ovarian cancer cell lines
Recently Published Documents


TOTAL DOCUMENTS

658
(FIVE YEARS 145)

H-INDEX

47
(FIVE YEARS 7)

2022 ◽  
Vol 12 (2) ◽  
pp. 365-372
Author(s):  
Chunhong Song ◽  
Juan Zhen ◽  
Aihua Gong ◽  
Longying Zhang

Background: The Cripto-1 (CR-1)/glucose-regulated protein 78 (GRP78) complex was involved in enhancing survival in different types of cells. CR-1 presented increased levels in ovarian carcinoma tissue. However, the potential mechanism of CR-1/GRP78 was unclear in ovarian cancer. Thus, the study aimed to analyze the role of CR-1/GRP78 in ovarian carcinoma cells. Methods and materials: The CR-1 and GRP78 expression in different ovarian cancer cell lines were detected by RT-qPCR and Western blot (WB). Immunoprecipitation assay was performed to analyze whether Cripto-1 interacted with GRP78. The CR-1 interfering plasmids or GRP-78 overexpressing plasmids transfected into cells were used to decrease endogenous CR-1 levels and increase GRP-78 levels. Cell clonogenicity and proliferation capabilities were separately evaluated by clone growth assay, along with the detection of cell migration and invasion abilities by transwell and wound healing assay. In addition, Matrix Metalloproteinases (MMPs) levels were detected by WB. The cell apoptosis was analyzed by Flow Cytometer and the detection of apoptosis-related proteins. Results: The results showed that CR-1 and GRP78 levels were higher in SKOV3 than other cell lines. Furthermore, CR-1 interacted with GRP78 in cells, which formed protein complex. CR-1 silence significantly decreased GRP-78 levels. Moreover, GRP78 overexpression blocked the anti-survival effects caused by CR-1 knockdown. Conclusion: CR-1 silence inhibited cell proliferation and promoted apoptosis via GRP78. It replied that GRP-78 overexpression might enhance the biological functions of CR-1/GRP78 complex ameliorated by CR-1 silence. Thus, CR-1/GRP78 could be a potential target for treating ovarian carcinoma.


2022 ◽  
Vol 23 (2) ◽  
pp. 833
Author(s):  
Sonia Capellero ◽  
Jessica Erriquez ◽  
Chiara Battistini ◽  
Roberta Porporato ◽  
Giulia Scotto ◽  
...  

Peritoneal metastases are the leading cause of morbidity and mortality in ovarian cancer. Cancer cells float in peritoneal fluid, named ascites, together with a definitely higher number of non neo-neoplastic cells, as single cells or multicellular aggregates. The aim of this work is to uncover the features that make these aggregates the metastasizing units. Immunofluorescence revealed that aggregates are made almost exclusively of ovarian cancer cells expressing the specific nuclear PAX8 protein. The same cells expressed epithelial and mesenchymal markers, such as EPCAM and αSMA, respectively. Expression of fibronectin further supported a hybrid epithelia-mesenchymal phenotype, that is maintained when aggregates are cultivated and proliferate. Hematopoietic cells as well as macrophages are negligible in the aggregates, while abundant in the ascitic fluid confirming their prominent role in establishing an eco-system necessary for the survival of ovarian cancer cells. Using ovarian cancer cell lines, we show that cells forming 3D structures neo-expressed thoroughly fibronectin and αSMA. Functional assays showed that αSMA and fibronectin are necessary for the compaction and survival of 3D structures. Altogether these data show that metastasizing units display a hybrid phenotype that allows maintenance of the 3D structures and the plasticity necessary for implant and seeding into peritoneal lining.


2022 ◽  
Vol 22 (1) ◽  
Author(s):  
Yang Zhou ◽  
Chunyan Wang ◽  
Jinye Ding ◽  
Yingying Chen ◽  
Yaoqi Sun ◽  
...  

Abstract Background Accumulating evidence has revealed that aberrant microRNA (miRNA) expression can affect the development of chemotherapy drug resistance by modulating the expression of relevant target proteins. Emerging evidence has demonstrated that miR-133a participates in the tumorigenesis of various cancers. However, whether miR-133a is associated with cisplatin resistance in ovarian cancer remains unclear. Objective To investigate the role of miR-133a in the development of cisplatin resistance in ovarian cancer. Methods MiR-133a expression in cisplatin-resistant ovarian cancer cell lines was assessed by reverse-transcription quantitative PCR (RT–qPCR). A cell counting kit-8 (CCK-8) assay was used to evaluate the viability of tumour cells treated with cisplatin in the presence or absence of miR-133a. A luciferase reporter assay was used to analyse the binding of miR-133a with the 3′ untranslated region (3′UTR) of YES proto-oncogene 1 (YES1). The YES1 expression level was analysed using a dataset from the International Cancer Genome Consortium (ICGC) and assessed by RT–qPCR and western blotting in vitro. The roles and mechanisms of YES1 in cell functions were further probed via gain- and loss-of-function analysis. Results The expression of miR-133a was significantly decreased in cisplatin-resistant ovarian cancer cell lines (A2780-DDP and SKOV3-DDP), and the overexpression of the miR-133a mimic reduced cisplatin resistance in A2780-DDP and SKOV3-DDP cells. Treatment with the miR-133a inhibitor increased cisplatin sensitivity in normal A2780 and SKOV3 cells. MiR-133a binds the 3’UTR of YES1 and downregulates its expression. Bioinformatics analysis revealed that YES1 expression was upregulated in recurrent cisplatin-resistant ovarian cancer tissue, and in vitro experiments also verified its upregulation in cisplatin-resistant cell lines. Furthermore, we discovered that miR-133a downregulated the expression of YES1 and thus inhibited cell autophagy to reduce cisplatin resistance. Yes1 knockdown significantly suppressed the cisplatin resistance of ovarian cancer cells by inhibiting autophagy in vitro. Xenograft tumour implantation further demonstrated that Yes1 overexpression promoted ovarian tumour development and cisplatin resistance. Conclusions Our results suggest that the miR-133a/YES1 axis plays a critical role in cisplatin resistance in human ovarian cancer by regulating cell autophagy, which might serve as a promising therapeutic target for ovarian cancer chemotherapy treatment in the future.


Author(s):  
Apratim Mukherjee ◽  
Haonan Zhang ◽  
Katherine Ladner ◽  
Megan Brown ◽  
Jacob Urbanski ◽  
...  

Ovarian cancer is routinely diagnosed long after the disease has metastasized through the fibrous sub-mesothelium. Despite extensive research in the field linking ovarian cancer progression to increasingly poor prognosis, there are currently no validated cellular markers or hallmarks of ovarian cancer that can predict metastatic potential. To discern disease progression across a syngeneic mouse ovarian cancer progression model, here, we fabricated extracellular-matrix mimicking suspended fiber networks: crosshatches of mismatch diameters for studying protrusion dynamics, aligned same diameter networks of varying inter-fiber spacing for studying migration, and aligned nanonets for measuring cell forces. We found that migration correlated with disease, while force-disease biphasic relationship exhibited f-actin stress-fiber network dependence. However, unique to suspended fibers, coiling occurring at tips of protrusions and not the length or breadth of protrusions displayed strongest correlation with metastatic potential. To confirm that our findings were more broadly applicable beyond the mouse model, we repeated our studies in human ovarian cancer cell lines and found that the biophysical trends were consistent with our mouse model results. Altogether, we report complementary high throughput and high content biophysical metrics capable of identifying ovarian cancer metastatic potential on time scale of hours. [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text]


2022 ◽  
Vol 11 ◽  
Author(s):  
Ruth M. Escalona ◽  
George Kannourakis ◽  
Jock K. Findlay ◽  
Nuzhat Ahmed

BackgroundThe tissue inhibitors of metalloproteinase (TIMPs) and their associated metalloproteinase (MMPs) are essential regulators of tissue homeostasis and are essential for cancer progression. This study analyzed the expression of TIMP-1,-2,-3 and the associated MMPs (MMP-2,-9,-11,-14) in different Stages, Grades and World Health Organization (WHO) classifications of serous ovarian tumors, ascites, ascites-derived cells from chemo-naïve (CN) and relapsed (CR) patients, and in ovarian cancer cell lines. The status of TIMPs and associated MMPs in response to chemotherapy treatment was assessed in cancer cell lines; TCGA data was interrogated to gauge TIMPs and associated MMPs as prognostic and platinum-response indicators.MethodsThe levels of TIMP-1, -2 and -3 were assessed by immunohistochemistry. The mRNA expression of TIMPs and MMPs was quantified by real time PCR (qRT-PCR). The chemosensitivity (IC50 values) to Cisplatin or Paclitaxel in cell lines was evaluated by MTT assay. The levels of TIMPs in ascites and cell lysates were analyzed by an ELISA assay.ResultsThe expression of TIMP-2 was significantly upregulated in Type 2 compared to Type 1 tumors and normal/benign ovarian tissues. TIMP-3 expression was significantly enhanced in Stage III, Grade 3 and Type 2 tumors compared to normal/benign ovarian tissues. The mRNA expression of MMP-9,-11 and -14 was significantly upregulated in Stage IV compared to normal/benign ovarian tissues. The expression of TIMP-1 was highest, followed by TIMP-2 and then TIMP-3 in CN ascites. At the cellular level, TIMP-2 mRNA expression was significantly higher in CN compared to CR epithelial cells in patients. The expression of TIMP-1 and -2, MMPs and cancer stem cells (CSCs) were upregulated in response to chemotherapy treatments in cancer cell lines. Interrogation of the TCGA dataset suggests shifts in platinum responses in patients consistent with genetic alterations in TIMP-2, -3 and MMP-2, -11 genes in tumors; and decreased overall survival (OS) and progression-free survival (PFS) in patients with altered MMP-14 genes.ConclusionsTIMPs and related MMPs are differentially expressed in serous ovarian tumors, ascites, ascites-derived cells and ovarian cancer cell lines. Chemotherapy treatment modulates expression of TIMPs and MMPs in association with increased expression of genes related to cancer stem cells.


Author(s):  
Heidelinde Fiegl ◽  
Judith Hagenbuchner ◽  
Christiana Kyvelidou ◽  
Beata Seeber ◽  
Sieghart Sopper ◽  
...  

Author(s):  
Ju Li ◽  
Khodabakhsh Rashidi ◽  
Behnam Mahdavi ◽  
Samaneh Goorani ◽  
Mohammad Karimian ◽  
...  

IntroductionRecently, various nanoparticles containing medicinal plants have been specifically designed to deliver anticancer drugs and nucleic acids such as DNA and RNA to cancer cells and as a result, they open up new avenues in cancer treatment strategies. In this study, gold nanoparticles were synthesized in aqueous medium using Nigella damascena extract as ‎stabilizing and reducing agents. ‎Material and methodsThe synthesized nanoparticles (AuNPs) were characterized using different techniques including UV-Vis. and ‎FT-IR spectroscopy, and Transmission electron microscopy (TEM). TEM images exhibited a uniform spherical ‎morphology in size of 21.64 nm for the biosynthesized nanoparticles. In the cellular and molecular part of the ‎recent study, the treated cells with AuNPs were assessed by MTT assay for 48h about the cytotoxicity and anti-‎human ovarian cancer ‎ properties on normal (HUVEC) and ovarian cancer cell lines i.e. PA-1, Caov-3, SW 626, ‎and SK-OV-3. ‎ResultsThe viability of malignant ovarian‎ cell line reduced dose-dependently in the presence of AuNPs. The IC50 of ‎AuNPs were 232‎, ‎204‎, ‎193‎, and ‎288 µg/mL against PA-1, Caov-3, SW 626, and SK-OV-3 cell lines, ‎respectively. In the antioxidant test, the IC50 of AuNPs and BHT against DPPH free radicals were 151 and 142 ‎‎µg/mL, respectively. ‎ConclusionsAfter clinical study, gold nanoparticles containing Nigella damascena leaf aqueous extract may be used to ‎formulate a new chemotherapeutic drug or supplement to treat the several types of human ovarian cancer.‎


2021 ◽  
Author(s):  
Chirukandath Sidhanth ◽  
Sadhanandhan Bindhya ◽  
Aboo Shabna ◽  
Shyama Krishnapriya ◽  
Pacharla Manasa ◽  
...  

LASP-1 was identified as a protein following mass spectrometric analysis of phosphoproteins consequent to signaling by ErbB2 in SKOV-3 cells. It has been previously identified as an oncogene and is located on chromosomal arm 17q 0.76Mb centromeric to ErbB2. It is expressed in serous ovarian cancer cell lines as a 40kDa protein. In SKOV-3 cells, it was phosphorylated and was inhibited by Lapatinib and CP7274714. LASP-1 co-immunoprecipitated with ErbB2 in SKOV-3 cells, suggesting a direct interaction. This interaction and phosphorylation were independent of the kinase activity of ErbB2. Moreover, the binding of LASP-1 to ErbB2 was independent of the  tyrosine phosphorylation of LASP-1. LASP-1 was neither expressed on the surface epithelium of the normal ovary nor in the fallopian tube. It was expressed in 28% of ovarian tumours (n=101) that did not significantly correlate with other clinical factors. In tumours from patients with invasive ductal carcinoma of the breast who had ErbB2 amplification (3+), LASP-1 was expressed in 3/20 (p <0.001). Analysis of the expression of an independent dataset of ovarian and breast tumors from TCGA showed the significant co-occurrence of ErbB2 and LASP-1 (p<0.01). These results suggest that LASP-1 and ErbB2 interaction could be important in the pathogenesis of ovarian cancer.


Cancers ◽  
2021 ◽  
Vol 13 (23) ◽  
pp. 5908
Author(s):  
Adam Neal ◽  
Tiffany Lai ◽  
Tanya Singh ◽  
Neela Rahseparian ◽  
Tristan Grogan ◽  
...  

Ovarian malignancies are a leading cause of cancer-related death for US women. High-grade serous ovarian carcinomas (HGSOCs), the most common ovarian cancer subtype, are aggressive tumors with poor outcomes. Mutations in TP53 are common in HGSOCs, with a subset resulting in p53 aggregation and misregulation. ReACp53 is a peptide designed to inhibit mutant p53 aggregation and has been shown efficacious in targeting cancer cells in vitro and in vivo. As p53 regulates apoptosis, combining ReACp53 with carboplatin represents a logical therapeutic strategy. The efficacy of this combinatorial approach was tested in eight ovarian cancer cell lines and 10 patient HGSOC samples using an in vitro organoid drug assay, with the SynergyFinder tool utilized for calculating drug interactions. Results demonstrate that the addition of ReACp53 to carboplatin enhanced tumor cell targeting in the majority of samples tested, with synergistic effects measured in 2 samples, additivity measured in 14 samples, and antagonism measured in 1 sample. This combination was found to be synergistic in OVCAR3 ovarian cancer cells in vitro through enhanced apoptosis, and survival of mice bearing OVCAR3 intraperitoneal xenografts was extended when treated with the addition of ReACp53 to carboplatin versus carboplatin alone. Results suggest that carboplatin and ReACp53 may be a potential strategy in targeting a subset of HGSOCs.


2021 ◽  
Author(s):  
Tatsuya Kobayashi ◽  
Akira Mitsuhashi ◽  
Piao Hongying ◽  
Masashi Shioya ◽  
Kyoko Nishikimi ◽  
...  

Abstract Purpose: Bexarotene is selectively activates retinoid X recepto, whichr is a commonly used anticancer agent for cutaneous T-cell lymphoma. In this study, we aimed to investigate the anticancer effect of bexarotene and its underlying mechanism in ovarian cancer in vitro.Methods: The ES2 and NIH:OVACAR3 ovarian cancer cell lines were treated with 0, 5, 10, or 20 µM bexarotene. After 24 hours, cell number measurement and lactate dehydrogenase (LDH) cytotoxicity assay were performed. The effect of bexarotene on CDKN1A expression, pyroptosis, and apoptosis were evaluated.Results: Bexarotene reduced cell proliferation in all concentrations in both cells. At concentrations above 10 µM, it increased extracellular LDH activity with cell rupture. In both cells, 10 µM bexarotene treatment increased the CDKN1A mRNA levels and reduced cell cycle related protein expression. In ES2 cells, caspase-4 and GSDME were activated, whereas caspase-3 was not, indicating that bexarotene-induced cell death might be pyroptosis. Conclusion: A clinical setting dose of bexarotene induced cell cycle arrest and cell death through caspase-4–mediated pyroptosis in ovarian cancer cell lines. Thus, bexarotene may serve as a novel therapeutic agent for ovarian cancer.


Sign in / Sign up

Export Citation Format

Share Document