Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Acta Biochim Biophys Sin (Shanghai) ; 55(1): 34-42, 2022 Dec 25.
Article in English | MEDLINE | ID: mdl-36647720

ABSTRACT

Despite favorable responses to platinum-based chemotherapy in ovarian cancer (OC), chemoresistance is still a major cause of treatment failure. Hence, we develop a novel synthetic agent, COM33, to relieve the chemoresistance caused by carboplatin. The anti-cancerous effects of the combination of COM33 and carboplatin on OC are evaluated by cell viability, wound healing, and transwell invasion assays. A mechanistic investigation is carried out by using RNA-Seq analysis and then verified by western blot analysis and immunofluorescence microscopy. The safety and efficacy in vivo are evaluated using SKOV3 tumor-bearing nude mice. Results show that the co-administration of COM33 enhances the inhibitory effects of carboplatin on cancer cell viability, migration, and invasion in vitro and tumor growth in vivo. Furthermore, COM33 suppresses the carboplatin-induced epithelial-mesenchymal transition (EMT) by inhibiting the ERK signaling pathway. Additionally, we show that Twist1, the effector of the ERK signaling pathway, participates in carboplatin-induced EMT and is also inhibited by COM33. Our data show that the combination of carboplatin with COM33 is beneficial for chemotherapy against OC, which may be a potential novel anti-tumor strategy.


Subject(s)
Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Ovarian Neoplasms , Twist-Related Protein 1 , Animals , Female , Humans , Mice , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Carboplatin/adverse effects , Carboplatin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Signal Transduction/drug effects , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/metabolism
2.
Drug Des Devel Ther ; 14: 5633-5644, 2020.
Article in English | MEDLINE | ID: mdl-33376307

ABSTRACT

BACKGROUND: Magnesium isoglycyrrhizinate (MI) was extracted from roots of the plant Glycyrrhiza glabra, which displays multiple pharmacological activities such as anti-inflammation, anti-apoptosis, and anti-tumor. Here, we aimed to investigate the effect of MI on the progression and epithelial-mesenchymal transition (EMT) of laryngeal cancer. METHODS: Forty laryngeal cancer clinical samples were used. The role of MI in the proliferation of laryngeal cancer cells was assessed by MTT assay, Edu assay and colony formation assay. The function of MI in the migration and invasion of laryngeal cancer cells was tested by transwell assays. The effect of MI on apoptosis of laryngeal cancer cells was determined by cell apoptosis assay. The impact of MI on tumor growth in vivo was analyzed by tumorigenicity analysis using Balb/c nude mice. qPCR and Western blot analysis were performed to measure the expression levels of gene and protein, respectively. RESULTS: We identified that EMT-related transcription factor Twist was significantly elevated in the laryngeal cancer tissues. The expression of Twist was also enhanced in the human laryngeal carcinoma HEP-2 cells compared with that in the primary laryngeal epithelial cells. The high expression of Twist was remarkably correlated with poor overall survival of patients with laryngeal cancer. Meanwhile, our data revealed that MI reduced cell proliferation, migration and invasion and enhanced apoptosis of laryngeal cancer cells in vitro. Moreover, MI decreased transcriptional activation and the expression levels of NF-κB and Twist, and alleviated EMT in vitro and in vivo. MI remarkably inhibited tumor growth and EMT of laryngeal cancer cells in vivo. CONCLUSION: MI restrains the progression of laryngeal cancer and induces an inhibitory effect on EMT in laryngeal cancer by modulating the NF-κB/Twist signaling. Our finding provides new insights into the mechanism by which MI inhibits laryngeal carcinoma development, enriching the understanding of the anti-tumor function of MI.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Laryngeal Neoplasms/drug therapy , NF-kappa B/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Saponins/pharmacology , Triterpenes/pharmacology , Twist-Related Protein 1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Twist-Related Protein 1/metabolism
3.
Biochem Pharmacol ; 180: 114152, 2020 10.
Article in English | MEDLINE | ID: mdl-32679125

ABSTRACT

α-Linolenic acid (ALA), an essential fatty acid, has anticancer activity in breast cancer, but the mechanism of its effects in triple-negative breast cancer (TNBC) remains unclear. We investigated the effect of ALA on Twist1, which is required to initiate epithelial-mesenchymal transition (EMT) and promotes tumor metastasis, and Twist1-mediated migration in MDA-MB231, MDA-MB468 and Hs578T cells. Twist1 protein was constitutively expressed in these TNBC cells, particularly MDA-MB-231 cells. Treatment with 100 µM ALA and Twist1 siRNA markedly decreased the Twist1 protein level and cell migration. Moreover, ALA transiently attenuated the nuclear accumulation of STAT3α as well as Twist1 mRNA expression. Treatment with ALA significantly attenuated the phosphorylation of JNK, ERK and Akt and decreased the phosphorylation of Twist1 at serine 68 in MDA-MB-231 cells. ALA accelerated Twist1 degradation in the presence of cycloheximide, whereas the ubiquitination and degradation of Twist1 by ALA was suppressed by MG-132. Pretreatment with ALA mimicked Twist1 siRNA, increased the protein expression of epithelial markers such as E-cadherin, and decreased the protein expression of mesenchymal markers including Twist1, Snail2, N-cadherin, vimentin, and fibronectin. Our findings suggest that ALA can be used not only to abolish EMT but also to suppress Twist1-mediated migration in TNBC cells.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/biosynthesis , Triple Negative Breast Neoplasms/metabolism , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/biosynthesis , alpha-Linolenic Acid/pharmacology , Cell Line, Tumor , Cell Movement , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Nuclear Proteins/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Twist-Related Protein 1/genetics , alpha-Linolenic Acid/therapeutic use
4.
Fitoterapia ; 137: 104275, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31351126

ABSTRACT

The non-small cell lung cancer (NSCLC) represents a malignant type of cancer worldwide. The atalantraflavone (AFL) is a natural product isolated from leaves of Atalantia monophylla (L.) DC. However, the function of atalantraflavone in NSCLC is still elusive. In present work, we have unraveled a novel function of AFL in NSCLC. AFL significantly inhibited NSCLC cell viability and colony formation. AFL increased sub-G1 fraction and apoptotic rates in a dose-dependent manner. Furthermore, Twist-related protein 1 (Twist1) was identified as the target of AFL. The association between AFL and Twist1 markedly decreased the stability of Twist1 via elevated ubiquitin mediated proteasomal degradation. AFL induced NSCLC suppression was mediated by Twist1 as Twist1 overexpression could partially reverse the inhibitory effect of AFL on migration and metastasis. Furthermore, AFL could also sensitize NSCLC cells to cisplatin treatment and consistently impair NSCLC proliferation and metastasis. Our current data have identified a tumor suppressive function for AFL in NSCLC by increasing Twist1 degradation. Therefore, the anti-tumor activity of AFL might provide critical insight into pharmaceutic lung cancer intervention to overcome cisplatin resistance.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Flavones/pharmacology , Nuclear Proteins/antagonists & inhibitors , Rutaceae/chemistry , Twist-Related Protein 1/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cisplatin , Drug Resistance, Neoplasm , Flavones/isolation & purification , Humans , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Plant Leaves/chemistry , Protein Stability
5.
Biochem Biophys Res Commun ; 516(3): 765-769, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31255287

ABSTRACT

The direct conversion of accessible cells such as human fibroblasts to inaccessible cells, particularly neurons, opens up many opportunities for using the human model system to study diseases and discover therapies. Previous studies have indicated that the neuronal conversion of adult human skin fibroblasts is much harder than that for human lung fibroblasts, which are used in many experiments. Here we formally report this differential plasticity of human skin versus lung fibroblasts in their transdifferentiation to induced neurons. Using RNAseq of isogenic and non-isogenic pairs of human skin and lung fibroblasts at different days in their conversion to neurons, we found that several master regulators (TWIST1, TWIST2, PRRX1 and PRRX2) in the fibroblast Gene Regulatory Network were significantly downregulated in lung fibroblasts, but not in skin fibroblasts. By knocking down each of these genes and other genes that suppress the neural fate, such as REST, HES1 and HEY2, we found that the combined attenuation of HEY2 and PRRX2 significantly enhanced the transdifferentiation of human skin fibroblasts induced by ASCL1 and p53 shRNA. The new method, which overexpressed ASCL1 and knocked down p53, HEY2 and PRRX2 (ApH2P2), enabled the efficient transdifferentiation of adult human skin fibroblasts to MAP2+ neurons in 14 days. It would be useful for a variety of applications that require the efficient and speedy derivation of patient-specific neurons from skin fibroblasts.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Fibroblasts/metabolism , Homeodomain Proteins/genetics , Repressor Proteins/genetics , Skin/metabolism , Tumor Suppressor Protein p53/genetics , Adult , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Transdifferentiation , Cellular Reprogramming , Fibroblasts/cytology , Gene Expression Regulation , Gene Regulatory Networks , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/metabolism , Humans , Lung/cytology , Lung/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Signal Transduction , Skin/cytology , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
6.
Mol Med Rep ; 19(6): 5301-5308, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31059108

ABSTRACT

Temozolomide (TMZ) is widely used as a chemotherapeutic agent in the treatment of glioma; however, the development of drug resistance remains a major obstacle in the effective treatment of glioblastoma. Increasing evidence has indicated that microRNAs (miRs) are involved in the drug resistance of glioma; however, the role of miR­186­5p in the TMZ resistance of glioblastoma remains unknown. In the present study, the role of miR­186­5p in the resistance of glioblastoma to TMZ was investigated. mRNA and protein expression levels were detected via reverse transcription­quantitative PCR and western blot analysis, respectively. It was determined that miR­186­5p was significantly downregulated in glioblastoma tissues and cell lines. Additionally, the expression of miR­186­5p was decreased, whereas that of Twist1 was upregulated during the development of drug resistance in glioma cells. The introduction of miR­186 into glioblastoma cells via transfection decreased the proliferation and TMZ resistance of glioblastoma cells, as determined via 5­ethynyl­2'­deoxyuridine and Cell Counting Kit­8 assays, whereas the inhibition of miR­186­5p induced opposing effects. Furthermore, luciferase reporter and expression rescue assays revealed that miR­186­5p bound to the 3'­untranslated region of Twist­related protein 1 (Twist1). In conclusion, the present study demonstrated that downregulation of miR­186­5p may contribute to the proliferation and drug resistance of glioblastoma cells via the regulation of Twist1 expression. These results suggested that miR­186­5p may be a novel therapeutic target in the treatment of glioblastoma.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , MicroRNAs/metabolism , Nuclear Proteins/metabolism , Twist-Related Protein 1/metabolism , 3' Untranslated Regions , Adult , Aged , Antagomirs/metabolism , Brain Neoplasms/metabolism , Case-Control Studies , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Drug Resistance, Neoplasm/genetics , Female , Glioblastoma/metabolism , Humans , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Temozolomide/pharmacology , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics
7.
Cell Death Dis ; 10(5): 352, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040266

ABSTRACT

Dysregulation of protein tyrosine phosphatase, receptor type B (PTPRB) correlates with the development of a variety of tumors. Here we show that PTPRB promotes metastasis of colorectal cancer (CRC) cells via inducing epithelial-mesenchymal transition (EMT). We find that PTPRB is expressed at significantly higher levels in CRC tissues compared to adjacent nontumor tissues and in CRC cell lines with high invasion. PTPRB knockdown decreased the number of invasive CRC cells in an in vitro wound healing model, and also reduced tumor metastasis in vivo. Conversely, PTPRB overexpression promoted CRC cell invasion in vitro and metastasis in vivo. PTPRB overexpression decreased vimentin expression and promoted E-cadherin expression, consistent with promotion of EMT, while PTPRB knockdown had the opposite effect. Hypoxic conditions induced EMT and promoted invasion in CRC cells, but these effects were eliminated by PTPRB knockdown. EMT blockade via TWIST1 knockdown inhibited the migration and invasiveness of CRC cells, and even increased PTPRB expression could not reverse this effect. Altogether, these data support the conclusion that PTPRB promotes invasion and metastasis of CRC cells via inducing EMT, and that PTPRB would be a novel therapeutic target for the treatment of CRC.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Transplantation, Heterologous , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
8.
Cell Death Dis ; 10(6): 410, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138781

ABSTRACT

Twist1, a key transcription factor regulating epithelial-mesenchymal transition and cancer metastasis, is highly expressed in invasive cancers in contrast to the loss of BTG2/TIS21 expression. Based on our observation that forced expression of BTG2/TIS21 downregulated Twist1 protein expression without altering mRNA level, we investigated molecular mechanisms of the BTG2/TIS21-inhibited Twist1 translation in the triple negative breast cancer (TNBC) cells and in vivo BTG2/TIS21-knockout (KO) mice and human breast cancer tissues. (1) C-terminal domain of Twist1 and Box B of BTG2/TIS21 interacted with each other, which abrogated Twist1 activity. (2) BTG2/TIS21 inhibited translational initiation by depleting eIF4E availability via inhibiting 4EBP1 phosphorylation. (3) Expression of BTG2/TIS21 maintained p-eIF2α that downregulates initiation of protein translation, confirmed by eIF2α-AA mutant expression and BTG2/TIS21 knockdown in MEF cells. (4) cDNA microarray analysis revealed significantly higher expression of initiation factors-eIF2A, eIF3A, and eIF4G2-in the BTG2/TIS21-KO mouse than that in the wild type. (5) BTG2/TIS21-inhibited translation initiation lead to the collapse of polysome formation and the huge peak of 80s monomer in the BTG2/TIS21 expresser, but not in the control. (6) mRNAs and protein expressions of elongation factors were also downregulated by BTG2/TIS21 expression in TNBC cells, but much higher in both TIS21-KO mice and lymph node-positive human breast cancers. (7) BTG2/TIS21-mediated Twist1 loss was not due to the protein degradation by ubiquitination and autophagy activation. (8) Twist1 protein level was significantly higher in various organs of TIS21-KO mice compared with that in the control, indicating the in vivo role of BTG2/TIS21 gene in the regulation of Twist1 protein level. Altogether, the present study support our hypothesis that BTG2/TIS21 is a promising target to combat with metastatic cancers with high level of Twist1 without BTG2/TIS21 expression.


Subject(s)
Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Chain Initiation, Translational/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Twist-Related Protein 1/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Down-Regulation , Eukaryotic Initiation Factor-2/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Lysosomes/genetics , Lysosomes/metabolism , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Peptide Elongation Factors/antagonists & inhibitors , Peptide Elongation Factors/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , Triple Negative Breast Neoplasms/genetics , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics
9.
Cell Physiol Biochem ; 50(5): 2004-2016, 2018.
Article in English | MEDLINE | ID: mdl-30404076

ABSTRACT

BACKGROUND/AIMS: Non-small-cell lung carcinoma (NSCLC) is the leading cause of cancer death, with tumor metastasis being mainly responsible for lung cancer-associated mortality. Our previous studies have found that tubulin polymerization promoting protein family member 3 (TPPP3) acted as a potential oncogene in NSCLC. Little is known about the function of TPPP3 in tumor metastasis. METHODS: RT-qPCR and IHC were used to investigate the expression of TPPP3 in NSCLC tissues. CCK8 assay and transwell assay were used to measure proliferation and migration of NSCLC cells in vitro and xenograft model was performed to assess the tumor growth and metastasis in vivo. RESULTS: In the present study, upregulation of TPPP3 was found to correlate with an increased metastasis capability of NSCLC. Ectopic expression of TPPP3 significantly enhanced cell proliferation in vitro and promoted tumor growth in vivo. Furthermore, overexpression of TPPP3 remarkably promoted NSCLC cell migration and invasion along with the upregulation of Twist1 both in vitro and in vivo. Further investigations showed that activation of STAT3 was required for TPPP3-mediated upregulation of Twist1, cell migration and invasion. A strong positive correlation between TPPP3 and Twist1 expression was identified in NSCLC tissues. Patients with low TPPP3 or low Twist1 in NSCLC tissues had a better prognosis with longer overall survival (OS) and disease-free survival (DFS). CONCLUSION: Overall, this study demonstrates that TPPP3 promotes the metastasis of NSCLC through the STAT3/Twist1 pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules/metabolism , Lung Neoplasms/pathology , Nuclear Proteins/metabolism , STAT3 Transcription Factor/metabolism , Twist-Related Protein 1/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Phenanthrenes/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics
10.
Cell Physiol Biochem ; 50(2): 501-511, 2018.
Article in English | MEDLINE | ID: mdl-30308498

ABSTRACT

BACKGROUND/AIMS: Lymph node metastasis is the primary cause of cancer-related death among patients with gastric cancer (GC), and cell division cycle 27 (CDC27) promotes the metastasis and epithelial-mesenchymal transition in many cancers. Till now, the mechanisms underlying CDC27-induced the epithelial-mesenchymal transition (EMT) of GC are still unclear. METHODS: We analyzed the expression levels of CDC27 and EMT-related biomarkers using immunohistochemistry and Western blot in 60 cases of GC tissues, and then GC cells with CDC27 shRNAs or plasmids were subjected to in vitro and in vivo assays, including CCK-8, wound healing and transwell assays. RESULTS: The CDC27 expression was obviously increased in GC tissues, and significantly correlates with EMT-related biomarkers, lymph node metastasis and poor 5-year overall survival. Additionally, in vitro and in vivo assays demonstrated that silencing of CDC27 expression effectively inhibited GC cell proliferation, invasion and metastasis. Conversely, CDC27 overexpression led to the opposite results. Finally, we demonstrated that Twist shRNA inhibited CDC27-meditated invasion and EMT of GC cells. CONCLUSION: CDC27 facilitates gastric cancer cell proliferation, invasion and metastasis via Twist-induced EMT; thus, this study offered a new therapy method for GC patients.


Subject(s)
Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Nuclear Proteins/metabolism , Stomach Neoplasms/pathology , Twist-Related Protein 1/metabolism , Adult , Aged , Animals , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/antagonists & inhibitors , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Lymphatic Metastasis , Male , Mice , Mice, Nude , Middle Aged , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Rate , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics
11.
J Biosci ; 42(4): 547-554, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29229873

ABSTRACT

Gastric cancer is one of the lethal causes of cancer-related deaths worldwide. The incidence and mortality rates of this disease is comparatively higher in China. In the current study, we evaluated the anticancer effects of Thymoquinone (TQ) against gastric cancer cells (MGC80-3 and SGC-7901) and normal noncancerous GES-1 cells and attempted to investigate the underlying mechanism. Our results indicated that TQ exhibited significant growth inhibitory effects on gastric cancer cells (MGC80-3 and SGC-7901). However, lower cytotoxicity was observed against normal GES-1 cells. Moreover, TQ could inhibit the colony formation potential of MGC80-3 and SGC-7901 cells in a dose-dependent manner. TQ also inhibited cell migration ability of the gastric cancer cells and down-regulated the expression of the mesenchymal genes such as N-cadherin, Vimentin, and TWIST. However, the epithelial markers such as E-cadherin and cytokeratin-19 were distinctly up-regulated in TQ-treated gastric cancer cells. Since PI3K/Akt/ mTOR plays an important role in progression and tumorigenesis, we also investigated the effect of TQ on PI3K/Akt/mTOR signalling pathway in gastric cancer cells. It was observed that TQ down-regulated the expression of some of the key proteins of this pathway. Taken together, we conclude that TQ may prove lead molecule for the treatment of gastric cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzoquinones/pharmacology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/drug effects , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gastric Mucosa/metabolism , Humans , Inhibitory Concentration 50 , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stomach/drug effects , Stomach/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Vimentin/antagonists & inhibitors , Vimentin/genetics , Vimentin/metabolism
12.
Eur Rev Med Pharmacol Sci ; 21(13): 3061-3068, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28742200

ABSTRACT

OBJECTIVE: To investigate whether hypoxia microenvironment induced hepatocellular carcinoma cells SMMC-7721 epithelial-mesenchymal transition (EMT) and to explore the underlying molecular mechanism. MATERIALS AND METHODS: In this study, SMMC-7721 cells were cultured under normoxia and hypoxia conditions, respectively. RT-PCR and Western blot were used to monitor the expression level of EMT-related markers, E-cadherin, and vimentin, as well as hypoxia inducible factor-1α (HIF-1α) and Twist1. Then we performed the transwell invasion assays to detect the ability of cell invasion. RESULTS: The results demonstrated that hypoxia micro-environment could induce hepatocellular carcinoma cells SMMC-7721 EMT and enhance the cell invasion ability. Furthermore, knockdown of Twist1 by using specific siRNA could reverse hypoxia-induced EMT process. CONCLUSIONS: Hypoxia promotes hepatocellular carcinoma cells SMMC-7721 EMT by upregulating the expression of Twist1.


Subject(s)
Cell Hypoxia , Epithelial-Mesenchymal Transition/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/metabolism , Antigens, CD , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Transcriptional Activation , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Vimentin/genetics , Vimentin/metabolism
13.
Oncol Rep ; 38(2): 1224-1232, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28677802

ABSTRACT

Epithelial-mesenchymal transition (EMT), plays a vital role in hepatocellular carcinoma (HCC) development and metastasis. Norcantharidin (NCTD; 7-oxabicyclo (2.2.1) heptane-2,3-dicarboxylic anhydride) plays anticancer roles in the regulation of tumor cell proliferation, apoptosis and migration. However, the molecular mechanism of HCC EMT and the effects of NCTD in the HCC EMT process have been either poorly elucidated or not studied. In this study, HCC EMT was induced by the treatment of IL-6 and various concentrations of NCTD (0, 30, 60 and 120 µM) were treated with HCC cell lines, HCCLM3 and SMMC-7721. We investigated the effect of NCTD on the invasion of HCC cells by using Transwell assay. Immunofluorescence staining, western blot analysis and quantitative RT-PCR were performed to evaluate the protein and mRNA expression levels of HCC cells. Here, using cell line models, our data demonstrated that interleukin 6 (IL-6) induced EMT through the JAK/STAT3/TWIST pathway in HCC. Moreover, our studies revealed that NCTD markedly inhibited IL-6-induced EMT and cell invasiveness. Signaling studies revealed that NCTD sufficiently suppressed JAK/STAT3/TWIST signaling to reverse the IL-6-promoting effects. Collectively, these data provide evidence for the use of NCTD as a potential anticancer drug in HCC metastatic patients.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Interleukin-6/pharmacology , Janus Kinase 2/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Twist-Related Protein 1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Janus Kinase 2/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasm Invasiveness , STAT3 Transcription Factor/metabolism , Tumor Cells, Cultured , Twist-Related Protein 1/metabolism
14.
Int J Oncol ; 51(2): 515-524, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28627611

ABSTRACT

Inherent and acquired chemoresistance reduce the effectiveness of irinotecan in the treatment of metastatic colorectal cancer (CRC). However, the molecular mechanisms underlying this resistance process are still unclear. Twist1 is one of the master transcription factors of epithelial-mesenchymal transition (EMT). Our previous study indicated that Twist1 is overexpressed in colon cancer tissues, and demonstrated that Twist1 plays a crucial role in the chemoresistance of CRC. In the present study, we further investigated how Twist1 contribute to acquired resistance to irinotecan in colon cancer. The irinotecan-resistant cells were established by gradual adaptation of increasing irinotecan concentrations in LoVo cells, named LoVo/CPT-11R cells. Results showed that cell viabilities to different anticancer drugs were markedly increased in LoVo/CPT-11R cells compared to LoVo cells. Moreover, LoVo/CPT-11R cells displayed EMT, CSC-like cellular morphology and relative biomarkers were also significantly increased. In addition, overexpressed Twist1 LoVo cells were established by lentivirus transfection assay, named LoVo/Twist1 cells. Results showed that the LoVo/Twist1 cells perform a distinctly decreased sensitivity to irinotecan, downregulated expression of E-cadherin, upregulated expression of cluster of differentiation 44 (CD44), and a significant enhancement of invasion and migration potential by regulation of MMP2 compared with control cells. In contrast, the inhibition of Twist1 transfected with siRNA could enhance the irinotecan sensitivity in LoVo/CPT-11R cells and downregulate the expression of vimentin and CD44. Our data provide evidence that EMT and CSC-like phenotype induced by Twist1 contribute to acquire resistance to irinotecan and enhanced migration and invasion in colon cancer.


Subject(s)
Camptothecin/analogs & derivatives , Cell Proliferation/genetics , Colonic Neoplasms/drug therapy , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , Camptothecin/administration & dosage , Camptothecin/adverse effects , Cell Movement/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Irinotecan , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/drug effects , Nuclear Proteins/antagonists & inhibitors , RNA, Small Interfering/genetics , Transfection , Twist-Related Protein 1/antagonists & inhibitors
15.
Oncol Rep ; 37(3): 1849-1856, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28184914

ABSTRACT

MicroRNA-361-5p (miR-361-5p) has been reported to be dysregulated in various human cancer types. However, the function of miR-361-5p in glioma remains unknown. In the present study, we aimed to investigate the biological functions of miR-361-5p in regulating glioma progression and the underlying molecular mechanism. We found that miR-361-5p was significantly decreased in glioma tissues and cell lines as detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis. Functional analysis revealed that miR-361-5p overexpression significantly inhibited glioma cell migration, invasion and epithelial-mesenchymal transition (EMT) whereas suppression of miR-361-5p showed opposite effects. Bioinformatic analysis showed that Twist1, a critical EMT inducer, was a predicted target of miR-361-5p which was validated by dual-luciferase reporter assay, RT-qPCR and western blot analysis. Further analysis indicated that miR-361-5p regulates the Twist1/Bmi-1 signaling axis. Rescue experiments showed that restoration of Twist1 expression significantly reversed the suppressive effect of miR-361-5p on cell migration, invasion and EMT. Taken together, the present study demonstrated an important role of miR-361-5p in glioma - which regulated the EMT of glioma cells by targeting and regulating Twist1. These findings provide novel insight into understanding the role and mechanism of miR-361-5p in regulating the biolo-gical behavior of glioma cells and suggest that miR-361-5p is a novel potential therapeutic target for glioma.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Glioma/pathology , MicroRNAs/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/metabolism , Blotting, Western , Cell Movement , Glioma/genetics , Glioma/metabolism , Humans , Neoplasm Invasiveness , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics
16.
Gene ; 600: 1-8, 2017 Feb 05.
Article in English | MEDLINE | ID: mdl-27884772

ABSTRACT

BACKGROUND: Associated with recent achievements in therapy for advanced lung adenocarcinoma, there will still be an unmet medical need for effective treatment of stage IIIb/IV, and the prognosis of lung cancer is not optimistic till now. OBJECTIVE: In order to obtain some essential evidences for a potential targeted therapy in lung adenocarcinoma, the effects of Ovol2 gene on Epithelial-to-Mesenchymal Transition (EMT) was observed and the probable mechanisms were analyzed. METHODS: Ovol2 expression was previously evaluated by immunochemistry in lung adenocarcinoma tissue, and Ovol2 was overexpressed by lentivirus infection in A549 cells. Subsequently, the migration and invasion ability of A549 cells was tested by Transwell and Wound healing experiments. The mRNA level of genes correlated to EMT was detected by Real-time PCR, and the expression of reasonable makers was probed by Western Blot. Finally, rescue experiment, Luciferase assay, and chromatin immunoprecipitation assay were performed to explore the probable mechanisms. RESULTS: After treated with Ovol2 overexpression, the expression level of E-cadherin was increased, while the expression level of Vimentin and Twist1 was declined not only in the mRNA level but also in the protein level. Moreover, we found that Ovol2 represses transcription of Twist1 by binding to its promoter directly. Wound healing and Transwell assays indicate that the migration and invasion ability were downregulated by Ovol2 in A549 cells. CONCLUSION: Ovol2 can suppress migration and invasion ability of A549 cells, and prevent EMT by inhibition of Twist1 transcription directly.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/genetics , Twist-Related Protein 1/antagonists & inhibitors , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Adult , Aged , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Models, Biological , Neoplasm Invasiveness/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Transcription Factors/agonists , Transcription Factors/metabolism , Transcription, Genetic , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Up-Regulation
17.
J Am Heart Assoc ; 5(12)2016 12 07.
Article in English | MEDLINE | ID: mdl-27927633

ABSTRACT

BACKGROUND: MicroRNA miR-214 has been implicated in many biological cellular functions, but the impact of miR-214 and its target genes on vascular smooth muscle cell (VSMC) proliferation, migration, and neointima smooth muscle cell hyperplasia is unknown. METHODS AND RESULTS: Expression of miR-214 was closely regulated by different pathogenic stimuli in VSMCs through a transcriptional mechanism and decreased in response to vascular injury. Overexpression of miR-214 in serum-starved VSMCs significantly decreased VSMC proliferation and migration, whereas knockdown of miR-214 dramatically increased VSMC proliferation and migration. Gene and protein biochemical assays, including proteomic analyses, showed that NCK associated protein 1 (NCKAP1)-a major component of the WAVE complex that regulates lamellipodia formation and cell motility-was negatively regulated by miR-214 in VSMCs. Luciferase assays showed that miR-214 substantially repressed wild-type but not the miR-214 binding site mutated version of NCKAP1 3' untranslated region luciferase activity in VSMCs. This result confirmed that NCKAP1 is the functional target of miR-214 in VSMCs. NCKAP1 knockdown in VSMCs recapitulates the inhibitory effects of miR-214 overexpression on actin polymerization, cell migration, and proliferation. Data from cotransfection experiments also revealed that inhibition of NCKAP1 is required for miR-214-mediated lamellipodia formation, cell motility, and growth. Importantly, locally enforced expression of miR-214 in the injured vessels significantly reduced NCKAP1 expression levels, inhibited VSMC proliferation, and prevented neointima smooth muscle cell hyperplasia after injury. CONCLUSIONS: We uncovered an important role of miR-214 and its target gene NCKAP1 in modulating VSMC functions and neointima hyperplasia. Our findings suggest that miR-214 represents a potential therapeutic target for vascular diseases.


Subject(s)
Membrane Proteins/physiology , MicroRNAs/physiology , Neointima/pathology , Angiogenesis Inducing Agents/pharmacology , Animals , Becaplermin , Binding Sites/genetics , Cell Line , Cell Movement/physiology , Cell Proliferation/physiology , Down-Regulation , Femoral Artery/surgery , Gene Knockdown Techniques , Hyperplasia/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Muscle, Smooth, Vascular/physiology , Mutation/genetics , Myocytes, Smooth Muscle , Proteomics , Proto-Oncogene Proteins c-sis/pharmacology , RNA, Small Interfering/physiology , Twist-Related Protein 1/antagonists & inhibitors
18.
Mol Cell ; 63(6): 1021-33, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27618486

ABSTRACT

Twist has been shown to cause treatment failure, cancer progression, and cancer-related death. However, strategies that directly target Twist are not yet conceivable. Here we reveal that K63-linked ubiquitination is a crucial regulatory mechanism for Twist activation. Through an E3 ligase screen and biochemical studies, we unexpectedly identified that RNF8 functions as a direct Twist activator by triggering K63-linked ubiquitination of Twist. RNF8-promoted Twist ubiquitination is required for Twist localization to the nucleus for subsequent EMT and CSC functions, thereby conferring chemoresistance. Our histological analyses showed that RNF8 expression is upregulated and correlated with disease progression, EMT features, and poor patient survival in breast cancer. Moreover, RNF8 regulates cancer cell migration and invasion and cancer metastasis, recapitulating the effect of Twist. Together, our findings reveal a previously unrecognized tumor-promoting function of RNF8 and provide evidence that targeting RNF8 is an appealing strategy to tackle tumor aggressiveness and treatment resistance.


Subject(s)
Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Damage , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Disease Progression , Epithelial-Mesenchymal Transition , Female , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Lysine/metabolism , MCF-7 Cells , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/metabolism , Ubiquitin-Protein Ligases , Ubiquitination
19.
Sci Rep ; 6: 21731, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26911838

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) signaling is constantly activated in human melanoma, and promotes melanoma metastasis. The dietary flavonoid apigenin is a bioactive compound that possesses low toxicity and exerts anti-metastatic activity in melanoma. However, the anti-metastasis mechanism of apigenin has not been fully elucidated. In the present study, we showed that apigenin suppressed murine melanoma B16F10 cell lung metastasis in mice, and inhibited cell migration and invasion in human and murine melanoma cells. Further study indicated that apigenin effectively suppressed STAT3 phosphorylation, decreased STAT3 nuclear localization and inhibited STAT3 transcriptional activity. Apigenin also down-regulated STAT3 target genes MMP-2, MMP-9, VEGF and Twist1, which are involved in cell migration and invasion. More importantly, overexpression of STAT3 or Twist1 partially reversed apigenin-impaired cell migration and invasion. Our data not only reveal a novel anti-metastasis mechanism of apigenin but also support the notion that STAT3 is an attractive and promising target for melanoma treatment.


Subject(s)
Apigenin/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Immunoblotting , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neoplasm Invasiveness/prevention & control , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
Oncotarget ; 7(8): 8931-43, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26840564

ABSTRACT

Inhibition of epithelial-mesenchymal transition (EMT)-inducing transcription factors Twist and Snail prevents tumor metastasis but enhances metastatic growth. Here, we report an unexpected role of a tumor suppressor tristetraprolin (TTP) in inhibiting Twist and Snail without enhancing cellular proliferation. TTP bound to the AU-rich element (ARE) within the mRNA 3'UTRs of Twist1 and Snail1, enhanced the decay of their mRNAs and inhibited the EMT of cancer cells. The ectopic expression of Twist1 or Snail1 without their 3'UTRs blocked the inhibitory effects of TTP on the EMT. We also observed that TTP overexpression suppressed the growth of cancer cells. Our data propose a new model whereby TTP down-regulates Twist1 and Snail1 and inhibits both the EMT and the proliferation of cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Snail Family Transcription Factors/antagonists & inhibitors , Tristetraprolin/pharmacology , Twist-Related Protein 1/antagonists & inhibitors , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Electrophoretic Mobility Shift Assay , Humans , Immunoprecipitation , Luciferases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors/genetics , Tumor Cells, Cultured , Twist-Related Protein 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...