Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Biomed Pharmacother ; 114: 108832, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30965236

ABSTRACT

Circular RNAs are known to participate in tumorigenesis through a variety of pathways, and as such, have potential to serve as molecular markers in tumor diagnosis and treatment. Here, using quantitative reverse transcription (qRT)-PCR, we showed that circ-CSPP1 is highly expressed in ovarian cancer (OC) tissues. Particularly, we detected circ-CSPP1 expression in three OC cell lines; of which, OVCAR3 and A2780 demonstrated higher levels of circ-CSPP1 expression, and CAOV3 showed lower circ-CSPP1 expression level. Subsequent silencing of circ-CSPP1 in OVCAR3 and A2780 cell lines revealed decreased cell growth, migration and invasion, while overexpression of circ-CSPP1 caused opposite results We also found that miR-1236-3p is a target of circ-CSPP1. Circ-CSPP1 silencing increased the expression of miR-1236-3p, and circ-CSPP1 overexpression decreased miR-1236-3p expression. MiR-1236-3p reportedly plays a tumor-suppressor role in OC by targeting zinc finger E-box binding homeobox 1 (ZEB1). In agreement with this, we showed that silencing circ-CSPP1 significantly decreased ZEB1 expression at both RNA and protein levels, and epithelial-mesenchymal transition (EMT) related markers (E-cadherin and N-cadherin) varied with ZEB1 expression. Circ-CSPP1 silencing also caused decreased expression of matrix metalloproteinase-2 (MMP-2) and vascular endothelial growth factor A (VEGFA), both of which are related to tumorigenesis. Overexpression of circ-CSPP1 had opposite effects. In addition, we indicated that the tumor-promoting effect was inhibited after we transfected miR-1236-3p into circ-CSPP1 overexpressing OC cells. Altogether, our findings suggest that by acting as a miR-1236-3p sponge, circ-CSPP1 impairs the inhibitory effect of miR-1236-3p on ZEB1, which subsequently promotes EMT and OC development.


Subject(s)
Cell Cycle Proteins/genetics , Cell Movement/genetics , Cell Proliferation/genetics , MicroRNAs/genetics , Microtubule-Associated Proteins/genetics , Ovarian Neoplasms/genetics , RNA/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Matrix Metalloproteinase 2/genetics , Ovarian Neoplasms/pathology , RNA, Circular , Vascular Endothelial Growth Factor A/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics
2.
Biomed Pharmacother ; 114: 108787, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30925458

ABSTRACT

CEMIP is a cell migration-inducing protein that is closely associated with carcinogenesis. However, the function of CEMIP has not been reported in ovarian cancer. Here we show that CEMIP expression level in ovarian cancer tissues was higher than that in normal ovaries. Silencing of CEMIP in ovarian cancer cell lines A2780 and OVCAR3 significantly decreased cell proliferation, cell migration and invasion capacity, while the proportion of apoptotic cells was increased. In addition, Western blotting demonstrated that knockdown of CEMIP inhibited activation of the PI3K/AKT signaling pathway. Altogether, these data suggest that CEMIP may promote the development of ovarian cancer by regulating PI3K/AKT signaling.


Subject(s)
Ovarian Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Apoptosis/physiology , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronoglucosaminidase , Ovarian Neoplasms/pathology
3.
Theranostics ; 9(3): 676-690, 2019.
Article in English | MEDLINE | ID: mdl-30809301

ABSTRACT

Cancer cells undergo metabolic reprogramming to support their energy demand and biomass synthesis. However, the mechanisms driving cancer metabolism reprogramming are not well understood. Methods: The differential proteins and interacted proteins were identified by proteomics. Western blot, qRT-PCR and IHC staining were used to analyze TBC1D8 levels. In vivo tumorigenesis and metastasis were performed by xenograft tumor model. Cross-Linking assays were designed to analyze PKM2 polymerization. Lactate production, glucose uptake and PK activity were determined. Results: We established two aggressive ovarian cancer (OVCA) cell models with increased aerobic glycolysis. TBC1D8, a member of the TBC domain protein family, was significantly up-regulated in the more aggressive OVCA cells. TBC1D8 is amplified and up-regulated in OVCA tissues. OVCA patients with high TBC1D8 levels have poorer prognoses. TBC1D8 promotes OVCA tumorigenesis and aerobic glycolysis in a GAP activity-independent manner in vitro and in vivo. TBC1D8 bound to PKM2, not PKM1, via its Rab-GAP TBC domain. Mechanistically, TBC1D8 binds to PKM2 and hinders PKM2 tetramerization to decreases pyruvate kinase activity and promote aerobic glycolysis, and to promote the nuclear translocation of PKM2, which induces the expression of genes which are involved in glucose metabolism and cell cycle. Conclusions:TBC1D8 drives OVCA tumorigenesis and metabolic reprogramming, and TBC1D8 serves as an independent prognosis factor for OVCA patients.


Subject(s)
Calcium-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , Ovarian Neoplasms/metabolism , Animals , Carcinogenesis , Carrier Proteins , Cell Line, Tumor , Dimerization , Female , Gene Expression Regulation, Neoplastic , Glycolysis , HEK293 Cells , Humans , Membrane Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Ovarian Neoplasms/genetics , Prognosis , Protein Domains , Pyruvate Kinase/metabolism , Thyroid Hormones , Up-Regulation , Thyroid Hormone-Binding Proteins
4.
Oncol Lett ; 8(2): 891-897, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25009663

ABSTRACT

Metastasis-associated in colon cancer 1 (MACC1) is a newly identified gene that has been shown to promote tumor cell invasion and metastasis. The present study investigated the effect of MACC1 downregulation on the biological characteristics of the ovarian cancer OVCAR3 cell line. In this study, MACC1 expression was blocked using the RNA interference technique. The downregulation of MACC1 mRNA and protein expression was confirmed using quantitative polymerase chain reaction and western blot analysis. The proliferative activity and adhesion rate of the cells were detected using cell counting kit-8 and a cell adhesion assay, while cell invasion was determined using a Matrigel invasion assay and migration capacity was observed using migration and wound-healing assays. A tube formation assay was also used to examine the angiogenic capacity of cells, and a luciferase assay was performed to assess whether MACC1 binds to the c-MET gene. The MACC1 mRNA and protein expression levels were significantly downregulated using sequence-specific small interfering RNA (siRNA). The inhibition of MACC1 expression markedly decreased the invasive, metastatic and angiogenic capacities of the cells, but only slightly inhibited growth and adhesion. In addition, a putative MACC1-binding site was identified in the 3'-untranslated region of c-MET. MACC1-siRNA was also found to significantly reduce the expression of the c-MET protein and a luciferase reporter assay confirmed that c-MET was the target gene of MACC1. These results demonstrated that the attenuation of MACC1 suppresses cell invasion and migration and that MACC1 may regulate cell metastasis through targeting the expression of c-MET. Inhibition of the function of MACC1 may represent a new strategy for treating ovarian cancer.

5.
Zhonghua Yi Xue Za Zhi ; 93(21): 1653-6, 2013 Jun 04.
Article in Chinese | MEDLINE | ID: mdl-24125676

ABSTRACT

OBJECTIVE: To evaluate the feasibility and safety of applying video endoscopic inguinal lymphadenectomy via hypogastric subcutaneous approach (VEIL-H) in the treatment of vulvar carcinoma. METHODS: From September 2009 to December 2012, 15 patients with vulvar carcinoma underwent VEIL-H plus radical vulvectomy at many participating hospitals. RESULTS: All were treated surgically. Two of them underwent laparoscopic pelvic lymphadenectomy (LPL) upon the positive results of parotid operations with frozen section. The mean operative duration of VEIL-H (bilateral groin) were (80.8 ± 2.9) minutes. The mean total volume of blood loss in VEIL was (5.5 ± 0.4) ml, the mean drainage duration (6 ± 2) days and the average postoperative hospitalization (11 ± 4) days. The mean follow-up period was 13.0 months. One patient suffered local recurrence at Month 2 postoperation. CONCLUSION: VEIL-H is both feasible and safe in inguinal lymphadenectomy.


Subject(s)
Groin , Laparoscopy , Lymph Node Excision/methods , Vulvar Neoplasms/surgery , Adult , Aged , Female , Humans , Male , Middle Aged
7.
J Zhejiang Univ Sci B ; 13(11): 894-903, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23125082

ABSTRACT

BACKGROUND AND OBJECTIVE: Gonadotropin-releasing hormone (GnRH) plays an important role in the regulation of ovarian function and ovarian cancer cell growth. In this study, we determined whether administration of the GnRH agonist (GnRHa), triporelin, prior to cisplatin treatment affects cisplatin and/or prevents cisplatin-induced ovarian damage. METHODS: nu/nu mice were injected with ovarian cancer OVCAR-3 cells intraperitoneally. After two weeks, the mice were treated with saline (control), cisplatin, GnRHa, or cisplatin plus GnRHa for four weeks. At the end of the experimental protocol, blood, tumor, ovary, and uterine tissues were resected for hematoxylin and eosin (H&E) staining, immunohistochemical analyses of Ki67, nuclear factor-κB (NF-κB), and caspase-3, transmission electron microscopy of apoptosis, or enzyme-linked immunosorbent assay (ELISA) analyses of anti-Mullerian hormone (AMH). RESULTS: Cisplatin treatment effectively inhibited tumor growth in mice treated with human ovarian cancer cells; however the treatment also induced considerable toxicity. Immunohistochemical analyses showed that Ki67 expression was reduced in cisplatin-treated mice compared to control (P<0.05), but there was no statistically significant differences between cisplatin-treated mice and cisplatin plus GnRHa-treated mice (P>0.05), while expressions of NF-κB and caspase-3 were reduced and induced, respectively, in cisplatin-treated mice and cisplatin plus GnRHa-treated mice. Apoptosis occurred in the GnRHa, cisplatin, and cisplatin plus GnRHa-treated mice, but not in control mice. Ovaries exposed to GnRHa in both GnRHa mice and cisplatin-treated mice (combination group) had significantly more primordial and growth follicles and serum levels of AMH than those in the control mice and cisplatin-treated mice (P<0.05). CONCLUSIONS: Administration of GnRHa to mice significantly decreased the extent of ovarian damage induced by cisplatin, but did not affect the anti-tumor activity of cisplatin.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Cystadenocarcinoma, Serous/drug therapy , Gonadotropin-Releasing Hormone/agonists , Ovarian Neoplasms/drug therapy , Ovary/drug effects , Triptorelin Pamoate/pharmacology , Animals , Anti-Mullerian Hormone/metabolism , Antineoplastic Agents/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Drug Interactions , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Microscopy, Electron, Transmission , NF-kappa B/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Random Allocation , Xenograft Model Antitumor Assays
8.
Oncol Lett ; 3(5): 1030-1036, 2012 May.
Article in English | MEDLINE | ID: mdl-22783386

ABSTRACT

Apoptosis induction by short hairpin RNA (shRNA) expression vectors may be an efficient and promising strategy for cancer gene therapy. Ultrasound-targeted microbubble destruction (UTMD) is an appealing technique; however, there few data are available to demonstrate the feasibility and to optimize the methodology for this technology. The aim of this study was to optimize this technique and to elucidate the effects on gene inhibition and apoptosis induction in vitro. Human cervical cancer cell lines were obtained and cultured.shRNA vectors were constructed, and the UTMD technique was examined to determine whether or not it was suitable for shRNA transfection into cells. Cells were then examined using flow cytometry. The results revealed that the optimal irradiation parameters obtained higher transfection efficiency and did not affect the integrity of plasmid DNA. We concluded that survivin downregulation with shRNA expression vectors, mediated by the optimal UTMD parameters, markedly induced cell apoptosis and caused cell cycle arrest, laying a foundation for further investigation of this cancer therapy.

9.
Zhonghua Fu Chan Ke Za Zhi ; 47(10): 764-8, 2012 Oct.
Article in Chinese | MEDLINE | ID: mdl-23302735

ABSTRACT

OBJECTIVE: To explore the lowest effective dosage of mifepristone combined with misoprostol in terminating ultra-early pregnancy. METHODS: All the cases of ultra-early pregnancy classified by amenorrhea days, ß-hCG and vaginal B-ultrasonic were randomly divided into two groups. One hundred cases in G1 group (minimized dosage) were orally administered 25 mg mifepristone once a day for 2 days and combined with 200 µg misoprostol 48 hours later, while 150 mg mifepristone combined with 600 µg misoprostol 48 hours later were given to 100 cases in G2 group (normal dosage). All cases were observed for 6 hours after taking misoprostol and returned for assessment three days later. RESULTS: None missing. Expulsion of conceptus: G1 and G2 group were 22 (22.0%, 22/100) and 25 (25.0%, 25/100; P > 0.05). Failure rate: cases with incomplete abortion were 1 (1.0%, 1/100) and 2 (2.0%, 2/100) in G1 and G2 group, hospitalization for suspected ectopic pregnancies both was 1 (1.0%). Bleeding: bleeding cases during the administration of mifepristone in G1 and G2 group were 71 (71.0%, 71/100) and 78 (78.0%, 78/100; P > 0.05); the mean bleeding time were (5.3 ± 1.4) days and (6.0 ± 1.5) days (P < 0.01). Other side effects: in G1 group, majority showed light nausea (7.0%, 7/100) and light abdominal pain (20.0%, 20/100). Menses recovery: 99 (99.0%, 99/100) for G1 group and 98 (98.0%, 98/100) for G2 group to recovery on scheduled time. Satisfactions: both were 99 (99.0%, 99/100). Except mean bleeding days and side-effects, the differences above showed no significance (P > 0.05). CONCLUSION: It is safe and effective treatment with the lowest dosages of mifepristone and misoprostol to terminate ultra-early pregnancies.


Subject(s)
Abortifacient Agents/administration & dosage , Abortion, Induced/methods , Mifepristone/administration & dosage , Misoprostol/administration & dosage , Abdominal Pain/etiology , Abortifacient Agents/adverse effects , Administration, Oral , Adult , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Mifepristone/adverse effects , Misoprostol/adverse effects , Nausea/etiology , Patient Satisfaction , Pregnancy , Treatment Outcome , Uterine Hemorrhage/etiology , Young Adult
10.
Oncol Rep ; 27(4): 1011-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22200669

ABSTRACT

Breast cancer metastasis suppressor 1 (BRMS1) is a predominantly nuclear protein that differentially regulates the expression of multiple genes, leading to suppression of metastasis without affecting orthotopic tumor growth. It has been demonstrated that BRMS1 may be correlated with advanced ovarian cancer. The aim of this study was to investi-gate the mechanisms of BRMS1 involvement in ovarian cancer metastasis. We constructed a plasmid containing a short hairpin RNA (shRNA) against BRMS1 and transfected it into the ovarian cancer cell line OVCAR3. Real-time reverse transcription polymerase chain reaction (real-time PCR) and Western blot analyses demonstrated that BRMS1 expression was efficiently downregulated. Stable suppression of BRMS1 significantly enhanced cell adhesion, migration, invasion and angiogenesis. We also found that chemokine receptor 4 (CXCR4) was upregulated at both the mRNA and protein levels. When approaching for the mechanism, we discovered that activation of the nuclear factor-κB (NF-κB) signaling pathway mediated CXCR4 upregulation, as demonstrated by the electrophoretic mobility shift assay (EMSA). Collectively, these results suggest that attenuation of BRMS1 may play a critical role in promoting migration, invasion and angiogenesis of ovarian cancer cells and BRMS1 may regulate the metastatic potential at least in part through upregulation of CXCR4 via NF-κB activation. Restoration of BRMS1 function is thus a potential new strategy for treating human ovarian cancer.


Subject(s)
Cell Movement , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Receptors, CXCR4/metabolism , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Down-Regulation , Electrophoretic Mobility Shift Assay , Female , Genetic Therapy , Human Umbilical Vein Endothelial Cells/metabolism , Humans , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neovascularization, Physiologic , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , RNA Interference , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/genetics , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transfection , Up-Regulation
11.
Ai Zheng ; 21(2): 122-6, 2002 Feb.
Article in Chinese | MEDLINE | ID: mdl-12479058

ABSTRACT

BACKGROUND & OBJECTIVE: Doc-1R gene is a new gene which was cloned in 1999. Recent studies suggest that Doc-1R gene is a potential tumor suppressor gene. In order to study the function of the Doc-1R gene, The aim of this study was to obtain its genomic sequences and to analyze its expression in the tissues. METHOD: According to the cDNA sequences of Doc-1R gene, the gene-specific primers were designed and synthesized. The sequence of Doc-1R gene was cloned through nested PCR using the Genome Walker kit. The sequence and splice donor/acceptor site were analyzed. Using the reverse transcription polymerase chain reaction(RT-PCR), the expression of Doc-1R gene in thirteen tissues, including mouse liver, spleen, pancreas, kidney, lung, intestine, heart, brain, bone, muscle, bladder, ovary, spermary were determined. RESULTS: The mouse Doc-1R gene has been obtained by two times genomic walking. This gene spans 2,787 bp and contains four exons and three introns. All of the splice donor/acceptor site sequences were in accordance with the consensus "GT-AG" rule. RT-PCR experiments demonstrated Doc-1R gene was expressed in the 13 tissue samples. CONCLUSION: The authors have successfully cloned the mouse Doc-1R gene, which will be the foundation for further investigating the function of that gene. The expression pattern suggests that the Doc-1R gene is a housekeeping gene which is important to keep the function of tissues and organs.


Subject(s)
Oncogene Proteins/genetics , Animals , Base Sequence , Cloning, Molecular , Genes, Tumor Suppressor , Mice , Molecular Sequence Data , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
12.
Ai Zheng ; 21(3): 240-4, 2002 Mar.
Article in Chinese | MEDLINE | ID: mdl-12451986

ABSTRACT

BACKGROUND & OBJECTIVE: DOC-1R gene is a candidate tumor suppressor gene that when connected specifically with CDK2 can control the course of cell cycle by restraining the reciprocity of CDK2 and cyclin. The aim of this study was to construct the antisense DOC-1R plasmid and to investigate the effect of DOR-1R gene on the growth of normal cell. METHOD: The recombinant antisense plasmid was constructed after screening the expression of DOC-1R gene. Following transfection, the effect of DOC-1R on cell growth was determined by assessing the ability of cell replication and observing soft agar culture. RESULT: The growth speed of NIH3T3 transfected by mouse DOC-1R gene was of significant difference from that transfected by empty vector. The pcDNA3-DOC-1R+ vector significantly inhibited the cell replication, while the pcDNA3-DOC-1R- vector stimulated the cell replication. In soft agar culture, the colony formation capacity was decreased in the recombinant sense vector group. The clone formation rate was decreased and the size of the colony formed was smaller as well. In contrast, the colony forming ability was remarkably increased in the antisense vector group. The clone formation rate was increased significantly, compared to that in the sense group. CONCLUSION: Mouse DOC-1R gene can significantly inhibit cell growth and colony formation capacity. It will be helpful for the study on the mechanism of normal cell growth and replication as well as for research in tumor treatment and prevention.


Subject(s)
DNA, Antisense/genetics , Oncogene Proteins/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Division/drug effects , Cell Division/physiology , DNA, Antisense/biosynthesis , DNA, Antisense/pharmacology , Genes, Tumor Suppressor/physiology , Genetic Vectors/genetics , Mice , Molecular Sequence Data , Oncogene Proteins/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...