80, and 120 mg·kg~(-1), respectively) TA groups were established and sorafenib(20 mg·kg~(-1)) was used as the positive control. The tumor volume and weight in each group were measured, and the expression of related proteins in the tumor tissue was determined by Western blot. The results showed that TA inhibited the proliferation of HepG2.2.15 cells in a concentration-dependent manner, with the IC_(50) of 66.65 and 23.09 µmol·L~(-1) at the time points of 24 and 48 h, respectively. The drug administration groups had small tumors with low mass. The tumor inhibition rates of sorafenib and low-, medium-and high-dose TA were 62.23%, 26.48%, 55.45%, and 62.36%, respectively. TA reduced migrating and invading cells and inhibited RhoC protein expression and RhoC GTPase activity in a concentration-dependent manner, dramatically reducing RhoC and membrane-bound RhoC GTPase. The expression of ROCK1, MLC, p-MLC, MMP2, and MMP9 downstream of RhoC can be significantly inhibited by TA, as confirmed in both in vitro and in vivo experiments. After HepG2.2.15 cells were transfected with pcDNA3.1-RhoC to overexpress RhoC, TA down-regulated the protein levels of RhoC, ROCK1, MLC, p-MLC, MMP2, and MMP9 and decreased the activity of RhoC GTPase, with the inhibition level comparable to that before overexpression. In summary, TA can inhibit the migration and invasion of HepG2.2.15 cells. It can inhibit the RhoC/ROCK1/MLC/MMP2/MMP9 signaling pathway by suppressing RhoC GTPase activity and down-regulating RhoC expression. This study provides a new idea for the development of autophagy modulators targeting HSP90α to block the proliferation and inhibit the invasion and migration of hepatocellular carcinoma cells via multiple targets of active components in traditional Chinese medicines.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Humans , rhoC GTP-Binding Protein/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Matrix Metalloproteinase 9/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , Sorafenib , Mice, Nude , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Cell Line, Tumor , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Cell Movement , Cell Proliferation
2.
Histol Histopathol ; 39(4): 497-509, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37702425

ABSTRACT

BACKGROUND: The interaction of microRNA with Chinese herbal medicines is a promising therapeutic approach for prevention of cervical cancer. METHODS: Western blotting or qRT-PCR were carried out to identify the expression of NCAPG2 and miR-638. A tetrandrine (TET) cell model was used to explore the effects of miR-638 and its target gene NCAPG2 using CCK-8, transwell, wound healing, and western blot assays. Furthermore, luciferase activity assay was conducted to measure the interaction among TET, NCAPG2 and miR-638. RESULTS: Under TET treatment, Hela and SiHa cells exhibited repressed cell viability, migration, invasion, and epithelial-mesenchymal transition (EMT), and these effects were further enhanced by high expression of miR-638. In contrast, NCAPG2 expression was low in TET-treated cells and had an opposite effect to that of miR-638. CONCLUSION: We highlighted that miR-638 suppresses cervical cancer progression by inhibiting NCAPG2 under tetrandrine treatment.


Subject(s)
Benzylisoquinolines , MicroRNAs , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Cell Line, Tumor , Neoplasm Invasiveness/genetics , Cell Proliferation , Cell Movement/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Chromosomal Proteins, Non-Histone/metabolism
3.
J Biochem Mol Toxicol ; 36(11): e23182, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35938691

ABSTRACT

Aberrant expression of microRNA-497 (miR-497) is associated with tumor progression, but the molecular mechanisms in tumorigenesis remain largely unknown. Here, we report that miR-497 expression is downregulated in esophageal squamous cell carcinoma (ESCC) clinical samples. Consistently, upregulation of miR-497 inhibits ESCC cell malignant properties and tumor growth in vivo. Importantly, we uncovered that miR-497 upregulation suppressed ESCC cell growth and tumor growth by inhibiting Smurf2. Mechanistically, we showed that Smurf2 was a target of miR-497, and mediated YY1 expression to elevate HIF2α expression, thereby enhancing the malignancy of ESCC cells. Together, our study uncovered the role of the miR-497-mediated Smurf2/YY1/HIF2α axis in tumor growth and metastasis, which might provide potential therapeutic targets for human ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , MicroRNAs , Humans , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , MicroRNAs/genetics , Cell Line, Tumor , Up-Regulation , Cell Proliferation , Gene Expression Regulation, Neoplastic , Cell Movement , Neoplasm Invasiveness/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
4.
Dev Cell ; 57(1): 32-46.e8, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35016005

ABSTRACT

We test the hypothesis that glioblastoma harbors quiescent cancer stem cells that evade anti-proliferative therapies. Functional characterization of spontaneous glioblastomas from genetically engineered mice reveals essential quiescent stem-like cells that can be directly isolated from tumors. A derived quiescent cancer-stem-cell-specific gene expression signature is enriched in pre-formed patient GBM xenograft single-cell clusters that lack proliferative gene expression. A refined human 118-gene signature is preserved in quiescent single-cell populations from primary and recurrent human glioblastomas. The F3 cell-surface receptor mRNA, expressed in the conserved signature, identifies quiescent tumor cells by antibody immunohistochemistry. F3-antibody-sorted glioblastoma cells exhibit stem cell gene expression, enhance self-renewal in culture, drive tumor initiation and serial transplantation, and reconstitute tumor heterogeneity. Upon chemotherapy, the spared cancer stem cell pool becomes activated and accelerates transition to proliferation. These results help explain conventional treatment failure and lay a conceptual framework for alternative therapies.


Subject(s)
Cell Survival/physiology , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/genetics , Cell Division/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/pathology , Heterografts , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Transcriptome/genetics
5.
Gene ; 808: 145994, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34626722

ABSTRACT

The involvement of store-operated calcium channels (SOCCs) in tumor initiation and metastatic dissemination has been extensively studied, but how its member ORAI3 influences tumor progression is still elusive. The present study aimed to evaluate the prognostic value of ORAI3 expression and examine the correlation between ORAI3 expression and immune cell infiltration within the tumor microenvironment (TME) in human muscle-invasive bladder cancer (MIBC). We examined the expression profile of ORAI3 in MIBC using data from two databases; analyzed the correlation between ORAI3 expression and patient survival; explored cellular pathways related to ORAI3 expression by Gene Set Enrichment Analysis (GSEA); and predicted potential drugs using Connectivity Map (CMap). ORAI3 was significantly lower expressed in tumor mass compared to normal samples in MIBC, with a higher level of methylation at the promoter region in tumor than in normal tissue, indicating that ORAI3 is suppressed during cancer progression. Survival analysis showed that higher expression of ORAI3 correlated with good prognosis in MIBC. GSEA demonstrated that ORAI3 expression inversely correlated with cell differentiation, development and gene silencing, with differential expression of genes involved in epidermal and keratinocyte differentiation pathways and inflammatory responses. RNA sequencing of an ORAI3-silenced human bladder cancer cell line (T24 cells) corroborated enhancement of pro-neoplastic pathways in absence of ORAI3. Western blottingMoreover, ORAI3 facilitated the recruitment of Th17 cells and natural killer cells, whereas hampered Th2 and macrophage infiltration. Our results revealed 4 molecules with potential to be beneficial as adjuvant drugs in MIBC treatment. We concluded that high ORAI3 expression correlates with increased survival in human MIBC.


Subject(s)
Calcium Channels/genetics , Gene Expression Profiling/methods , Urinary Bladder Neoplasms/genetics , Biomarkers, Tumor/genetics , Calcium Channels/metabolism , China , Databases, Genetic , Disease Progression , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Predisposition to Disease/genetics , Humans , Neoplasm Invasiveness/genetics , Prognosis , Survival Analysis , Transcriptome/genetics , Tumor Microenvironment/immunology , Urinary Bladder/pathology
6.
Int J Mol Sci ; 22(22)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34830011

ABSTRACT

The Chinese medicine monomer cynaroside (Cy) is a flavonoid glycoside compound that widely exists in plants and has a variety of pharmacological effects, such as its important role in the respiratory system, cardiovascular system and central nervous system. Studies have reported that Cy has varying degrees of anticancer activity in non-small cell lung cancer, cervical cancer, liver cancer, esophageal cancer and other cancers. However, there are no relevant reports about its role in gastric cancer. The MET/AKT/mTOR signaling pathway plays important roles in regulating various biological processes, including cell proliferation, apoptosis, autophagy, invasion and tumorigenesis. In this study, we confirmed that Cy can inhibit the cell growth, migration and invasion and tumorigenesis in gastric cancer. Our finding shows that Cy can block the MET/AKT/mTOR axis by decreasing the phosphorylation level of AKT, mTOR and P70S6K. Therefore, the MET/AKT/mTOR axis may be an important target for Cy. In summary, Cy has anti-cancer properties and is expected to be a potential drug for the treatment of gastric cancer.


Subject(s)
Glucosides/pharmacology , Luteolin/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-met/genetics , Stomach Neoplasms/drug therapy , TOR Serine-Threonine Kinases/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medicine, Chinese Traditional , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
7.
J Pharmacol Sci ; 146(2): 71-81, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33941323

ABSTRACT

BACKGROUND: Dihydroartemisinin (DHA) possesses an inhibitory effect on ovarian cancer and promotes reversion-inducing cysteine-rich protein with Kazal motifs (RECK) expression in glioma cells. This study explored the role of DHA and RECK on ovarian cancer. METHODS: The RECK level in ovarian cancer was analyzed under GEPIA 2 database and proved by RT-qPCR. After being treated with DHA or infected with siRECK lentivirus, the viability, apoptosis, migration, and invasion of ovarian cancer cells were evaluated by CCK-8, flow cytometry, wound healing, and transwell assays. Also, the expressions of factors related to apoptosis and epithelial-mesenchymal transition were measured by Western blot or RT-qPCR. RESULTS: DHA-treatment weakened the viability, migration, invasion, and enhanced apoptosis of ovarian cancer cells. DHA also down-regulated the levels of Bcl-2, N-cadherin, and Vimentin, and up-regulated the levels of Bax, C-caspase-3 and E-cadherin in ovarian cancer cells. RECK was lowly expressed in both ovarian cancer tissues and cells. siRECK not only had an effect opposite to DHA on the viability, apoptosis, migration, invasion, and related-factors of ovarian cancer cells but also offset the effect of DHA on ovarian cancer cells. CONCLUSION: DHA regulated apoptosis, migration, and invasion of ovarian cancer cells via mediating RECK.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Artemisinins/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Neoplasm Invasiveness/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Artemisinins/therapeutic use , Female , Humans , Ovarian Neoplasms/drug therapy , Phytotherapy , Tumor Cells, Cultured , Up-Regulation/drug effects
8.
Int J Mol Sci ; 22(8)2021 Apr 12.
Article in English | MEDLINE | ID: mdl-33921242

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies. Present-day treatments have not shown real improvements in reducing the high mortality rate and the short survival of the disease. The average survival is less than 5% after 5 years. New innovative treatments are necessary to curtail the situation. The very dense pancreatic cancer stroma is a barrier that impedes the access of chemotherapeutic drugs and at the same time establishes a pro-proliferative symbiosis with the tumor, thus targeting the stroma has been suggested by many authors. No ideal drug or drug combination for this targeting has been found as yet. With this goal in mind, here we have explored a different complementary treatment based on abundant previous publications on repurposed drugs. The cell surface protein CD44 is the main receptor for hyaluronan binding. Many malignant tumors show over-expression/over-activity of both. This is particularly significant in pancreatic cancer. The independent inhibition of hyaluronan-producing cells, hyaluronan synthesis, and/or CD44 expression, has been found to decrease the tumor cell's proliferation, motility, invasion, and metastatic abilities. Targeting the hyaluronan-CD44 pathway seems to have been bypassed by conventional mainstream oncological practice. There are existing drugs that decrease the activity/expression of hyaluronan and CD44: 4-methylumbelliferone and bromelain respectively. Some drugs inhibit hyaluronan-producing cells such as pirfenidone. The association of these three drugs has never been tested either in the laboratory or in the clinical setting. We present a hypothesis, sustained by hard experimental evidence, suggesting that the simultaneous use of these nontoxic drugs can achieve synergistic or added effects in reducing invasion and metastatic potential, in PDAC. A non-toxic, low-cost scheme for inhibiting this pathway may offer an additional weapon for treating pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Hyaluronan Receptors/genetics , Hyaluronan Synthases/genetics , Hyaluronic Acid/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Bromelains/therapeutic use , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronan Synthases/antagonists & inhibitors , Hyaluronic Acid/antagonists & inhibitors , Hymecromone/therapeutic use , Molecular Targeted Therapy , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Pyridones/pharmacology , Pyridones/therapeutic use , Signal Transduction/drug effects
9.
Cancer Lett ; 500: 119-131, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33338532

ABSTRACT

Nav1.5, encoded by SCN5A, has been associated with metastasis in colorectal cancer (CRC). Here, we investigated the mechanism by which Nav1.5 regulates tumor progression and whether Nav1.5 influences chemosensitivity to 5-fluorouracil (5-FU) in CRCs. CRC cases were evaluated for Nav1.5 expression. Elevated Nav1.5 expression was associated with poor prognosis in CRCs, whereas stage II/III patients with upregulated SCN5A expression could have better survival after receiving 5-FU-based adjuvant chemotherapy. In CRC cells, SCN5A knockdown reduced the proliferation, migration and invasion. According to RNA sequencing, SCN5A knockdown inhibited both the cell cycle and epithelial-mesenchymal transition. In addition, Nav1.5 stabilized the KRas-calmodulin complex to modulate Ras signaling, promoting Ca2+ influx through the Na+-Ca2+ exchanger and Ca2+ release-activated calcium channel. Meanwhile, SCN5A knockdown increased the 50% inhibitory concentration to 5-FU by upregulating 5-FU-stimulated apoptosis in CRCs. In conclusion, Nav1.5 could progress to proliferation and metastasis through Ca2+/calmodulin-dependent Ras signaling in CRC, and it could also enhance 5-FU-stimulated apoptosis. Clinically, patients with stage II/III CRCs with elevated SCN5A expression demonstrated poor prognosis, yet those patients could benefit more from 5-FU-based chemotherapy than patients with lower SCN5A expression.


Subject(s)
Calmodulin/genetics , Colorectal Neoplasms/drug therapy , Fluorouracil/pharmacology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Apoptosis/drug effects , Calmodulin/ultrastructure , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemotherapy, Adjuvant/adverse effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Fluorouracil/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Proto-Oncogene Proteins p21(ras)/ultrastructure
10.
Hum Cell ; 34(1): 187-200, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33040228

ABSTRACT

Cholangiocarcinoma (CCA) is one of the most aggressive and lethal malignancies. Long noncoding RNAs (lncRNAs) are being found to play crucial roles in CCA progression. This work aims to investigate the roles of long intergenic non-protein coding RNA 667 (LINC00667) in progression of CCA. RT-qPCR and western blot were applied to detect gene expression. Clinical correlation and survival were analyzed by statistical methods. Overexpression and RNA interference approaches were used to investigate the effects of LINC00667 on CCA cells. Tumor xenograft assay was performed to detect the function of LINC00667 in vivo. Transcriptional regulation and competing endogenous RNA (ceRNA) mechanism were predicted via bioinformatics analysis. ChIP, luciferase reporter, and Ago2 RIP assays further confirmed the predicted results. Our data indicated that LINC00667 was highly expressed in CCA tissues and cells, and transcription factor Yin Yang 1 (YY1) induced LINC00667 expression in CCA cells. Up-regulated LINC00667 was significantly associated with lymph node metastasis, advanced TNM stage, and poor prognosis. Knockdown of LINC00667 suppressed the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) of CCA cells, while overexpression of LINC00667 acquired opposite effects. Moreover, knockdown of LINC00667 inhibited tumor growth in vivo. In addition, LINC00667 was demonstrated to function as a ceRNA for miR-200c-3p, and then LINC00667 up-regulated pyruvate dehydrogenase kinase 1 (PDK1) to promote CCA development by inhibiting miR-200c-3p. These findings identified a pivotal role of LINC00667 in tumorigenesis and development of CCA. Targeting the YY1/LINC00667/miR-200c-3p/PDK1 axis may provide a new therapeutic strategy for CCA treatment.


Subject(s)
Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , RNA, Long Noncoding/physiology , Up-Regulation/genetics , YY1 Transcription Factor/physiology , Cell Line, Tumor , Humans
11.
PLoS One ; 15(8): e0236553, 2020.
Article in English | MEDLINE | ID: mdl-32756597

ABSTRACT

OBJECTIVES: The importance of clinical outcome prediction models using artificial intelligence (AI) is being emphasized owing to the increasing necessity of developing a clinical decision support system (CDSS) employing AI. Therefore, in this study, we proposed a "Dr. Answer" AI software based on the clinical outcome prediction model for prostate cancer treated with radical prostatectomy. METHODS: The Dr. Answer AI was developed based on a clinical outcome prediction model, with a user-friendly interface. We used 7,128 clinical data of prostate cancer treated with radical prostatectomy from three hospitals. An outcome prediction model was developed to calculate the probability of occurrence of 1) tumor, node, and metastasis (TNM) staging, 2) extracapsular extension, 3) seminal vesicle invasion, and 4) lymph node metastasis. Random forest and k-nearest neighbors algorithms were used, and the proposed system was compared with previous algorithms. RESULTS: Random forest exhibited good performance for TNM staging (recall value: 76.98%), while k-nearest neighbors exhibited good performance for extracapsular extension, seminal vesicle invasion, and lymph node metastasis (80.24%, 98.67%, and 95.45%, respectively). The Dr. Answer AI software consisted of three primary service structures: 1) patient information, 2) clinical outcome prediction, and outcomes according to the National Comprehensive Cancer Network guideline. CONCLUSION: The proposed clinical outcome prediction model could function as an effective CDSS, supporting the decisions of the physicians, while enabling the patients to understand their treatment outcomes. The Dr. Answer AI software for prostate cancer helps the doctors to explain the treatment outcomes to the patients, allowing the patients to be more confident about their treatment plans.


Subject(s)
Artificial Intelligence , Decision Support Systems, Clinical , Prognosis , Prostatic Neoplasms/epidemiology , Adult , Aged , Aged, 80 and over , Algorithms , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness/genetics , Probability , Prostate/pathology , Prostate/surgery , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/blood , Prostatic Neoplasms/physiopathology , Prostatic Neoplasms/therapy , Seminal Vesicles/pathology , Seminal Vesicles/surgery , Treatment Outcome
12.
Am J Chin Med ; 48(5): 1203-1220, 2020.
Article in English | MEDLINE | ID: mdl-32668971

ABSTRACT

Lymph node migration results in poor prognoses for nasopharyngeal carcinoma (NPC) patients. Tricetin, a flavonoid derivative, regulates tumorigenesis activity through its antiproliferative and antimetastatic properties. However, the molecular mechanism of tricetin affecting the migration and invasion of NPC cells remains poorly understood. In this paper, we examined the antimetastatic properties of tricetin in human NPC cells. Our results demonstrated that tricetin at noncytotoxic concentrations (0-80 3M) noticeably reduced the migration and invasion of NPC cells (HONE-1, NPC-39, and NPC-BM). Moreover, tricetin suppressed the indicative protease, presenilin-1 (PS-1), as indicated by protease array. PS-1 was transcriptionally inhibited via the Akt signaling pathway but not mitogen-activated protein kinase pathways, such as the JNK, p38, and ERK1/2 pathways. In addition to upregulating GSK-3[Formula: see text] phosphorylation through Akt suppression, tricetin may downregulate the activity of PS-1. Overall, our study provides new insight into the role of tricetin-induced molecular regulation in the suppression of NPC metastasis and suggests that tricetin has prospective therapeutic applications for patients with NPC.


Subject(s)
Cell Movement/drug effects , Chromones/pharmacology , Nasopharyngeal Neoplasms/genetics , Presenilin-1/genetics , Presenilin-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness/genetics
13.
Molecules ; 25(10)2020 May 21.
Article in English | MEDLINE | ID: mdl-32455624

ABSTRACT

Vitis coignetiae Pulliat (Meoru in Korea) has been used in Korean folk medicine for the treatment of inflammatory diseases and cancers. Evidence suggests that NF-κB activation is mainly involved in cancer cell proliferation, invasion, angiogenesis, and metastasis. TNF-α also enhances the inflammatory process in tumor development. Recently, flavonoids from plants have been reported to have inhibitory effects on NF-κB activities. We investigated the effects of anthocyanins extracted from the fruits of Vitis coignetiae Pulliat (AIM, anthocyanins isolated from Meoru (AIM)) on TNF-α-induced NF-κB activities in MCF-7 human breast cancer cells and the molecules involved in AIM-induced anti-cancer effects, especially on cancer metastasis. We performed cell viability assay, gelatin zymography, invasion assay, and western blot analysis to unravel the anti-NF-κB activity of AIMs on MCF-7 cells. AIM suppressed the TNF-α effects on the NF-κB-regulated proteins involved in cancer cell proliferation (COX-2, C-myc), invasion, and angiogenesis (MMP-2, MMP9, ICAM-1, and VEGF). AIM also increased the expression of E-cadherin, which is one of the hallmarks of the epithelial-mesenchymal transition (EMT) process. In conclusion, this study demonstrates that the anthocyanins isolated from the fruits of Vitis coignetiae Pulliat acts as an inhibitor of TNF-α induced NF-κB activation, and subsequent downstream molecules involved in cancer proliferation, invasion, adhesion, angiogenesis, and thus have anti-metastatic activities in MCF-7 breast cancer cells.


Subject(s)
Anthocyanins/pharmacology , Breast Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/genetics , Vitis/chemistry , Anthocyanins/chemistry , Anthocyanins/isolation & purification , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Fruit/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Proteins/genetics
14.
Am J Chin Med ; 48(4): 1021-1034, 2020.
Article in English | MEDLINE | ID: mdl-32471314

ABSTRACT

Codonopsis lanceolata roots have been widely used in Korean cuisine and traditional medicine. This study aimed to investigate the antimetastatic effects of lancemaside A, a major triterpenoid saponin, isolated from the roots of C. lanceolata, in human ovarian cancer cells. Lancemaside A significantly suppressed the migration and invasion and the expression of matrix metalloproteinases (MMPs)-2 and -9 in ovarian cancer A2780 and SKOV3 cells. Treatment with lancemaside A generated reactive oxygen species (ROS) in ovarian cancer cells. However, treatment with anti-oxidant N-acetyl-L-cysteine (NAC) significantly negated the anti-invasive activity of lancemaside A. Additionally, lancemaside A activated p38 MAP kinase, which is mediated by ROS generation. This is the first study, to our knowledge, to reveal that lancemaside A isolated from the roots of C. lanceolata exerts antimetastatic activity through inhibition of MMP expression and cancer cell invasion via activation of the ROS-mediated p38 pathway.


Subject(s)
Codonopsis/chemistry , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Ovarian Neoplasms/pathology , Plant Roots/chemistry , Reactive Oxygen Species/metabolism , Saponins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic , Cell Line, Tumor , Female , Gene Expression , Humans , Saponins/isolation & purification , p38 Mitogen-Activated Protein Kinases/genetics
15.
J Biochem Mol Toxicol ; 34(5): e22471, 2020 May.
Article in English | MEDLINE | ID: mdl-32057176

ABSTRACT

BACKGROUND: Medulloblastoma is a common tumor originates from central nervous system in children with metastatic potential. Geniposide is the major active ingredient separated from the fruit of Gardenia jasminoides Ellis. Herein, we tested the possible anticancer activity of geniposide on human medulloblastoma cells, as well as the potential underlying molecular mechanisms. METHODS: Firstly, followed by geniposide incubation, cell viability, proliferation, apoptosis, migration, and invasion of medulloblastoma Daoy cells, along with microRNA-373 (miR-373) expression were tested, respectively. Then, the influences of miR-373 overexpression in the reduction of medulloblastoma cell proliferation, migration, and invasion and the elevation of apoptosis, triggered by geniposide treatment, were re-investigated. Finally, the Ras/Raf/MEK/ERK pathway activity was analyzed. RESULTS: Geniposide treatment inhibited medulloblastoma cell viability, proliferation, migration, and invasion, but promoted cell apoptosis. Surprisingly, miR-373 expression in medulloblastoma cells was obviously downregulated by geniposide treatment. miR-373 overexpression reversed the effects of geniposide on Daoy cells. Furthermore, geniposide hindered the Ras/Raf/MEK/ERK pathway by downregulating miR-373 expression. CONCLUSION: Geniposide exhibited anticancer activity on human medulloblastoma cells and blocked Ras/Raf/MEK/ERK pathway by downregulating miR-373 expression.


Subject(s)
Antineoplastic Agents/pharmacology , Cerebellar Neoplasms/metabolism , Down-Regulation/drug effects , Fruit/chemistry , Gardenia/chemistry , Iridoids/pharmacology , Medulloblastoma/metabolism , MicroRNAs/metabolism , Plant Extracts/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cerebellar Neoplasms/pathology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Medulloblastoma/pathology , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Transfection
16.
Metallomics ; 12(3): 449-459, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32039426

ABSTRACT

Selenium (Se) is an essential trace element that has several functions in cellular processes related to cancer prevention. While the cancericidal effect of Se has been reported in liver cancer, the mechanism has not been clarified. MiR-29a has widely been reported as a tumor suppressor; however, it also acts as a carcinogenic agent by increasing cell invasion in human epithelial cancer cells and hepatoma cells. In a previous study, we found that miR-29a-3p is a Se-sensitive miRNA. However, its effect in the chicken hepatocellular carcinoma cell line (LMH) is still unknown. In the present study, we found that the expression of miR-29a-3p in LMH cells was decreased by Se supplementation and increased under Se-deficient conditions. Flow cytometry and CCK-8 results suggested that Se decreased LMH cell proliferation induced by miR-29a-3p overexpression. Transwell and gap-closure assays implied that Se mediated LMH cell invasion and migration by downregulating miR-29a-3p. Quantitative real-time polymerase chain reaction and Western blotting results suggested that Se mitigated miR-29a-3p overexpression-induced LMH cell proliferation by downregulating CDK2, cyclin-D1, CDK6, and cyclin-E1. We further demonstrated that collagen type IV alpha 2 (COL4A2) is a target gene of miR-29a-3p. COL4A2 activates the RhoA/ROCK pathway to promote LMH cell invasion and migration. In conclusion, Se mediated miR-29a-3p overexpression induced LMH cell invasion and migration by targeting COL4A2 to inactivate the RhoA/ROCK pathway.


Subject(s)
Avian Proteins/genetics , Carcinoma, Hepatocellular/veterinary , Collagen Type IV/genetics , Liver Neoplasms/veterinary , MicroRNAs/genetics , Poultry Diseases/genetics , Selenium/pharmacology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/prevention & control , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chickens/genetics , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/genetics , Liver Neoplasms/prevention & control , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/prevention & control , Poultry Diseases/prevention & control
17.
J Recept Signal Transduct Res ; 40(2): 157-165, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32019397

ABSTRACT

Introduction: This study aimed to elucidate the regulatory role and molecular regulation mechanism of miR-130b gene in the process of invasion and metastasis of hepatocarcinoma, and provide a theoretical basis for seeking of effective prevention and treatment of new targets for hepatocellular carcinoma.Materials and methods: The expression level of miR-130b gene in hepatocarcinoma tissues was determined by qRT-PCR. The biological function and mechanism of miR-130b gene were verified by cell and animal models, and the target gene was verified by double luciferase assay.Results: In the liver cancer tissues of patients with metastasis, the expression level of miR-130b gene was increased, and the difference was significantly significant (p < 0.05). Evaluation of independent risk factors for overall survival showed significant difference (p < 0.01). Up-regulation of miR-130b in MHCC97L- subpopulation cells significantly enhanced the invasion and migration ability, and the difference was statistically significant (p < 0.05). The invasion and migration ability of MHCC97H + subpopulation cells with increased expression of miR-130b was significantly decreased, and the difference was notably significant (p < 0.05). When the expression of miR-130b in MHCC97H + subpopulation cells was inhibited, the expressions of Notch-Dll1 and SOX2, Nanog and E2F3 proteins in transplanted tumor tissues were significantly higher than those in other groups (p < 0.05). When miR-130b in MHCC97L- subpopulation cells was up-regulated, the expressions of Notch-Dll1 and Bcl-2, CCND1, Nanog and MET proteins in transplanted tumor tissues were significantly increased than those in other groups (p < 0.05). The prediction results of bioinformatics data suggest that the target gene of miR-130b may be Notch-Dll1 gene. The experiment of luciferase reporter gene confirmed that miR-130b gene can be inhibited and contains fluorescent reporter gene with complementary binding site, lost activity.Conclusion: The miR-130b gene can inhibit the protein expression of Notch-Dll1, and it can promote the invasion and metastasis of liver cancer cells.


Subject(s)
Calcium-Binding Proteins/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Animals , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease-Free Survival , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Liver Neoplasms/epidemiology , Liver Neoplasms/pathology , Male , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Receptors, Notch/genetics , Signal Transduction/genetics
18.
Clin Exp Metastasis ; 37(2): 283-292, 2020 04.
Article in English | MEDLINE | ID: mdl-32020377

ABSTRACT

We explored the role of the transcription factor, NF-κB, and its upstream kinase IKKß in regulation of migration, invasion, and metastasis of cisplatin-resistant head and neck squamous cell carcinoma (HNSCC). We showed that cisplatin-resistant HNSCC cells have a stronger ability to migrate and invade, as well as display higher IKKß/NF-κB activity compared to their parental partners. Importantly, we found that knockdown of IKKß, but not NF-κB, dramatically impaired cell migration and invasion in these cells. Consistent with this, the IKKß inhibitor, CmpdA, also inhibited cell migration and invasion. Previous studies have already shown that N-Cadherin, an epithelial-mesenchymal transition (EMT) marker, and IL-6, a pro-inflammatory cytokine, play important roles in regulation of HNSCC migration, invasion, and metastasis. We found that cisplatin-resistant HNSCC expressed higher levels of N-Cadherin and IL-6, which were significantly inhibited by CmpdA. More importantly, we showed that CmpdA treatment dramatically abated cisplatin-resistant HNSCC cell metastasis to lungs in a mouse model. Our data demonstrated the crucial role of IKKß in control of migration, invasion, and metastasis, and implicated that targeting IKKß may be a potential therapy for cisplatin-resistant metastatic HNSCC.


Subject(s)
Head and Neck Neoplasms/drug therapy , I-kappa B Kinase/antagonists & inhibitors , Lung Neoplasms/prevention & control , NF-kappa B/metabolism , Oxazines/therapeutic use , Pyridines/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Knockdown Techniques , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/genetics , Neoplasm Metastasis/prevention & control , Oxazines/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/secondary , Xenograft Model Antitumor Assays
19.
Biomed Pharmacother ; 124: 109887, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31986414

ABSTRACT

PURPOSE: Long non-coding RNA (LncRNA) HOX transcript antisense RNA (HOTAIR) and Yin Yang 1 (YY1) are reported to be involved in tumorigenesis. However, the effect and molecular mechanism of HOTAIR on YY1 expression remains poorly understood. The study aimed to investigate the functions and molecular mechanism of LncRNA HOTAIR in medulloblastoma progression. METHODS: qPCR was performed to detect HOTAIR and YY1 mRNA in tissues and cells, as well as that of miR-1 and miR-206 expression levels. Western blot assay was used to test YY1 and EMT-related biomarkers' protein levels. Cell proliferation was tested with CCK-8 assay and colony formation assay. Migration and invasion abilities were tested with Transwell migration and invasion assays. Tumor growth was tested with an in vivo animal study. Cell apoptosis was tested with an Annexin V-FITC/PI kit. Luciferase assay was used to test the luciferase intensity of YY1 and HOTAIR. RNA pull down assay was used to detect the combination between HOTAIR and miR-1/miR-206. RESULTS: In this study, we found that HOTAIR and YY1 were up-regulated in medulloblastoma tissues and cell lines, and HOTAIR increased YY1 expression. The molecular mechanism demonstrated that HOTAIR negatively regulated miR-1 and miR-206 expression, which can directly target YY1 in medulloblastoma cells. Moreover, HOTAIR increased YY1 expression through binding to miR-1 and miR-206. The functional experiments showed that HOTAIR knockdown suppressed medulloblastoma cell proliferation, tumor growth, migration and invasion, and promoted cell apoptosis via the modulation of the miR-1/miR-206-YY1 axis, as well as epithelial to mesenchymal transition (EMT). CONCLUSION: These data indicate that HOTAIR promotes medulloblastoma progression via acting as a competing endogenous RNA (ceRNA) to regulate YY1 expression through binding to miR-1 and miR-206.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , RNA, Long Noncoding/genetics , YY1 Transcription Factor/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cerebellar Neoplasms/pathology , Disease Progression , Female , Gene Knockdown Techniques , Humans , Medulloblastoma/pathology , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Xenograft Model Antitumor Assays
20.
Oncogene ; 39(3): 617-636, 2020 01.
Article in English | MEDLINE | ID: mdl-31527668

ABSTRACT

The basic understanding of the biological effects of eukaryotic translation initiation factors (EIFs) remains incomplete, notably for their roles independent of protein translation. Different EIFs exhibit nuclear localization and DNA-related functions have been proposed, but the understanding of EIFs novel functions beyond protein translation lacks of integrative analyses between the genomic and the proteomic levels. Here, the noncanonical function of EIF3F was studied in human lung adenocarcinoma by combining methods that revealed both the protein-protein and the protein-DNA interactions of this factor. We discovered that EIF3F promotes cell metastasis in vivo. The underpinning molecular mechanisms involved the regulation of a cluster of 34 metastasis-promoting genes including Snail2, as revealed by proteomics combined with immuno-affinity purification of EIF3F and ChIP-seq/Q-PCR analyses. The interaction between EIF3F and signal transducer and activator of transcription 3 (STAT3) controlled the EIF3F-mediated increase in Snail2 expression and cellular invasion, which were specifically abrogated using the STAT3 inhibitor Nifuroxazide or knockdown approaches. Furthermore, EIF3F overexpression reprogrammed energy metabolism through the activation of AMP-activated protein kinase and the stimulation of oxidative phosphorylation. Our findings demonstrate the role of EIF3F in the molecular control of cell migration, invasion, bioenergetics, and metastasis. The discovery of a role for EIF3F-STAT3 interaction in the genetic control of cell migration and metastasis in human lung adenocarcinoma could lead to the development of diagnosis and therapeutic strategies.


Subject(s)
Adenocarcinoma of Lung/genetics , Cell Nucleus/metabolism , Energy Metabolism/genetics , Eukaryotic Initiation Factor-3/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , STAT3 Transcription Factor/metabolism , A549 Cells , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cell Nucleus/genetics , Cell Nucleus/pathology , Datasets as Topic , Energy Metabolism/drug effects , Eukaryotic Initiation Factor-3/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Hydroxybenzoates/pharmacology , Lung/cytology , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Mutation , Neoplasm Invasiveness/genetics , Nitrofurans/pharmacology , Oxidative Phosphorylation/drug effects , RNA, Small Interfering/metabolism , RNA-Seq , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Snail Family Transcription Factors/genetics , Survival Analysis , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL