Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Oncogene ; 42(36): 2659-2672, 2023 09.
Article in English | MEDLINE | ID: mdl-37516803

ABSTRACT

Melanoma is an aggressive form of skin cancer due to its high metastatic abilities and resistance to therapies. Melanoma cells reside in a heterogeneous tumour microenvironment that acts as a crucial regulator of its progression. Snail1 is an epithelial-to-mesenchymal transition transcription factor expressed during development and reactivated in pathological situations including fibrosis and cancer. In this work, we show that Snail1 is activated in the melanoma microenvironment, particularly in fibroblasts. Analysis of mouse models that allow stromal Snail1 depletion and therapeutic Snail1 blockade indicate that targeting Snail1 in the tumour microenvironment decreases melanoma growth and lung metastatic burden, extending mice survival. Transcriptomic analysis of melanoma-associated fibroblasts and analysis of the tumours indicate that stromal Snail1 induces melanoma growth by promoting an immunosuppressive microenvironment and a decrease in anti-tumour immunity. This study unveils a novel role of Snail1 in melanoma biology and supports its potential as a therapeutic target.


Subject(s)
Melanoma , Skin Neoplasms , Tumor Microenvironment , Animals , Mice , Epithelial-Mesenchymal Transition , Immunosuppression Therapy , Melanoma/genetics , Skin Neoplasms/genetics , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/immunology , Snail Family Transcription Factors/metabolism , Transcription Factors/genetics
2.
Biomed Pharmacother ; 141: 111870, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34246192

ABSTRACT

Caffeine is an active ingredient found in coffee and energy beverages. Its hepatoprotective effects against liver fibrosis are well-documented. Nonetheless, its renoprotective effects against renal fibrogenesis and epithelial-mesenchymal transition (EMT) processes remain unclear and under-investigated. In this study, the protective effects of caffeine against oxalate-induced EMT in renal tubular cells were evaluated by various assays to measure expression levels of epithelial and mesenchymal markers, cell migrating activity, level of oxidized proteins, and expression of Nrf2 and Snail1. Oxalate at sublethal dose significantly suppressed cell proliferation but increased cell elongation, spindle index and migration. Oxalate also decreased expression of epithelial markers (zonula occludens-1 (ZO-1) and E-cadherin) but increased expression of mesenchymal markers (fibronectin, vimentin and α-smooth muscle actin (α-SMA)). All of these EMT-inducing effects of oxalate could be prevented by pretreatment with caffeine. While oxalate increased oxidized proteins and Snail1 levels, it decreased Nrf2 expression. Caffeine could preserve all these molecules to their basal (control) levels. Finally, silencing of Nrf2 expression by small interfering RNA (siRNA) could abolish such protective effects of caffeine on oxalate-induced EMT. Our data indicate that the renoprotective effects of caffeine against oxalate-induced EMT is mediated, at least in part, by its anti-oxidative property through activation of Nrf2 signaling and suppression of Snail1 transcription factor.


Subject(s)
Antioxidants/pharmacology , Caffeine/pharmacology , Epithelial-Mesenchymal Transition/drug effects , NF-E2-Related Factor 2/metabolism , Oxalates/toxicity , Snail Family Transcription Factors/metabolism , Animals , Cell Movement/drug effects , Cell Movement/physiology , Dogs , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/physiology , Gene Knockdown Techniques , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Madin Darby Canine Kidney Cells , NF-E2-Related Factor 2/genetics , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics
3.
Nat Commun ; 12(1): 3932, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168145

ABSTRACT

Chemical descriptors encode the physicochemical and structural properties of small molecules, and they are at the core of chemoinformatics. The broad release of bioactivity data has prompted enriched representations of compounds, reaching beyond chemical structures and capturing their known biological properties. Unfortunately, bioactivity descriptors are not available for most small molecules, which limits their applicability to a few thousand well characterized compounds. Here we present a collection of deep neural networks able to infer bioactivity signatures for any compound of interest, even when little or no experimental information is available for them. Our signaturizers relate to bioactivities of 25 different types (including target profiles, cellular response and clinical outcomes) and can be used as drop-in replacements for chemical descriptors in day-to-day chemoinformatics tasks. Indeed, we illustrate how inferred bioactivity signatures are useful to navigate the chemical space in a biologically relevant manner, unveiling higher-order organization in natural product collections, and to enrich mostly uncharacterized chemical libraries for activity against the drug-orphan target Snail1. Moreover, we implement a battery of signature-activity relationship (SigAR) models and show a substantial improvement in performance, with respect to chemistry-based classifiers, across a series of biophysics and physiology activity prediction benchmarks.


Subject(s)
Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Cell Line, Tumor , Databases, Pharmaceutical , Drug Evaluation, Preclinical/methods , Humans , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
4.
Cell Biochem Funct ; 39(6): 813-820, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34128234

ABSTRACT

Prostate cancer (PCa) is the second most prevalent cancer in men worldwide. Most cases of death from PCa are due to metastasis. Early stages of metastasis are mediated by epithelial-mesenchymal transition (EMT) process through which cancer cells acquire motility and invasive characteristics. Thus, more potent and novel therapeutic strategies must be designed based on the inhibition of EMT or metastasis. Herein, we employ a co-culture system to evaluate the anti-EMT effects of human amniotic mesenchymal stromal cells (hAMSCs) on LNCaP PCa cells. The RNA of treated (sample) and untreated cancer cells (control) and whole-cell lysates of related cells were prepared and analysed through quantitative real-time polymerase chain reaction (qRT-PCR) and western blot, respectively. Based on the results, the expression of vimentin, Snail and Zeb1 in LNCaP cells decreased and the expression of E-cadherin increased after treatment with hAMSCs. Furthermore, induction of the cellular apoptosis in LNCaP cells was detected. The anti-cancer activity of conditioned medium from hAMSCs was shown using hanging drop technique (a 3D cell culture model). Our findings support the idea that stem cells can be considered as a novel therapeutic approach to inhibit prostate cancer cells. SIGNIFICANCE OF THE STUDY: The anti-tumour activity of hAMSCs on LNCaP prostate cancer cells using 2D and 3D cell culture models via induction of apoptosis, suppression of EMT process and down-regulation of EGFR was shown. The results of the present study support this idea that hAMSCs may be a potent therapeutic tool to suppress tumour growth in LNCaP prostate cancer cells.


Subject(s)
Apoptosis/drug effects , Culture Media, Conditioned/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Mesenchymal Stem Cells/drug effects , Snail Family Transcription Factors/antagonists & inhibitors , Vimentin/antagonists & inhibitors , Zinc Finger E-box-Binding Homeobox 1/antagonists & inhibitors , Coculture Techniques , Culture Media, Conditioned/chemistry , Down-Regulation/drug effects , Humans , Mesenchymal Stem Cells/metabolism , Snail Family Transcription Factors/metabolism , Tumor Cells, Cultured , Vimentin/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism
5.
J Ethnopharmacol ; 275: 114111, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33848610

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Imperata cylindrica (L.) Raeusch (Gramineae) is a medicinal spice traditionally used in the treatment of hypertension and cancer. AIM OF THE STUDY: To assess the anti-metastatic potential of the methanol extract of I. cylindrica roots and determined its mechanisms of action. MATERIAL AND METHODS: The growth inhibition activity of I. cylindrica root extract in vitro and in vivo in human cervical cancer. The scratch assay and Boyden Chamber assay were used to determine the anti-migrative and anti-invasion actions of the plant extract. The whole-genome gene expression profiling using RNA-Seq was performed to determine the differentially expressed genes in CaSki cells after exposure to I. cylindrica to identify its targeted genes related to metastasis. Using protein analysis (western blotting) and gene expression analysis (RTqPCR), the targeted pathways of the key genes that were initially identified with RNA-Seq, were evaluated. RESULTS: I. cylindrica extract showed dose-dependent cytotoxicity in vitro and in vivo in mice bearing tumors. Furthermore, I. cylindrica root extract significantly inhibited cell migration and cell invasion. After the genome-wide transcriptome analysis, we found that important genes involved in cancer progression and metastasis of cervical cancer, that is, CD24 and TIMP-4 were significantly downregulated and upregulated, respectively. Moreover, I. cylindrica root extract significantly inhibited the PI3/AKT/Snail signaling pathway and blocked the EMT of CaSki cells. CONCLUSION: These findings provide an anti-metastatic mechanism of action of I. cylindrica root extract toward the human cervical cancer suggesting that this plant maybe developed into selective chemotherapy.


Subject(s)
CD24 Antigen/genetics , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/pharmacology , Poaceae/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Snail Family Transcription Factors/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinases/genetics , Animals , Antigens, CD/metabolism , CD24 Antigen/antagonists & inhibitors , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Matrix Metalloproteinases/metabolism , Mice, SCID , Plant Extracts/therapeutic use , Plant Roots/chemistry , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinases/metabolism , Up-Regulation/drug effects , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-4
6.
Bioorg Chem ; 108: 104646, 2021 03.
Article in English | MEDLINE | ID: mdl-33484941

ABSTRACT

The transcriptional repressor Snail trriggers epithelial-mesenchymal transition (EMT), the process allowing cancer cells with invasive and metastasis properties. In this study, we screened medicinal plants for the Snail inhibitory active components by high content screen (HCS) and found that the crude extract of Xylopia vielana leaves showed potential activity. Subsequently, bioassay-guided isolation of the extract of Xylopia vielana was performed to obtain twenty-four dimeric guaianes (1-24), including 16 new analogues (1-5, 8-11, 13-15, 17, 18, 21, and 22). Their structures were elucidated by the comprehensive application of multiple spectroscopic methods. Compounds 1, 11, 12, and 16 were initially identified as the active compounds. Wound healing assay, transwell migration assay and western blot experiments verified that compounds 1 and 12 inhibited the expression of Snail in a concentration-dependent manner, and compound 12 was verified as a potent tumor migration inhibitory agent. This work showed a practical strategy for the discovery of new Snail inhibitors from natural products and provided potential insights for dimeric guaianes as anticancer lead compounds specifically targeting Snail protein.


Subject(s)
Plants, Medicinal/chemistry , Sesquiterpenes, Guaiane/pharmacology , Snail Family Transcription Factors/antagonists & inhibitors , Xylopia/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Plant Leaves/chemistry , Sesquiterpenes, Guaiane/chemistry , Sesquiterpenes, Guaiane/isolation & purification , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Int J Oncol ; 58(1): 100-110, 2021 01.
Article in English | MEDLINE | ID: mdl-33367934

ABSTRACT

Parthenolide has been demonstrated to have anticancer effects against various types of cancer. However, the functional role of parthenolid has yet to be clearly reported in renal cell carcinoma (RCC). The aim of the present study was to investigate the effect of parthenolide in RCC 786­O and ACHN cells. CCK­8 and colony­formation assays were used to observe the proliferation of RCC 786­O and ACHN cells. Migration and invasion abilities were assessed through Transwell assays. The stem cell­like properties of RCC cell lines were evaluated by mammosphere formation assay. Western blot analysis was used to investigate the metastasis and epithelial­mesenchymal transition (EMT) induced by parthenolide on the expression levels of MMP2, MMP9, E­cadherin, N­cadherin, vimentin and snail. The results revealed that when the cells were treated with various concentrations of parthenolide, the rate of proliferation and growth was decreased in 786­O and ACHN cells. The number of invasive cells in a field was approximately 170, 90, 40 and 190, 150, 70 in 786­O and ACHN cells with 0, 4 and 8 µM of parthenolide treatment. MMP­2/­9 expression (P<0.05) was inhibited by parthenolide. The protein levels of E­cadherin were increased (P<0.05) and N­cadherin, vimentin and snail were decreased (P<0.05) by parthenolide treatment. In addition, Parthenolide inhibited the expression of cancer stem cell markers and the PI3K/AKT pathway. The present study confirmed that parthenolide inhibited RCC cell proliferation and metastasis and suppressed the stem cell­like properties of RCC cell lines, which could be a potential strategy to treat RCC. However, further molecular mechanisms of parthenolide in RCC should be observed and reported in the future.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Sesquiterpenes/pharmacology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Self Renewal/drug effects , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Sesquiterpenes/therapeutic use , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/metabolism
8.
Aging (Albany NY) ; 13(2): 1947-1961, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33323539

ABSTRACT

Erythrocyte membrane protein band 4.1-like 3 (EPB41L3) is an important membrane skeletal protein that may interact with numerous membrane proteins. Loss of EPB41L3 is reported in multiple cancer types, and it is originally identified as a tumor suppressor. In this study, through analyzing expression profiling retrieved from the Gene Expression Omnibus (GEO) dataset, we find that EPB41L3 is upregulated in primary osteosarcoma (OS) and osteosarcoma cell lines. Importantly, EPB41L3 may promote osteosarcoma cell proliferation and suppress osteosarcoma cell migration and invasion. Reduced EPB41L3 leads to a decrease of E-cadherin as well as an increase of N-cadherin and Vimentin, implying a prominent epithelial-to-mesenchymal transition. Furthermore, we demonstrate that EPB41L3 inhibits the epithelial-to-mesenchymal transition through destabilizing the Snai1 protein, one of the most important transcription factors of the epithelial-to-mesenchymal transition process. Collectively, our study has first established the complex and vital roles of EPB41L3 and implicated EPB41L3 as a potential biomarker in osteosarcoma.


Subject(s)
Bone Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Microfilament Proteins/genetics , Osteosarcoma/genetics , Snail Family Transcription Factors/biosynthesis , Biomarkers/analysis , Bone Neoplasms/mortality , Cadherins/biosynthesis , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Osteosarcoma/mortality , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Tumor Stem Cell Assay , Vimentin/biosynthesis
9.
Oncol Rep ; 44(5): 2130-2142, 2020 11.
Article in English | MEDLINE | ID: mdl-32901893

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is the main subtype of esophageal cancer in China, and the prognosis of patients remains poor mainly due to the occurrence of lymph node and distant metastasis. The long non­coding RNA (lncRNA) maternally expressed gene 3 (MEG3) has been shown to have tumor­suppressive properties and to play an important role in epithelial­to­mesenchymal transition (EMT) in some solid tumors. However, whether MEG3 is involved in EMT in ESCC remains unclear. In the present study, the MEG3 expression level and its association with tumorigenesis were determined in 43 tumor tissues of patients with ESCC and in ESCC cells using reverse transcription­quantitative PCR analysis. Gene microarray analysis was performed to detect differentially expressed genes (DEGs). Based on the functional annotation results, the effects of ectopic expression of MEG3 on cell growth, migration, invasion and EMT were assessed. MEG3 expression level was found to be markedly lower in tumor tissues and cells. Statistical analysis revealed that MEG3 expression was significantly negatively associated with lymph node metastasis and TNM stage in ESCC. Fluorescence in situ hybridization assay demonstrated that MEG3 was expressed mainly in the nucleus. Ectopic expression of MEG3 inhibited cell proliferation, migration, invasion and cell cycle progression in EC109 cells. Gene microarray results demonstrated that 177 genes were differentially expressed ≥2.0 fold in MEG3­overexpressing cells, including 23 upregulated and 154 downregulated genes. Functional annotation revealed that the DEGs were mainly involved in amino acid biosynthetic process, mitogen­activated protein kinase signaling, and serine and glycine metabolism. Further experiments indicated that the ectopic expression of MEG3 significantly suppressed cell proliferation, migration, invasion and EMT by downregulating phosphoserine aminotransferase 1 (PSAT1). In pathological tissues, PSAT1 and MEG3 were significantly negatively correlated, and high expression of PSAT1 predicted poor survival. Taken together, these results suggest that MEG3 may be a useful prognostic biomarker and may suppress EMT by inhibiting the PSAT1­dependent glycogen synthase kinase­3ß/Snail signaling pathway in ESCC.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , RNA, Long Noncoding/metabolism , Snail Family Transcription Factors/antagonists & inhibitors , Transaminases/antagonists & inhibitors , Adult , Aged , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Down-Regulation , Epithelial-Mesenchymal Transition/physiology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Female , Glycogen Synthase Kinase 3 beta/metabolism , Heterografts , Humans , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , RNA, Long Noncoding/genetics , Signal Transduction , Snail Family Transcription Factors/metabolism , Survival Rate , Transaminases/metabolism
10.
J Med Chem ; 63(19): 10855-10878, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32886511

ABSTRACT

Capuramycin displays a narrow spectrum of antibacterial activity by targeting bacterial translocase I (MraY). In our program of development of new N-acetylglucosaminephosphotransferase1 (DPAGT1) inhibitors, we have identified that a capuramycin phenoxypiperidinylbenzylamide analogue (CPPB) inhibits DPAGT1 enzyme with an IC50 value of 200 nM. Despite a strong DPAGT1 inhibitory activity, CPPB does not show cytotoxicity against normal cells and a series of cancer cell lines. However, CPPB inhibits migrations of several solid cancers including pancreatic cancers that require high DPAGT1 expression in order for tumor progression. DPAGT1 inhibition by CPPB leads to a reduced expression level of Snail but does not reduce E-cadherin expression level at the IC50 (DPAGT1) concentration. CPPB displays a strong synergistic effect with paclitaxel against growth-inhibitory action of a patient-derived pancreatic adenocarcinoma, PD002: paclitaxel (IC50: 1.25 µM) inhibits growth of PD002 at 0.0024-0.16 µM in combination with 0.10-2.0 µM CPPB (IC50: 35 µM).


Subject(s)
Aminoglycosides/pharmacology , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Neoplasms/pathology , Aminoglycosides/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/chemistry , Humans , Paclitaxel/pharmacology , Snail Family Transcription Factors/antagonists & inhibitors , Structure-Activity Relationship
11.
Eur Rev Med Pharmacol Sci ; 24(3): 1041-1053, 2020 02.
Article in English | MEDLINE | ID: mdl-32096174

ABSTRACT

OBJECTIVE: Ovarian cancer is a highly invasive type of cancer. A previous study demonstrated that E-cadherin expression was upregulated in a human ovarian cancer cell line with a high expression of WW domain-containing oxidoreductase (WWOX), which is a tumor suppressor. Also, the migration and invasion ability of these cells was reduced. Snail family members are involved in the epithelial-to-mesenchymal transition (EMT) of ovarian cancer cells, and the expression of Snail family members is regulated by the transcription factor Elf5. The aim of the present research was to elucidate the role of WWOX in EMT of ovarian carcinoma cells through the Elf5/Snail pathway by gain and loss of function approaches in in vitro experiments. MATERIALS AND METHODS: First, a WWOX gene expressing plasmid was transfected into CD133+CD117+ HO8910 ovarian carcinoma cells, and an Elf5 shRNA plasmid was transfected into these cells to assess the changes in EMT-related factors, including Snail1, and the invasive ability of tumor cells ability. Second, the human ovarian carcinoma cell lines HO8910 and SKOV3 were divided into six groups to detect the same indicators. RESULTS: The results demonstrated that the high expression of WWOX resulted in an increased E-cadherin expression, decreased Snail1 activity, and decreased invasion ability in CD133+CD117+ HO8910 cells. Elf5 shRNA transfection did not affect the WWOX expression; however, it decreased the expression of E-cadherin and Elf5 activity, while increasing Snail1 activity and invasion ability in CD133+CD117+ HO8910 cells. It was also observed that WWOX overexpression in HO8910 and SKOV3 cells inhibited the expression of EMT-related proteins and inhibited cell migration and invasion. CONCLUSIONS: Taken together, the results of the present report suggest that WWOX can decrease Snail1 activity by enhancing the activity of Elf5, thus upregulating E-cadherin expression and eventually inhibiting EMT of ovarian carcinoma.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , DNA-Binding Proteins/biosynthesis , Epithelial-Mesenchymal Transition/physiology , Ovarian Neoplasms/metabolism , Snail Family Transcription Factors/biosynthesis , Transcription Factors/biosynthesis , Tumor Suppressor Proteins/biosynthesis , WW Domain-Containing Oxidoreductase/biosynthesis , Carcinoma, Ovarian Epithelial/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Ovarian Neoplasms/genetics , Signal Transduction/physiology , Snail Family Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , WW Domain-Containing Oxidoreductase/genetics
12.
Life Sci ; 248: 117454, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32088211

ABSTRACT

AIMS: Dihydroartemisinin (DHA) is currently considered as the promising cancer therapeutic drug. In this study, we aimed to investigate the anti-proliferative and anti-metastasis effects of DHA. MAIN METHODS: Utilizing breast cancer cells MCF-7, MDA-MB-231 and BT549, cell proliferation, migration and invasion were detected. RT-qPCR was performed to detect CIZ1, TGF-ß1 and Snail expression, and the interactions of these related molecules were analyzed by GeneMANIA database. Western blot detected CIZ1, TGF-ß1/Smads signaling and Snail expression in DHA-treated cells, in TGFß1-induced cells with enhanced metastatic capacity, and in cells treated with DHA plus TGFß1/TGFß1 inhibitor SD-208. KEY FINDINGS: Results indicated DHA inhibited breast cancer cell proliferation and migration, with more potent effects compared with that of artemisinin. RT-qPCR and Western blot showed DHA inhibited CIZ1, TGF-ß1 and Snail expression, and these molecules were shown to have protein-protein interactions by bioinformatics. Furthermore, TGFß1-treatment enhanced MCF-7 migration and invasion, and CIZ1, TGF-ß1/Smads signaling and snail activities; DHA, SD-208, combination of DHA and SD-208 reversed these conditions, preliminarily proving the cascade regulation between TGF-ß1 signaling and CIZ1. MCF-7 xenografts model demonstrated the inhibition of DHA on tumor burden, and its mechanisms and well-tolerance in vivo; combination of DHA and SD-208 tried by us for the first time showed better treatment effects, but possible liver impairment made its use still keep cautious. SIGNIFICANCE: DHA treatment inhibits the proliferation and metastasis of breast cancer, through suppressing TGF-ß1/Smad signaling and CIZ1, suggesting the promising potential of DHA as a well-tolerated antitumor TGF-ß1 pathway inhibitor.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Artemisinins/pharmacology , Breast Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Nuclear Proteins/genetics , Transforming Growth Factor beta1/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Epithelial-Mesenchymal Transition , Female , Humans , Lymphatic Metastasis , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Pteridines/pharmacology , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Cancer Med ; 9(4): 1307-1320, 2020 02.
Article in English | MEDLINE | ID: mdl-31863638

ABSTRACT

Ovarian cancer is the most lethal gynecological malignancy. Currently, new chemotherapeutic strategies are required to improve patient outcome and survival. Biguanides, classic anti-diabetic drugs, have gained importance for theiri antitumor potency demonstrated by various studies. Olaparib is a PARP inhibitor approved for maintenance therapy following platinum-based chemotherapy. Furthermore, Snai1, a transcription factor that works as a master regulator of the epithelial/mesenchymal transition process (EMT) is involved in ovarian cancer resistance and progression. Here we aimed to demonstrate the possible cross talk between biguanides and Snail in response to olaparib combination therapy. In this study, we have shown that while in A2780CR cells biguanides reduced cell survival (single treatments ~20%; combined treatment ~44%) and cell migration (single treatments ~45%; biguanide-olaparib ~80%) significantly, A2780PAR exhibited superior efficacy with single (~60%) and combined treatments (~80%). Moreover, our results indicate that knock-down of Snail further enhances the attenuation of migration, inhibits EMT related-proteins (~90%) and induces a synergistic effect in biguanide-olaparib treatment. Altogether, this work suggests a novel treatment strategy against drug-resistant or recurrent ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinogenesis/drug effects , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Snail Family Transcription Factors/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biguanides/pharmacology , Biguanides/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Knockdown Techniques , Humans , Neoplasm Recurrence, Local/pathology , Ovarian Neoplasms/pathology , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Snail Family Transcription Factors/genetics
14.
Biochem Biophys Res Commun ; 521(1): 152-157, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31653342

ABSTRACT

Epithelial-mesenchymal transition (EMT) is associated with cancer malignancies such as invasion, metastasis, and drug resistance. In this study, HCT116 human colorectal cancer cells were transduced with SLUG or SNAIL retroviruses, and EMT cells with mesenchymal morphology were established. The EMT cells showed a high invasive activity and resistance to several anticancer agents such as methotrexate, SN-38, and cisplatin. Furthermore, they contained about 1-10% side population (SP) cells that were not stained by Hoechst 33342. This SP phenotype was not stable; the isolated SP cells generated both SP and non-SP cells, suggesting a potential for differentiation. Gene expression analysis of SP cells suggested the alteration of genes that are involved in epigenetic changes. Therefore, we examined the effect of 74 epigenetic inhibitors, and found that two inhibitors, namely I-BET151 and bromosporine, targeting the bromodomain and extra-terminal motif (BET) proteins, decreased the ratio of SP cells to <50% compared with the control, without affecting the immediate efflux of Hoechst 33342 by transporters. In addition, compared with the parental cells, the EMT cells showed a higher sensitivity to I-BET151 and bromosporine. This study suggests that EMT development and SP phenotype can be independent events but both are regulated by BET inhibitors in SLUG- or SNAIL-transducted HCT116 cells.


Subject(s)
Colorectal Neoplasms/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacology , Proteins/antagonists & inhibitors , Snail Family Transcription Factors/antagonists & inhibitors , Cell Differentiation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , HCT116 Cells , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Phenotype , Proteins/metabolism , Snail Family Transcription Factors/metabolism
15.
Cell Death Dis ; 10(5): 373, 2019 05 09.
Article in English | MEDLINE | ID: mdl-31073122

ABSTRACT

Increasing evidence suggest that lncRNAs (long noncoding RNAs) play important roles in human cancer. Breast cancer is a heterogeneous disease and the potential involvement of lncRNAs in breast cancer remains unexplored. In this study, we characterized a novel lncRNA, RP1-5O6.5 (termed as RP1). We found that RP1 was highly expressed in breast cancer and predicted poor prognosis of breast cancer patients. Gain-of-function and loss-of-function assays showed that RP1 promoted the proliferation and metastasis of breast cancer cells in vitro and in vivo. Mechanistically, RP1 maintained the EMT and stemness states of breast cancer cells via repressing p27kip1 protein expression. RP1 combined with the complex p-4E-BP1/eIF4E to prevent eIF4E from interacting with eIF4G, therefore attenuating the translational efficiency of p27kip1 mRNA. Furthermore, we found that p27kip1 evidently downregulated Snail1 but not ZEB1 to inhibit invasion of breast cancer cells. Kruppel-like factor 5 (KLF5) was positively correlated with RP1 in breast cancer tissues. Moreover, we demonstrated that KLF5 recruited p300 to the RP1 promoter to enhance RP1 expression. Taken together, our findings demonstrated that KLF5-regulated RP1 plays an oncogenic role in breast cancer by suppressing p27kip1, providing support for the clinical investigation of therapeutic approaches focusing on RP1.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Kruppel-Like Transcription Factors/metabolism , RNA, Long Noncoding/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cyclin-Dependent Kinase Inhibitor p27/genetics , Eukaryotic Initiation Factor-4E/metabolism , Female , Humans , Mice , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Up-Regulation , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , p300-CBP Transcription Factors/metabolism
16.
Sci Adv ; 5(5): eaav3235, 2019 05.
Article in English | MEDLINE | ID: mdl-31086816

ABSTRACT

Resistance to platinum-based chemotherapy is a common event in patients with cancer, generally associated with tumor dissemination and metastasis. Whether platinum treatment per se activates molecular pathways linked to tumor spreading is not known. Here, we report that the ubiquitin-specific protease 1 (USP1) mediates ovarian cancer cell resistance to platinum, by regulating the stability of Snail, which, in turn, promotes tumor dissemination. At the molecular level, we observed that upon platinum treatment, USP1 is phosphorylated by ATM and ATR and binds to Snail. Then, USP1 de-ubiquitinates and stabilizes Snail expression, conferring resistance to platinum, increased stem cell-like features, and metastatic ability. Consistently, knockout or pharmacological inhibition of USP1 increased platinum sensitivity and decreased metastatic dissemination in a Snail-dependent manner. Our findings identify Snail as a USP1 target and open the way to a novel strategy to overcome platinum resistance and more successfully treat patients with ovarian cancer.


Subject(s)
Apoptosis/drug effects , Coordination Complexes/pharmacology , Platinum/chemistry , Snail Family Transcription Factors/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Coordination Complexes/therapeutic use , Drug Resistance, Neoplasm , Female , Gene Editing , Humans , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Xenograft Model Antitumor Assays
17.
Biosci Rep ; 39(5)2019 05 31.
Article in English | MEDLINE | ID: mdl-30975732

ABSTRACT

Snail1 plays an important role in epithelial to mesenchymal transition (EMT) during tumor metastasis; however, whether Snai1 potentiates the process of neoangiogenesis is completely unknown. In the present study, tube formation assay was used to evaluate neoangiogenesis in vitro The expression of Snai1 and other pro-neoangiogenic factors was measured by quantitative real time PCR. Tumor derived endothelial cells (TDECs) were stimulated with fibroblast growth factor 1 (FGF1) or VEGF and formed more tubes compared with untreated, whereas cells treated with Sulforaphane had less tube formation. Silencing SNAI1 significantly attenuated tube formation accompanied by decreased CD31, CD34, and VWF expression in TDECs compared with control. In contrast, overexpression of Snai1 led to more CD31, CD34, and VWF expression and tube formation. To determine if the observed effects of SNAI1 on tube formation was a global phenomenon, the same assay was conducted in normal mesenchymal stem cells (MSCs). SNAI1 silencing did not have any effect on tube formation in MSCs. The expression of TIMP2, ENG, and HIF1A was up-regulated 3-fold or higher after silencing SNAI1, and ID1, VEGFA, PLG, LECT1, HPSE were shown down-regulated. Taken together, our study elucidates an important role of EMT inducer Snai1 in regulating tumor neoangiogenesis, suggesting a potential therapeutic target for overcoming tumor EMT.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Neovascularization, Pathologic/genetics , Snail Family Transcription Factors/genetics , Anticarcinogenic Agents/pharmacology , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Endoglin/genetics , Endoglin/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Fibroblast Growth Factor 1/pharmacology , Glucuronidase/genetics , Glucuronidase/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Isothiocyanates/pharmacology , Lung/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/metabolism , Sulfoxides , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
18.
J Cell Biochem ; 120(6): 10342-10350, 2019 06.
Article in English | MEDLINE | ID: mdl-30582213

ABSTRACT

Chlorogenic acid (CA) is a polyphenol compound that possesses anticancer effects on several types of tumors. However, there are few previous studies concerning the protective effects of CA on osteosarcoma. The current study aimed to examine the toxicity of CA to osteosarcoma cells and to explore the potential mechanisms. Cell growth was evaluated using cell counting kit-8 assay and Western blot analysis of proliferating cell nuclear antigen (PCNA). Apoptosis was assessed by flow cytometry analysis using flow cytometry and caspase-3/7 activity assay. The expression changes of the signal transducer and activator of transcription 3 (STAT3)/Snail pathway were detected by Western blot analysis. We found that CA dose-dependently inhibited cell viability and PCNA expression in osteosarcoma cells. Meanwhile, CA treatment increased the apoptotic rate and caspase-3/7 activity in osteosarcoma cells in a concentration-dependent manner. We found that CA concentration-dependently inhibited the activation of the STAT3/Snail pathway in osteosarcoma cells. Inhibition of the STAT3/Snail pathway by si-STAT3 retarded the growth and induced apoptosis of osteosarcoma cells. Mechanistically, activation of the STAT3/Snail pathway by pcDNA-STAT3 reversed the effects of CA on osteosarcoma cell growth and apoptosis. In conclusion, CA inhibited osteosarcoma carcinogenesis by suppressing osteosarcoma cell growth and inducing apoptosis, which was involved in inactivation of the STAT3/Snail pathway. Therefore, our study suggested that CA might have good therapy prospects in osteosarcoma therapy.


Subject(s)
Bone Neoplasms/drug therapy , Carcinogenesis/drug effects , Chlorogenic Acid/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Osteosarcoma/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Snail Family Transcription Factors/antagonists & inhibitors , Apoptosis , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Tumor Cells, Cultured
19.
PLoS One ; 13(9): e0202747, 2018.
Article in English | MEDLINE | ID: mdl-30208064

ABSTRACT

Snail2 is a zinc-finger transcription factor best known to repress expression of genes encoding cell adherence proteins to facilitate induction of the epithelial-to-mesenchymal transition. While this role has been best documented in the developmental migration of the neural crest and mesoderm, here we expand on previously reported preliminary findings that morpholino knock-down of snai2 impairs the generation of hematopoietic stem cells (HSCs) during zebrafish development. We demonstrate that snai2 morphants fail to initiate HSC specification and show defects in the somitic niche of migrating HSC precursors. These defects include a reduction in sclerotome markers as well as in the Notch ligands dlc and dld, which are known to be essential components of HSC specification. Accordingly, enforced expression of the Notch1-intracellular domain was capable of rescuing HSC specification in snai2 morphants. To parallel our approach, we obtained two mutant alleles of snai2. In contrast to the morphants, homozygous mutant embryos displayed no defects in HSC specification or in sclerotome development, and mutant fish survive into adulthood. However, when these homozygous mutants were injected with snai2 morpholino, HSCs were improperly specified. In summary, our morpholino data support a role for Snai2 in HSC development, whereas our mutant data suggest that Snai2 is dispensable for this process. Together, these findings further support the need for careful consideration of both morpholino and mutant phenotypes in studies of gene function.


Subject(s)
Snail Family Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Embryo, Nonmammalian/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Genotype , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Morpholinos/metabolism , Mutagenesis, Site-Directed , PAX9 Transcription Factor/metabolism , Phenotype , Receptors, Notch/metabolism , Signal Transduction , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
20.
Chem Biol Interact ; 296: 134-144, 2018 Dec 25.
Article in English | MEDLINE | ID: mdl-30266538

ABSTRACT

Dictamnine (DTM) is a natural alkaloid isolated from the root of Dictamnus dasycarpus Turcz and has been shown to exhibit multiple biological functions, including anti-inflammatory, antifungal, anti-angiogenic and anticancer activity. However, the mechanisms by which dictamnine inhibits tumor growth are not fully understood. In this study, we investigated the effectiveness of dictamnine as a treatment for cancer and to identify the underlying mechanisms of its anticancer activity. Here, dictamnine showed the potent inhibitory activity against HIF-1α and Slug activation induced by hypoxia in various human cancer cell lines. This compound markedly decreased the hypoxia-induced accumulation of HIF-1α and Slug protein in a dose-dependent manner. Further analysis revealed that dictamnine inhibited HIF-1α protein synthesis, without affecting its degradation. Our results demonstrated that dictamnine reduced HIF-1α protein synthesis by downregulating the mTOR/p70S6K/eIF4E and MAPK pathways, and reduced the expression of Slug by inhibiting the GSK-3ß/Slug signaling pathway. Moreover, epithelial-mesenchymal transition (EMT) was inhibited in dictamnine-treated tumors by downregulation of HIF-1α and Slug, as reflected by the upregulation of E-cadherin and Occludin, and the downregulation of N-cadherin and Vimentin. Phenomenological experiments showed that dictamnine reduced migration and invasion, inhibited HCT116 cell proliferation and promoted HCT116 cell apoptosis by downregulating HIF-1α and Slug. In vivo studies further confirmed that dictamnine treatment caused significant inhibition of tumor growth in a xenograft tumor model. These findings suggest that dictamnine is a potent cancer inhibitor, providing a rationale for anticancer pathway-targeted therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Neoplasms/drug therapy , Quinolines/pharmacology , Snail Family Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Cell Proliferation/drug effects , Dictamnus/chemistry , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Plant Roots/chemistry , Quinolines/chemistry , Quinolines/isolation & purification , Signal Transduction/drug effects , Snail Family Transcription Factors/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...