Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Chinese Medical Journal ; (24): 1098-1110, 2023.
Article in English | WPRIM | ID: wpr-980838

ABSTRACT

BACKGROUND@#Ovarian cancer is one of the most widespread malignant diseases of the female reproductive system worldwide. The plurality of ovarian cancer is diagnosed with metastasis in the abdominal cavity. Epithelial-mesenchymal transition (EMT) exerts a vital role in tumor cell metastasis. However, it remains unclear whether long non-coding RNA (lncRNA) are implicated in EMT and influence ovarian cancer cell invasion and metastasis. This study was designed to investigate the impacts of lncRNA AC005224.4 on ovarian cancer.@*METHODS@#LncRNA AC005224.4, miR-140-3p, and snail family transcriptional repressor 2 ( SNAI2 ) expression levels in ovarian cancer and normal ovarian tissues were determined using real-time quantitative polymerase chain reaction (qRT-PCR). Cell Counting Kit-8 (CCK-8) and Transwell (migration and invasion) assays were conducted to measure SKOV3 and CAOV-3 cell proliferation and metastasis. E-cadherin, N-cadherin, Snail, and Vimentin contents were detected using Western blot. Nude mouse xenograft assay was utilized to validate AC005224.4 effects in vivo . Dual-luciferase reporter gene assay confirmed the targeted relationship between miR-140-3p and AC005224.4 or SNAI2 .@*RESULTS@#AC005224.4 and SNAI2 upregulation and miR-140-3p downregulation were observed in ovarian cancer tissues and cells. Silencing of AC005224.4 observably moderated SKOV3 and CAOV-3 cell proliferation, migration, invasion, and EMT process in vitro and impaired the tumorigenesis in vivo . miR-140-3p was a target of AC005224.4 and its reduced expression level was mediated by AC005224.4. miR-140-3p mimics decreased the proliferation, migration, and invasion of ovarian cancer cells. SNAI2 was identified as a novel target of miR-140-3p and its expression level was promoted by either AC005224.4 overexpression or miR-140-3p knockdown. Overexpression of SNAI2 also facilitated ovarian cancer cell viability and metastasis.@*CONCLUSION@#AC005224.4 was confirmed as an oncogene via sponging miR-140-3p and promoted SNAI2 expression, contributing to better understanding of ovarian cancer pathogenesis and shedding light on exploiting the novel lncRNA-directed therapy against ovarian cancer.


Subject(s)
Animals , Mice , Humans , Female , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Ovarian Neoplasms/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Snail Family Transcription Factors/metabolism
2.
Int. j. med. surg. sci. (Print) ; 9(2): 1-9, June 2022. ilus, graf
Article in English | LILACS | ID: biblio-1512600

ABSTRACT

Cisplatin, the first platinum compound approved for cancer treatment, is widely used in the treatment of various cancers including hepatocellular carcinoma (HCC). HCC incidence rates rise globally. Epithelial mesenchymal transition (EMT) is implicated in cancer invasion and metastasis, which are associated with increased mortality. Cisplatin dose might influence cancer invasion and metastatic behavior of the cells. The aim of the study was to investigate the effect of low-dose cisplatin treatment on EMT- related changes in HepG2 cells. Following treatment with 4 µM cisplatin, HepG2 cells were evaluated morphologically. Gene expression of E-cadherin, Vimentin, Snail1 was assessed by quantitative PCR. Immunofluorescence analyses of NA-K ATPase were performed. Although the low-dose cisplatin treated cells exhibited a more stretched morphology, no statistical difference was detected in gene expression of E-cadherin, Vimentin, Snail1 and immunofluorescence of NA-K ATPase. Findings on low-dose cisplatin effects in HepG2 might contribute to the knowledge of antineoplastic inefficacy by further understanding the molecular mechanisms of drug action.


El cisplatino, el primer compuesto de platino aprobado para el tratamiento del cáncer, es ampliamente utilizado en el tratamiento de varios tipos de cáncer, incluido el carcinoma hepatocelular (CHC). Las tasas de incidencia de CHC aumentan a nivel mundial. La transición mesenquimal epitelial (EMT) está implicada en la invasión del cáncer y la metástasis, que se asocian con un aumento de la mortalidad. La dosis de cisplatino podría influir en la invasión del cáncer y el comportamiento metastásico de las células. El objetivo del estudio fue investigar el efecto del tratamiento con dosis bajas de cisplatino en los cambios relacionados con la EMT en las células HepG2. Tras el tratamiento con cisplatino de 4 µM, se evaluaron morfológicamente las células HepG2. La expresión génica de E-cadherina, vimentina, caracol1 se evaluó mediante PCR cuantitativa. Se realizaron análisis de inmunofluorescencia de NA-K ATPasa . Aunque las células tratadas con cisplatino en dosis bajas exhibieron una morfología más estirada, no se detectaron diferencias estadísticas en la expresión génica de E-cadherina, vimentina, Snail1 e inmunofluorescencia de NA-K ATPasa. Los hallazgos sobre los efectos del cisplatino en dosis bajas en HepG2 podrían contribuir al conocimiento de la ineficacia antineoplásica al comprender mejor los mecanismos moleculares de la acción del fármaco.


Subject(s)
Humans , Cisplatin/administration & dosage , Antineoplastic Agents/administration & dosage , Vimentin/drug effects , Vimentin/genetics , Vimentin/metabolism , Cadherins/drug effects , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Fluorescent Antibody Technique , Microscopy, Confocal , Hep G2 Cells , Epithelial-Mesenchymal Transition , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors/drug effects , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Neoplasm Invasiveness
3.
Chinese Journal of Applied Physiology ; (6): 101-106, 2019.
Article in Chinese | WPRIM | ID: wpr-776551

ABSTRACT

OBJECTIVE@#To investigate the effects of Yiqi Huayu Hutan decoction on pulmonary fibrosis of rats which induced by bleomycin.@*METHODS@#The rat model of pulmonary fibrosis was induced by intratracheal injection of bleomycin hydrochloride (5 mg/kg). Sixty SD rats were randomly divided into the normal group (group N), the model group (group M), the positive control group (group Y), group of low concentration (group LC), group of medium concentration (group MC) and group of high concentration of Yiqi Huayu Hutan decoction (group HC). After 4 weeks, the experimental groups were treated with low concentration decoction, medium concentration decoction and high concentration decoction respectively, and the Y group was treated with hydrocortisone acetate, the Group N and group M were treated with saline by intragastric administration. Twelve weeks later, rats were killed and the pathomorphism of pulmonary tissues of each group was observed by HE staining and Masson staining. Further, the expressions of transforming growth factor-β1(TGF-β1), Snail1, E-cadherin and Fibronectin in pulmonary tissues of each group were detected by qTR-PCR and Western blot.@*RESULTS@#Compared with the model group, the collagen sediment in the interstitial was reduced in the experimental groups, especially in the group of medium concentration, which was observed by HE staining and Masson staining .Compared with the model group, the expressions of TGF-β1, Snail1 and Fibronectin protein in pulmonary tissues of the treatment groups were decreased in the experimental group, especially in the group of medium concentration, which were detected by qRT-PCR and Western blot.@*CONCLUSION@#Yiqi Huayu Hutan decoction can significantly improve the pulmonary fibrosis which is induced by bleomycin, and the mechanism is related to the inhibition of the expression of TGF-β/Snail pathway of transcription TGF-β1.


Subject(s)
Animals , Rats , Bleomycin , Cadherins , Metabolism , Drugs, Chinese Herbal , Pharmacology , Fibronectins , Metabolism , Idiopathic Pulmonary Fibrosis , Drug Therapy , Lung , Metabolism , Pathology , Random Allocation , Rats, Sprague-Dawley , Snail Family Transcription Factors , Metabolism , Transforming Growth Factor beta1 , Metabolism
4.
Journal of Southern Medical University ; (12): 1325-1330, 2018.
Article in Chinese | WPRIM | ID: wpr-771473

ABSTRACT

OBJECTIVE@#To explore the role of SIRT1 in the occurrence of epithelial-mesenchymal transition (EMT) in EC-9706 and Eca-109 cells and the possible mechanism.@*METHODS@#Three chemically synthesized siRNA targeting SIRT1 were transfected into EC-9706 and Eca-109 cells with the non-transfected cells and cells transfected with the negative siRNAs as controls. Real-time PCR and Western blotting were used to detect the expressions of SIRT1, E-cadherin, vimentin, Snail, Twist1 and ZEB in the cells. Transwell invasion assay and wounding healing assay were used to examine the changes in the invasion and metastasis abilities of the cells after transfection.@*RESULTS@#EC-9706 and Eca-109 cells transfected with SIRT1 siRNA1 and SIRT1 siRNA3 showed significantly decreased mRNA and protein expressions of SIRT1 ( < 0.05). Transwell invasion assay and wounding healing assay showed that transfection with SIRT1 siRNA1 and SIRT1 siRNA3 caused significantly lowered invasion and metastasis abilities in EC-9706 and Eca-109 cells ( < 0.05). In EC-9706 and Eca-109 cells transfected with SIRT1 siRNA1 and SIRT1 siRNA3, the expression level of E-cadherin was significantly increased while the expressions of vimentin, Snail and Twist were significantly lowered ( < 0.05).@*CONCLUSIONS@#SIRT1 participates in the invasion and metastasis of EC-9706 and Eca- 109 cells probably by inducing EMT via regulating the expression of Snail.


Subject(s)
Humans , Antigens, CD , Metabolism , Cadherins , Metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Physiology , Neoplasm Invasiveness , Nuclear Proteins , Metabolism , RNA, Messenger , Metabolism , RNA, Small Interfering , Metabolism , Sirtuin 1 , Genetics , Metabolism , Snail Family Transcription Factors , Metabolism , Transfection , Twist-Related Protein 1 , Metabolism , Vimentin , Metabolism , Zinc Finger E-box-Binding Homeobox 1 , Metabolism
5.
Braz. j. med. biol. res ; 51(2): e6808, 2018. tab, graf
Article in English | LILACS | ID: biblio-889020

ABSTRACT

Pituitary adenomas account for 10-15% of primary intracranial tumors. Growth hormone (GH)-secreting adenomas account for 13% of all pituitary adenomas and cause acromegaly. These tumors can be aggressive, invade surrounding structures and are highly recurrent. The objective of this study was to evaluate E-cadherin, Slug and neural cell adhesion molecule (NCAM) expression in GH-secreting pituitary adenomas and its relationship to tumor invasiveness. A cross-sectional study of patients who underwent hypophysectomy due to GH-secreting pituitary adenoma from April 2007 to December 2014 was carried out. The medical records were reviewed to collect clinical data. Immediately after surgery, tumor samples were frozen in liquid nitrogen and stored in a biofreezer at -80°C for assessment of E-cadherin 1 (CDH1), SLUG (SNAI2), and NCAM (NCAM1) by real-time PCR. The samples were fixed in formalin and embedded in paraffin for immunohistochemical analysis of E-cadherin and NCAM. Thirty-five patients with acromegaly were included in the study. Of these, 65.7% had invasive tumors. Immunohistochemically, E-cadherin was expressed in 96.7% of patients, and NCAM in 80% of patients. There was no statistically significant relationship between tumor grade or invasiveness and immunohistochemical expression of these markers. Regarding gene expression, 50% of cases expressed CDH1, none expressed SNAI2, and 53.3% expressed NCAM1. There was no statistically significant relationship between tumor grade or invasiveness and gene expression of CDH1, SNAI2, and NCAM1. The absence of Slug overexpression and of E-cadherin and NCAM suppression suggests that expression of these markers is not associated with tumor invasiveness in GH-secreting pituitary adenomas.


Subject(s)
Humans , Male , Female , Adolescent , Adult , Middle Aged , Aged , Young Adult , Acromegaly/pathology , Adenoma/pathology , Cadherins/analysis , Neural Cell Adhesion Molecules/analysis , Snail Family Transcription Factors/analysis , Acromegaly/genetics , Acromegaly/metabolism , Immunohistochemistry , Biomarkers, Tumor/analysis , Adenoma/genetics , Adenoma/chemistry , Gene Expression , Cross-Sectional Studies , Neoplasm Grading
6.
Journal of Southern Medical University ; (12): 802-806, 2016.
Article in Chinese | WPRIM | ID: wpr-286895

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of small interfering RNA (siRNA)-mediated silencing of PC4 and SFRS1 interacting protein 1 (PSIP1) on invasion and migration of human glioma U87 cells.</p><p><b>METHODS</b>Chemically synthesized siRNA targeting PSIP1 gene was transfected into U87 cells via lipofectamine, and the gene silencing effect was determined using real-time PCR. The changes in the invasion and migration abilities of the transfected cells were assessed with Transwell assay and wound healing assay, respectively. Western blotting was used to analyze the expression of N-cadherin, β-catenin and the transcription factor Slug.</p><p><b>RESULTS</b>The mRNA and protein level of PSIP1 was significantly reduced in U87 cells after transfection with PSIP1 siRNA (P<0.0001). PSIP1 knockdown in U87 cells resulted in significant suppression of cell invasion and migration abilities (P<0.01) and also reduced N-cadherin, β-catenin and Slug expressions.</p><p><b>CONCLUSION</b>s Silencing of PSIP1 impairs the invasion and migration abilities of glioma cells and lowers the expressions of N-cadherin, β-catenin and Slug, suggesting that PSIP1 may regulate Slug by classical Wnt/β-catenin signaling pathway to modulate epithelial-mesenchymal transition and promote the invasion and migration of glioma cells.</p>


Subject(s)
Humans , Adaptor Proteins, Signal Transducing , Genetics , Metabolism , Antigens, CD , Metabolism , Cadherins , Metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Glioma , Pathology , Neoplasm Invasiveness , RNA Interference , RNA, Messenger , Genetics , Metabolism , RNA, Small Interfering , Genetics , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors , Genetics , Metabolism , Transfection , Wnt Signaling Pathway , beta Catenin , Metabolism
7.
Journal of Southern Medical University ; (12): 532-537, 2016.
Article in Chinese | WPRIM | ID: wpr-264009

ABSTRACT

<p><b>OBJECTIVE</b>To identify potential markers for predicting invasion, metastasis, and prognosis of gastric adenocarcinoma (GAC).</p><p><b>METHODS</b>The expressions of Slug, ZEB1 and KISS-1 were detected immunohistochemically in 261 GAC tissues and 80 normal gastric tissues.</p><p><b>RESULTS</b>The positivity rates of Slug, ZEB1, and KISS-1 in gastric tissues were 2.5%, 1.3%, and 87.5%, respectively, significantly different from the rates of 62.1%, 28.4%, and 41.1% in GAC tissues (P<0.05). The expression level of Slug was significantly correlated with the depth of invasion, lymph node metastasis, and pTNM stages; the positivity rates of both ZEB1 and KISS-1 were significantly correlated with the tumor grade, depth of invasion, lymph node metastasis and pTNM stages. Slug expression was positively correlated with ZEB1 expression, and KISS-1 expression was inversely correlated with Slug and ZEB1 expressions. Kaplan-Meier analysis showed that the overall survival time of patients with positive expressions of Slug and ZEB1 was significantly shorter than that of the negative patients, and the survival time of patients positive for KISS-1 was significantly longer than the negative patients. COX multivariate analysis showed that positive Slug, ZEB1 and KISS-1 protein expressions and pTNM stages were independent prognostic factors of GAC (P<0.05).</p><p><b>CONCLUSION</b>The abnormal expressions of Slug, ZEB1 and KISS-1 may contribute to the tumorigenesis of GAC and are related with lymph node metastasis, pTNM stages, and prognosis of GAC. The combined detection of Slug, ZEB1, and KISS-1 expression has an important value in predicting the progression and prognosis of GAC.</p>


Subject(s)
Humans , Adenocarcinoma , Metabolism , Pathology , Disease Progression , Homeodomain Proteins , Metabolism , Immunohistochemistry , Kaplan-Meier Estimate , Kisspeptins , Metabolism , Lymphatic Metastasis , Neoplasm Grading , Prognosis , Proportional Hazards Models , Snail Family Transcription Factors , Stomach Neoplasms , Metabolism , Pathology , Transcription Factors , Metabolism , Zinc Finger E-box-Binding Homeobox 1
8.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 839-845, 2016.
Article in English | WPRIM | ID: wpr-238433

ABSTRACT

Gefitinib is used as a first-line treatment for advanced non-small cell lung cancer (NSCLC). Unfortunately, most NSCLC patients inevitably develop gefitinib resistance during treatment. In addition to EGFR mutation status, the mechanisms involved are largely unknown. In this study, we showed that miR-124, a tumor suppressor, was significantly down-regulated in gefitinib-resistant NSCLC patients and cell lines compared with gefitinib-sensitive patients and cell lines. In addition, the miR-124 depletion induced gefitinib resistance, and miR-124 overexpression sensitized gefitinib-resistant cells to gefitinib. Mechanistic analysis revealed that miR-124 decreased SNAI2 and STAT3 expression by directly targeting their 3'UTRs and that knocking down SNAI2 or STAT3 partly reversed the gefitinib resistance induced by miR-124 depletion. Our data demonstrate that the miR-124 plays a new critical role in acquired resistance to gefitinib and that the manipulation of miR-124 might provide a therapeutic strategy for reversing acquired gefitinib resistance.


Subject(s)
Humans , 3' Untranslated Regions , Antineoplastic Agents , Pharmacology , Therapeutic Uses , Carcinoma, Non-Small-Cell Lung , Drug Therapy , Genetics , Metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Genetics , HEK293 Cells , Lung Neoplasms , Drug Therapy , Genetics , Metabolism , MicroRNAs , Genetics , Quinazolines , Pharmacology , Therapeutic Uses , STAT3 Transcription Factor , Genetics , Metabolism , Snail Family Transcription Factors , Genetics , Metabolism
9.
Chinese Journal of Plastic Surgery ; (6): 128-133, 2015.
Article in Chinese | WPRIM | ID: wpr-353189

ABSTRACT

<p><b>OBJECTIVE</b>To construct and characterize the TGF-β1, induced epithelial-mesenchymal transition (EMT) model of keloid epithelial cells in vitro, and to investigate the expression of epithelial stem cells related surface markers in keloid epithelial cells during EMT induction.</p><p><b>METHODS</b>The epithelial cells from 3 keloid samples of ears were cultured in vitro and induced by transforming growth factor betal (TGF-β1, 1 ng/ml) for 5 days, which was the experimental group, the same cells untreated were considered as the negative control group. The expressions of EMT-associated markers and regulative genes were detected using immunofluorescence staining, real-time PCR and western blot analysis. Then the surface markers of epithelial stem cells were detected using real-time PCR. Statistical significance was determined using Independent-Samples t Test, a p value less than 0. 05 was considered statistically significant.</p><p><b>RESULTS</b>The mRNA expression of transcription factor snail2 and mesenchymal-specific marker vimentin increased significantly in TGF-β1, induced keloid epithelial cells (P < 0. 05), in which snail2 increasing from 0. 91 ± 0. 23 to 1. 69 ± 0. 10, and vimentin from 5. 86 ± 2. 07 to 24. 29 ± 5. 39. Whereas the mRNA expression of epithelial-specific marker E-cadherin decreased from 1. 06 ± 0. 19 to 0. 65 ± 0. 09. The mRNA expression of CD29 and Lgr6, two surface markers of epithelial stem cells, significantly increased after induction of the TGF-β1, (P < 0. 05), from 0. 55 ± 0. 14 and 1. 61 ± 0. 31 to 1. 19 ± 0. 12 and 3. 84 t 0. 62 respectively. In induced cells, the immunofluorescence results showed staining of E- cadherin became faint, but the number of positive staining cells of vimentin increased. Western blot confirmed the protein expression of E-cadherin weakened, and the vimentin and p-Smad3 enhanced (P < 0. 05).</p><p><b>CONCLUSIONS</b>TGF-β1, initiated EMT in keloid epithelial cells by inducing the up-regulation of snail2, and TGF-β1,/Smad3 signaling pathway was involved in EMT. EMT could change the phenotype of epithelial stem cells in keloid.</p>


Subject(s)
Humans , Biomarkers , Metabolism , Cadherins , Genetics , Metabolism , Epithelial Cells , Physiology , Epithelial-Mesenchymal Transition , Physiology , In Vitro Techniques , Keloid , Pathology , RNA, Messenger , Metabolism , Signal Transduction , Smad3 Protein , Genetics , Metabolism , Snail Family Transcription Factors , Transcription Factors , Genetics , Metabolism , Transforming Growth Factor beta1 , Metabolism , Pharmacology , Up-Regulation , Vimentin , Genetics , Metabolism
10.
Journal of Southern Medical University ; (12): 1297-1302, 2015.
Article in Chinese | WPRIM | ID: wpr-333637

ABSTRACT

<p><b>OBJECTIVE</b>To explore expressions of CD133, E-cadherin and Snail in human epithelial ovarian cancer (EOC) and elucidate their relationship with the clinicopathologic features and prognosis of the patients.</p><p><b>METHODS</b>The expression of CD133, E-cadherin and Snail were detected by immunohistochemical staining in 150 specimens of EOC and 50 specimens of benign ovarian epithelial tumor tissues.</p><p><b>RESULTS</b>The positivity rates of CD133, E-cadherin and Snail protein in EOC were 58.7%, 60.7% and 32.7%, respectively, significantly different from the rates in benign epithelial tumor tissues (10%, 8.0%, and 70%, respectively; P<0.05). The expressions of CD133, E-cadherin and Snail in EOC were significantly correlated with abdominal organ and lymphnode metastases and FIGO stage (P<0.01). E-cadherin expression was inversely correlated with Snail and CD133 expression (r=-0.545 and -0.570, P<0.01), and the latter two were positively correlated (r=0.599, P<0.01). Overexpressions of CD133 and Snail and a decreased expression of E-cadherin were all related to a poor prognosis of the patients (P<0.05). FIGO stage and expressions of CD133, E-cadherin and Snail were all independent prognostic factors of EOC (P<0.05).</p><p><b>CONCLUSION</b>The expressions of CD133, E-cadherin and Snail are related to lymph node metastasis, clinical stage, and prognosis of EOC. Combined detection of these indexes provides important evidence for predicting the progression and prognosis of EOC.</p>


Subject(s)
Female , Humans , AC133 Antigen , Antigens, CD , Metabolism , Cadherins , Metabolism , Disease Progression , Glycoproteins , Metabolism , Lymphatic Metastasis , Neoplasms, Glandular and Epithelial , Metabolism , Pathology , Ovarian Neoplasms , Metabolism , Pathology , Peptides , Metabolism , Prognosis , Snail Family Transcription Factors , Transcription Factors , Metabolism
11.
Chinese Journal of Medical Genetics ; (6): 810-813, 2015.
Article in Chinese | WPRIM | ID: wpr-287983

ABSTRACT

<p><b>OBJECTIVE</b>To explore the molecular etiology of two pedigrees affected with type II Waardenburg syndrome (WS2) and to provide genetic diagnosis and counseling.</p><p><b>METHODS</b>Blood samples were collected from the proband and his family members. Following extraction of genomic DNA, the coding sequences of PAX3, MITF, SOX10 and SNAI2 genes were amplified with PCR and subjected to DNA sequencing to detect potential mutations.</p><p><b>RESULTS</b>A heterozygous deletional mutation c.649_651delAGA in exon 7 of the MITF gene has been identified in all patients from the first family, while no mutation was found in the other WS2 related genes including PAX3, MITF, SOX10 and SNAI2.</p><p><b>CONCLUSION</b>The heterozygous deletion mutation c.649_651delAGA in exon 7 of the MITF gene probably underlies the disease in the first family. It is expected that other genes may also underlie WS2.</p>


Subject(s)
Female , Humans , Male , Base Sequence , DNA Mutational Analysis , Exons , Genetics , Family Health , Genetic Predisposition to Disease , Genetics , Heterozygote , Microphthalmia-Associated Transcription Factor , Genetics , Molecular Sequence Data , Mutation , PAX3 Transcription Factor , Paired Box Transcription Factors , Genetics , Pedigree , Polymerase Chain Reaction , SOXE Transcription Factors , Genetics , Sequence Deletion , Snail Family Transcription Factors , Transcription Factors , Genetics , Waardenburg Syndrome , Classification , Diagnosis , Genetics
12.
Chinese Journal of Oncology ; (12): 585-590, 2015.
Article in Chinese | WPRIM | ID: wpr-286776

ABSTRACT

<p><b>OBJECTIVE</b>To detect the expression of interleukin-17A(IL-17A) in hepatocellular carcinoma (HCCs) tissues, and to analyze its relationship with clinicopathological characteristics and epithelial-mesenchymal transition (EMT).</p><p><b>METHODS</b>The expression of IL-17A, E-cadherin, vimentin proteins and Snail mRNA were detected by immunohistochemistry and in situ hybridization histochemistry in the hepatocellular carcinoma (HCC) tissues of 74 patients.</p><p><b>RESULTS</b>IL-17A staining was detected in 54.1% (40/74) specimens of human HCCs, but only 25.0% (5/20) in corresponding peritumoral tissues (P<0.05). The positive rate of IL-17A expression in HCC patients with grade III+IV and UICC stage III+IV tumors was significantly higher than those with grade I+II and UICC stage I+II tumors. The expression of IL-17A was positively correlated with portal vein tumor thrombus and microvascular invasion (all P<0.05). The 1- and 2-year recurrence-free survival rates were 27.6% and 17.2% in the patients with positive IL-17A expression, but 79.3% and 58.5% in IL-17A-negative HCCs. The 1- and 2-year overall survival rates were 69.0% and 27.8% in the cases with positive IL-17A expression, while 91.3% and 87.0% in IL-17A-negative cases. Patients with IL-17A-positive HCCs showed significantly shorter recurrence-free and overall survival compared with the patients with IL-17A-negative HCCs (P<0.05). Multivariate analysis indicated that IL-17A expression was an independent factor for recurrence-free and overall survival of HCCs. IL-17A-positive HCCs were characterized by increased expression of vimentin (r=0.492, P<0.01) or Snail (r=0.410, P<0.05) and loss of E-cadherin expression (r=-0.404, P<0.05).</p><p><b>CONCLUSIONS</b>Our results suggest that IL-17A is closely related to epithelial-mesenchymal transition in hepatocellular carcinoma. IL-17A-positive hepatocellular carcinoma demonstrates more aggressive biological behavior, and IL-17A may serve as a potential prognostic marker for this cancer.</p>


Subject(s)
Humans , Cadherins , Genetics , Metabolism , Carcinoma, Hepatocellular , Metabolism , Mortality , Pathology , Disease-Free Survival , Epithelial-Mesenchymal Transition , Immunohistochemistry , Interleukin-17 , Metabolism , Liver Neoplasms , Metabolism , Mortality , Pathology , Neoplasm Invasiveness , Neoplasm Proteins , Genetics , Metabolism , Neoplasm Recurrence, Local , Prognosis , RNA, Messenger , Metabolism , Snail Family Transcription Factors , Survival Rate , Transcription Factors , Genetics , Metabolism , Vimentin , Genetics , Metabolism
13.
Chinese Journal of Oncology ; (12): 258-265, 2015.
Article in Chinese | WPRIM | ID: wpr-248372

ABSTRACT

<p><b>OBJECTIVE</b>To explore the role of S100A4 in the epithelial-mesenchymal transition (EMT) in esophageal squamous cell carcinoma and its possible molecular mechanism.</p><p><b>METHODS</b>Three chemically synthesized S100A4 siRNA sequences were transiently transfected into esophageal carcinoma EC9706 cells. EC9706 cells transfected with negative siRNA, lipofectamine 2000, and vacant EC9706 cells were used as control. Fluorescence quantitative RT-PCR and Western blot were used to detect the inhibition rate of S100A4 siRNA. S100A4 siRNA2 with the best inhibition rate was chosen to transiently transfect into EC9706 cells under the same conditions. The EC9706 cells transfected with negative siRNA, lipofectamine 2000 and vacant EC9706 cells were also used as control. Fluorescence quantitative RT-PCR and Western blot were used to detect the mRNA and protein expressions of E-cadherin, vimentin and snail. The morphology of EC9706 cells was observed under an inverted microscope. Boyden chamber and scratch test were used to detect the invasion and migration ability of EC9706 cells, and CCK8 assay was used to detect the proliferation ability of EC9706 cells. EC9706 cells transfected with S100A4 siRNA2 were further transfected with snail eukaryotic expression vector. The EC9706 cells transfected with S100A4 siRNA, EC9706 cells transfected with snail eukaryotic expression vector and vacant EC9706 cells were used as control. The above indexes of all the groups were observed, too.</p><p><b>RESULTS</b>The S100A4 mRNA and protein expression levels of the S100A4 siRNA2 group were 0.417 ± 0.041 and 0.337 ± 0.039, the transmembrane cell number was 61.608 ± 8.937, the scratch healing distance was (0.216 ± 0.064) mm, the A value was 0.623 ± 0.084, the E-cadherin mRNA and protein levels were 0.619 ± 0.032 and 0.495 ± 0.034, the vimentin mRNA and protein levels were 0.514 ± 0.032 and 0.427 ± 0.028, the snail mRNA and protein levels were 0.573 ± 0.029 and 0.429 ± 0.041. These data were significantly different with the liposome group, the negative control group and the blank group (P < 0.05 for all). After the S100A4 siRNA2 treatment for 24 h, the appearance of EC9706 cells changed to epithelial cell morphology. The transmembrane cell number and the scratch healing distance of the S100A4 siRNA2+snail eukaryotic expression vector group were (69.382 ± 9.666) cells and (0.274 ± 0.029) mm, the A value was 0.823 ± 0.101, the snail mRNA and protein levels were 0.704 ± 0.037 and 0.625 ± 0.031, the vimentin mRNA and protein levels were 0.712 ± 0.046 and 0.609 ± 0.038, and these data were significantly higher than those of the Sl00A4 siRNA2 group (P < 0.05 for all). The E-cadherin mRNA and protein levels of the S100A4 siRNA2+eukaryotic expression vector group were 0.437 ± 0.038 and 0.381 ± 0.031, significantly lower than those of the S100A4 siRNA2 group (P < 0.05 for all). However, snail had no effect on the morphology of EC9706 cells.</p><p><b>CONCLUSIONS</b>S100A4 may be involved in the EMT process of esophageal squamous-cell carcinoma by regulating the expression of snail and then plays a role in the invasion and metastasis of esophageal carcinoma.</p>


Subject(s)
Humans , Cadherins , Carcinoma, Squamous Cell , Metabolism , Pathology , Cell Line, Tumor , Epithelial Cells , Epithelial-Mesenchymal Transition , Esophageal Neoplasms , Metabolism , Pathology , Indicators and Reagents , Lipids , RNA, Messenger , RNA, Small Interfering , Physiology , S100 Calcium-Binding Protein A4 , S100 Proteins , Genetics , Physiology , Snail Family Transcription Factors , Transcription Factors , Genetics , Transfection , Vimentin , Genetics
14.
Journal of Zhejiang University. Medical sciences ; (6): 479-485, 2015.
Article in Chinese | WPRIM | ID: wpr-255166

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of sunitinib on the migration of ovarian cells and its mechanism of the negative regulation TGF-β mediated of epithelial-mesenchymal transition(EMT) by sunitinib to inhibit ovarian cancer metastasis.</p><p><b>METHODS</b>The migration of human ovarian cancer cells SKOV3 was evaluated by wound-healing and transwell assays. The effects of sunitinib on TGF-β-induced E-cadherin expression was assessed by Western-blotting, real time RT-PCR and immunofluorescence assay. The protein levels of Snail and the transcriptional activity of Smad in sunitinib-treated cells were examined by Western-blotting and SBE-luciferase assay.</p><p><b>RESULTS</b>Sunitinib suppressed the migration of SKOV3 cells in a concentration-dependent manner. TGF-β stimulation reduced E-cadherin protein level, which was attenuated by sunitinib. Sunitinib inhibited the up-regulation of Snail protein level induced by TGF-β treatment. The SBE reporter was constructed by linking the Smad-binding elements promoter upstream of luciferase reporter gene. A remarkable increment of transcriptional activity of Smads complexes was observed in SKOV3 cells exposed to TGF-β, which was significantly prohibited by sunitinib.</p><p><b>CONCLUSION</b>Sunitinib can inhibit the migration of SKOV3 cells and attenuate the down-regulation of E-cadherin protein level induced by TGF-β. Sunitinib can abolish TGF-β-induced up-regulation of Snail protein and decrease the transcriptional activity of Smad complexes. The results indicate that sunitinib suppresses migration of ovarian cancer cells through negative modulation of TGF-β-mediated epithelial-mesenchymal transition.</p>


Subject(s)
Female , Humans , Cadherins , Metabolism , Cell Line, Tumor , Cell Movement , Down-Regulation , Epithelial-Mesenchymal Transition , Indoles , Pharmacology , Ovarian Neoplasms , Pathology , Pyrroles , Pharmacology , Smad Proteins , Metabolism , Snail Family Transcription Factors , Transcription Factors , Metabolism , Transforming Growth Factor beta , Pharmacology , Up-Regulation
15.
Journal of Southern Medical University ; (12): 179-184, 2015.
Article in Chinese | WPRIM | ID: wpr-239216

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of MSX2 interference on epithelial-mesenchymal transitions (EMT) of pancreatic cancer cell line PANC-1.</p><p><b>METHODS</b>Three vectors containing short hairpin RNAs (shRNAs) of MSX2 (shMSX2-1, shMSX2-2, and shMSX2-3) and the empty vector (negative control) were transfected separately into PANC-1 cell line with Lipofectamine2000. Real-time RT-PCR and Western blotting were used to observe changes in the expressions of MSX2, E-cadherin, and vimentin in the cells. CCK-8 assay was used to assess the changes in the cell growth, and wound scratch assay and Transwell assay were employed to evaluate the cell invasion and metastasis after the transfection.</p><p><b>RESULTS</b>Among the 3 shRNA, shMSX2-1 showed the highest interference efficiency. MSX2 knockdown by the specific shRNA of MSX2 significantly increased E-cadherin expressions, lowered vimentin expressions, and suppressed the invasion, metastasis and proliferation of the cells (P<0.05). MSX2 knockdown also resulted in morphological changes of the cells into cobblestone-like cells in close contact. RT-PCR results revealed significantly reduced mRNA expressions of the transcription factors snail and twist (P<0.05) without affecting slug and zeb1 expressions in the cells with MSX2 knockdown. Conclusion MSX2 knockdown can reverse EMT and induce MET in PANC1 cells, in which process the transcription factors snail and twist may play a role.</p>


Subject(s)
Humans , Cadherins , Metabolism , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Homeodomain Proteins , Metabolism , Nuclear Proteins , Metabolism , Pancreas , Pancreatic Neoplasms , Pathology , RNA, Small Interfering , Snail Family Transcription Factors , Transcription Factors , Metabolism , Transfection , Twist-Related Protein 1 , Metabolism , Vimentin , Metabolism
16.
Journal of Southern Medical University ; (12): 1564-1569, 2015.
Article in Chinese | WPRIM | ID: wpr-232570

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of RbAp48 knockdown on the migration and invasion of human cervical cancer cells and explore the mechanism.</p><p><b>METHODS</b>A small interference RNA (siRNA) was used to knock down the expression of RbAp48 in MS751 cells. The changes in cell migration and invasion were evaluated using wound healing assay and Transwell assay, respectively, and the expressions of RbAp48, vimentin, N-cadherin, E-cadherin, Snail, Twist, MMP-2 and TIMP-2 were determined with Western blotting.</p><p><b>RESULTS</b>After siRNA-mediated RbAp48 knockdown, MS751 cells showed a significantly reduced expression of RbAp48 with significantly suppressed cell migration and invasion (P<0.01). RbAp48 knockdown induced obvious down-regulation of the expressions of interstitial cell phenotype proteins vimentin, N-cadherin, and MMP-2 and up-regulation of epithelial cell phenotype proteins E-cadherin and TIMP-2, suggesting the inhibition of epithelial- mesenchymal transition of the cells. The expressions of Snail and Twist were significantly down-regulated in the cells following RbAp48 knockdown.</p><p><b>CONCLUSION</b>Knockdown of RbAp48 can significantly inhibit epithelial-mesenchymal transition and suppress the migration and invasion of cervical cancer cell line MS751, the mechanism of which may involve the down-regulation of Snail and Twist expressions.</p>


Subject(s)
Female , Humans , Antigens, CD , Metabolism , Cadherins , Metabolism , Cell Line, Tumor , Cell Movement , Down-Regulation , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Matrix Metalloproteinase 2 , Metabolism , Neoplasm Invasiveness , Nuclear Proteins , Metabolism , RNA, Small Interfering , Retinoblastoma-Binding Protein 4 , Genetics , Snail Family Transcription Factors , Tissue Inhibitor of Metalloproteinase-2 , Metabolism , Transcription Factors , Metabolism , Twist-Related Protein 1 , Metabolism , Up-Regulation , Uterine Cervical Neoplasms , Pathology , Vimentin , Metabolism
17.
Journal of Southern Medical University ; (12): 1733-1738, 2015.
Article in Chinese | WPRIM | ID: wpr-232536

ABSTRACT

<p><b>OBJECTIVE</b>To explore the expression of Snail and Slug in primary cervical squamous cell carcinoma (CSCC) and their relationship with KAI1 expression.</p><p><b>METHODS</b>The expressions of Snail, Slug, and KAI1 proteins were examined by immunohistochemistry in 154 specimens of CSCC tissues, 50 specimens of cervical intraepithelial neoplasm (CIN), and 40 specimens of normal cervical tissues.</p><p><b>RESULTS</b>The positivity rates of Snail, Slug, and KAI1 expression were 0%, 2.5%, and 95.0% in normal cervical tissues, 32.0%, 34.0% and 64.0% in CIN tissues, and 66.2%, 66.9%, and 43.5% in CSCC tissues, respectively, showing significant differences in the rates among the 3 groups (P<0.05). The expressions of Snail, Slug, and KAI1 were significantly correlated with the histological grades of the tumor, depth of invasion, lymph node metastasis, International Federation of Gynecology and Obstetrics (FIGO) stages, and postoperative survival time (P<0.05). The expressions of Snail and Slug were positively correlated (r=0.752, P<0.001), and both of them were negatively correlated with the expression of KAI1 (P<0.001). Kaplan-Meier analysis showed that patients positive for Snail and Slug had significantly lower survival rates than the negative patients (P<0.001), while a positive expression of KAI1 was associated with a higher survival rate of the patients. Cox regression analysis identified Snail, KAI1, and FIGO stage as independent factors that affected the outcomes of CSCC (P<0.05).</p><p><b>CONCLUSION</b>The expressions of Snail, Slug, and KAI1 are related to the tumor grade, FIGO stage, invasive depth, lymph node metastasis, and prognosis of CSCC, and their combined detection can help estimate the outcomes of the patients.</p>


Subject(s)
Female , Humans , Carcinoma, Squamous Cell , Metabolism , Pathology , Uterine Cervical Dysplasia , Metabolism , Pathology , Immunohistochemistry , Kangai-1 Protein , Metabolism , Kaplan-Meier Estimate , Lymphatic Metastasis , Neoplasm Staging , Prognosis , Snail Family Transcription Factors , Survival Rate , Transcription Factors , Metabolism , Uterine Cervical Neoplasms , Metabolism , Pathology
18.
Biomedical and Environmental Sciences ; (12): 544-548, 2015.
Article in English | WPRIM | ID: wpr-264548

ABSTRACT

Epithelial-mesenchymal transition (EMT) plays an important role in fibrotic diseases. We have previously showed that silica induces EMT in human bronchial epithelial cells (BECs); however, the underlying mechanism of silica-induced EMT is poorly understood. In the present study, we investigated the role of Snail in silica-induced EMT in human BECs in vitro. Human BECs were treated with silica at various concentrations and incubation times. Then MTT assay, western blot, electrophoretic mobility shift assay (EMSA), and small interfering RNA (siRNA) transfection were performed. We found that silica increased the expression and DNA binding activity of Snail in human BECs. SNAI siRNA inhibited the silica-induced expression of Snail. Moreover, SNAI siRNA upregulated the expression of epithelial marker E-cadherin, but attenuated the expression of mesenchymal marker α-smooth muscle actin and vimentin in silica-stimulated cells. These results suggest that Snail mediates the silica-induced EMT in human BECs.


Subject(s)
Humans , Actins , Metabolism , Blotting, Western , Bronchi , Cell Biology , Metabolism , Cadherins , Metabolism , Cell Culture Techniques , Cell Line , Cell Survival , Electrophoretic Mobility Shift Assay , Epithelial Cells , Cell Biology , Metabolism , Epithelial-Mesenchymal Transition , Particle Size , RNA, Small Interfering , Genetics , Silicon Dioxide , Toxicity , Snail Family Transcription Factors , Transcription Factors , Genetics , Metabolism
19.
Journal of Central South University(Medical Sciences) ; (12): 883-888, 2012.
Article in Chinese | WPRIM | ID: wpr-814770

ABSTRACT

OBJECTIVE@#To investigate the simultaneous inhibition of X-linked inhibitor of apoptosis protein (XIAP) and survivin expression on epithelial-mesenchymal transition (EMT) and invasiion of pancreatic cancer cells Panc-1, and its mechanism.@*METHODS@#On the established human pancreatic cancer cells Panc-1-XS, the expression of XIAP and survivin was inhibited simultaneously. Cell invasion and migration were detected by Transwell chamber experiments and scratch test, and the expression of epithelial marker E-cadherin, mesenchymal markers Slug, phosphatase and tensin homolog deleted on chromosome ten (PTEN) and P-Akt protein was determined by Western blot.@*RESULTS@#Cell invasion and migration of Panc-1-XS cells decreased significantly, accompanied by significantly upregulated protein expression of E-cadherin, and significantly declined protein expression of the Slug, indicating increased mesenchymal-epithelial conversion (MET); and increased protein expression of PTEN, and declined protein expression of P-Akt.@*CONCLUSION@#Simultaneously inhibiting the expression of XIAP and survivin can partially reverse EMT phenotype of pancreatic cancer Panc-1 cells, which then significantly reduces the cell invasion and migration of Panc-1 cell lines. This process may be regulated by PTEN/PI3K/Akt signaling pathway.


Subject(s)
Humans , Antigens, CD , Cadherins , Metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Genetics , Inhibitor of Apoptosis Proteins , Genetics , Metabolism , Neoplasm Invasiveness , PTEN Phosphohydrolase , Metabolism , Pancreatic Neoplasms , Pathology , Proto-Oncogene Proteins c-akt , Metabolism , Signal Transduction , Snail Family Transcription Factors , Survivin , Transcription Factors , Metabolism , X-Linked Inhibitor of Apoptosis Protein , Genetics , Metabolism
20.
Acta Academiae Medicinae Sinicae ; (6): 319-323, 2012.
Article in English | WPRIM | ID: wpr-352908

ABSTRACT

<p><b>OBJECTIVE</b>To study the effect of LY294002 on the adriamycin- induced epithelial-mesenchymal transition in human breast carcinoma cells.</p><p><b>METHODS</b>Human breast carcinoma cells MCF-7 was cultured in vitro and then exposed to adriamycin with or without LY294002. The protein expression levels of Akt, phosphorylated-Akt (p-Akt), Snail, and E-cadherin was detected by Western blot analysis. The mRNA expressions of Snail and E-cadherin were determined by RT-PCR.</p><p><b>RESULTS</b>Adriamycin significantly increased the protein expression of Snail and depressed the protein expression of E-cadherin (P<0.05). The pre-treatment with LY294002 significantly reversed the changes of activities and levels of the above proteins (P<0.05).</p><p><b>CONCLUSION</b>LY294002 could reverse the adriamycin-induced epithelial-mesenchymal transition in human breast carcinoma cells by regulating the expressions of Snail and E-cadherin through suppressing PI3K/Akt signaling pathway.</p>


Subject(s)
Humans , Breast Neoplasms , Metabolism , Pathology , Cadherins , Metabolism , Chromones , Pharmacology , Doxorubicin , Pharmacology , Epithelial-Mesenchymal Transition , MCF-7 Cells , Morpholines , Pharmacology , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Metabolism , Signal Transduction , Snail Family Transcription Factors , Transcription Factors , Metabolism
SELECTION OF CITATIONS
SEARCH DETAIL