33 results on '"Yudith Ramos Valdes"'
Search Results
2. OVCAR8-STK11KO spheroid from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Single OVCAR8-STK11KO spheroid formation and growth over time as imaged by IncuCyte ZOOM
- Published
- 2023
3. Supplementary Figure S6 from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Overexpression of DUSP4 in FTE and EOC cells does not alter spheroid formation or viability.
- Published
- 2023
4. Supplemental Figures and Legends 1-6 from A Systematic Analysis of Negative Growth Control Implicates the DREAM Complex in Cancer Cell Dormancy
- Author
-
Frederick A. Dick, Gabriel E. DiMattia, Larissa Litovchick, Pirunthan Perampalam, Yudith Ramos-Valdes, and James MacDonald
- Abstract
S1. Cumulative viability score of siRNA knockdowns. S2. Spheroid morphology of control and gene knock down cells. S3. Reattachment of control and gene knockdown spheroid cells after 72 hours. S4. Relative protein expression levels of Dyrk1A and Dyrk1B in cell lines used in this study. S5. Expression and function of CIP/KIPproteins in non-adherent culture. S6. Flow cytometry analysis of p130 depleted cells.
- Published
- 2023
5. Supplementary Figure Legends from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Figure legends for Supplementary Figures S1-6
- Published
- 2023
6. HeyA8 spheroid from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Single HeyA8 spheroid formation and growth over time as imaged by IncuCyte ZOOM
- Published
- 2023
7. Supplementary Figure S5 from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
LKB1 loss does not alter MAPK phosphorylation in EOC cells and spheroids.
- Published
- 2023
8. Supplementary Figure S3 from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Spheroids isolated from tumor cell mouse xenograft ascites.
- Published
- 2023
9. Supplementary Table S2 from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Data from reverse phase protein array analysis (data used to calculate log2-fold change for Figure 6A)
- Published
- 2023
10. HeyA8-STK11KO spheroid from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Single HeyA8-STK11KO spheroid formation and growth over time as imaged by IncuCyte ZOOM
- Published
- 2023
11. Supplementary Figure S4 from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Loss of LKB1 does not alter sensitivity of OVCAR8 cells to several metabolic stresses.
- Published
- 2023
12. Supplementary Figure S1 from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Morphology of STK11KO cell lines in adherent culture.
- Published
- 2023
13. OVCAR8 spheroid from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Single OVCAR8 spheroid formation and growth over time as imaged by IncuCyte ZOOM
- Published
- 2023
14. Supplementary Table S1 from AMPK-Independent LKB1 Activity Is Required for Efficient Epithelial Ovarian Cancer Metastasis
- Author
-
Trevor G. Shepherd, Gabriel E. DiMattia, Yudith Ramos-Valdes, and Adrian Buensuceso
- Abstract
Number of mice with tumor lesions at specific anatomical sites and ascites
- Published
- 2023
15. Loss of LKB1-NUAK1 Signalling Enhances NF-κB Activity in a Spheroid Model of High-Grade Serous Ovarian Cancer
- Author
-
Olga Collins, Trevor G. Shepherd, Jamie Lee Fritz, Adrian Buensuceso, Gabriel E. DiMattia, and Yudith Ramos Valdes
- Subjects
chemistry.chemical_compound ,Signalling ,Chemistry ,Serous ovarian cancer ,NUAK1 ,Cancer research ,Spheroid ,NF-κB - Abstract
High-grade serous ovarian cancer (HGSOC) is an aggressive malignancy often diagnosed at an advanced stage. Although most HGSOC patients respond initially to debulking surgery combined with cytotoxic chemotherapy, many ultimately relapse with platinum-resistant disease. Thus, improving outcomes requires new ways of limiting metastasis and eradicating residual disease. We identified previously that Liver kinase B1 (LKB1) and its substrate NUAK1 are implicated in EOC spheroid cell viability and are required for efficient metastasis in orthotopic mouse models. Here, we sought to identify additional signalling pathways altered in EOC cells due to LKB1 or NUAK1 loss-of-function. Transcriptome analysis revealed that inflammatory signalling mediated by NF-κB transcription factors is hyperactive due to LKB1-NUAK1 loss in HGSOC cells and spheroids. Upregulated NF-κB signalling due to NUAK1 loss suppresses reactive oxygen species (ROS) production and sustains cell survival in spheroids. NF-κB signalling is also activated in HGSOC precursor fallopian tube secretory epithelial cell spheroids, and is further enhanced by NUAK1 loss. Finally, immunohistochemical analysis of OVCAR8 xenograft tumors lacking NUAK1 displayed increased RelB expression and nuclear staining. Our results support the idea that NUAK1 and NF-κB signalling pathways together regulate ROS and inflammatory signalling, supporting cell survival during each step of HGSOC pathogenesis. We propose that their combined inhibition may be efficacious as a novel therapeutic strategy for advanced HGSOC.
- Published
- 2021
16. A novel role for NUAK1 in promoting ovarian cancer metastasis through regulation of fibronectin production in Spheroids
- Author
-
Brett Larsen, Jamie Lee Fritz, Anne-Claude Gingras, Kyle E. Francis, Kevin R. Brown, Trevor G. Shepherd, Karen Colwill, Olga Collins, Parima Saxena, Yudith Ramos Valdes, Robert Rottapel, and Adrian Buensuceso
- Subjects
0301 basic medicine ,Cancer Research ,LKB1 ,Cell ,NUAK1 ,lcsh:RC254-282 ,Article ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,fibronectin ,Ovarian cancer ,medicine ,metastasis ,Anoikis ,Fibronectin ,biology ,Chemistry ,Spheroid ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,ovarian cancer ,Oncology ,Cell culture ,spheroid ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,biology.protein - Abstract
Epithelial ovarian cancer (EOC) has a unique mode of metastasis, where cells shed from the primary tumour, form aggregates called spheroids to evade anoikis, spread through the peritoneal cavity, and adhere to secondary sites. We previously showed that the master kinase Liver kinase B1 (LKB1) is required for EOC spheroid viability and metastasis. We have identified novel (nua) kinase 1 (NUAK1) as a top candidate LKB1 substrate in EOC cells and spheroids using a multiplex inhibitor beads-mass spectrometry approach. We confirmed that LKB1 maintains NUAK1 phosphorylation and promotes its stabilization. We next investigated NUAK1 function in EOC cells. Ectopic NUAK1-overexpressing EOC cell lines had increased adhesion, whereas the reverse was seen in OVCAR8-NUAK1KO cells. In fact, cells with NUAK1 loss generate spheroids with reduced integrity, leading to increased cell death after long-term culture. Following transcriptome analysis, we identified reduced enrichment for cell interaction gene expression pathways in OVCAR8-NUAK1KO spheroids. In fact, the FN1 gene, encoding fibronectin, exhibited a 745-fold decreased expression in NUAK1KO spheroids. Fibronectin expression was induced during native spheroid formation, yet this was completely lost in NUAK1KO spheroids. Co-incubation with soluble fibronectin restored the compact spheroid phenotype to OVCAR8-NUAK1KO cells. In a xenograft model of intraperitoneal metastasis, NUAK1 loss extended survival and reduced fibronectin expression in tumours. Thus, we have identified a new mechanism controlling EOC metastasis, through which LKB1-NUAK1 activity promotes spheroid formation and secondary tumours via fibronectin production.
- Published
- 2020
17. AMPK-independent LKB1 activity is required for efficient epithelial ovarian cancer metastasis
- Author
-
Yudith Ramos-Valdes, Adrian Buensuceso, Trevor G. Shepherd, and Gabriel E. DiMattia
- Subjects
0301 basic medicine ,Cancer Research ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system diseases ,AMP-Activated Protein Kinases ,Carcinoma, Ovarian Epithelial ,Protein Serine-Threonine Kinases ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,AMP-Activated Protein Kinase Kinases ,Downregulation and upregulation ,Mice, Inbred NOD ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Protein kinase A ,skin and connective tissue diseases ,Molecular Biology ,030304 developmental biology ,Ovarian Neoplasms ,0303 health sciences ,Chemistry ,Kinase ,Spheroid ,AMPK ,medicine.disease ,female genital diseases and pregnancy complications ,3. Good health ,030104 developmental biology ,Oncology ,Apoptosis ,Cell culture ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,Heterografts ,Female - Abstract
Epithelial ovarian cancer (EOC) spreads by direct dissemination of malignant cells and multicellular clusters, known as spheroids, into the peritoneum followed by implantation and growth on abdominal surfaces. Using a spheroid model system of EOC metastasis, we discovered that Liver kinase B1 (LKB1), encoded by the STK11 gene, and its canonical substrate AMP-activated protein kinase (AMPK) are activated in EOC spheroids, yet only LKB1 is required for cell survival. We have now generated STK11-knockout cell lines using normal human FT190 cells and three EOC cell lines, OVCAR8, HeyA8, and iOvCa147. STK11KO did not affect growth and viability in adherent culture, but it decreased anchorage-independent growth of EOC cells. EOC spheroids lacking LKB1 had markedly impaired growth and viability, whereas there was no difference in normal FT190 spheroids. To test whether LKB1 loss affects EOC metastasis, we performed intraperitoneal injections of OVCAR8-, HeyA8-, and iOvCa147-STK11KO cells, and respective controls. LKB1 loss exhibited a dramatic reduction on tumor burden and metastatic potential; in particular, OVCAR8-STK11KO tumors had evidence of extensive necrosis, apoptosis, and hypoxia. Interestingly, LKB1 loss did not affect AMPKα phosphorylation in EOC spheroids and tumor xenografts, indicating that LKB1 signaling to support EOC cell survival in spheroids and metastatic tumor growth occurs via other downstream mediators. We identified the dual-specificity phosphatase DUSP4 as a commonly upregulated protein due to LKB1 loss; indeed, DUSP4 knockdown in HeyA8-STK11KO cells partially restored spheroid formation and viability. Implications: LKB1 possesses key tumor-promoting activity independent of downstream AMPK signaling during EOC metastasis.
- Published
- 2019
- Full Text
- View/download PDF
18. Characterization of Mutational Status, Spheroid Formation, and Drug Response of a New Genomically-Stable Human Ovarian Clear Cell Carcinoma Cell Line, 105C
- Author
-
Wonjae Lee, Trevor G. Shepherd, Hal W. Hirte, Hiroaki Itamochi, Bart Kolendowski, Mark S. Carey, Yudith Ramos Valdes, and Gabriel E. DiMattia
- Subjects
epithelial ovarian cancer ,mTOR inhibitor ,DNA Mutational Analysis ,Mice, SCID ,OCCC ,clear cell ,Mutation Rate ,Mice, Inbred NOD ,Chromosomes, Human ,lcsh:QH301-705.5 ,Ovarian Neoplasms ,biology ,TOR Serine-Threonine Kinases ,cell line ,General Medicine ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Phenotype ,Clear cell carcinoma ,Adenocarcinoma ,Female ,Signal Transduction ,DNA Copy Number Variations ,Cell Survival ,Morpholines ,Antineoplastic Agents ,Genomic Instability ,Article ,Cell Line, Tumor ,Spheroids, Cellular ,Cell Adhesion ,medicine ,Animals ,Humans ,ovarian ,cancer ,PTEN ,Gene ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Genome, Human ,medicine.disease ,Xenograft Model Antitumor Assays ,lcsh:Biology (General) ,spheroid ,Cell culture ,Mutation ,Cancer research ,biology.protein ,Proto-Oncogene Proteins c-akt ,Clear cell ,Transcription Factors - Abstract
Ovarian clear cell carcinoma (OCCC) is a rare subtype of gynecological cancer for which well-characterized and authenticated model systems are scarce. We provide an extensive characterization of &lsquo, 105C&rsquo, a cell line generated from an adenocarcinoma of the clear cell histotype using targeted next-generation sequencing, cytogenetic microarrays, along with analyses of AKT/mTOR signaling. We report that that the 105C cell line is a bona fide OCCC cell line, carrying PIK3CA, PTEN, and ARID1A gene mutations, consistent with OCCC, yet maintain a stable genome as reflected by low copy number variation. Unlike KOC-7c, TOV-21G, and RMG-V OCCC lines also mutated for the above genes, the 105C cells do not carry mutations in mismatch repair genes. Importantly, we show that 105C cells exhibit greater resistance to mTOR inhibition and carboplatin treatment compared to 9 other OCCC cell lines in 3D spheroid cultures. This resistance may be attributed to 105C cells remaining dormant in suspension culture which surprisingly, contrasts with several other OCCC lines which continue to proliferate in long-term suspension culture. 105C cells survive xenotransplantation but do not proliferate and metastasize. Collectively, we show that the 105C OCCC cell line exhibits unique properties useful for the pre-clinical investigation of OCCC pathobiology.
- Published
- 2020
19. Spatial and temporal epithelial ovarian cancer cell heterogeneity impacts Maraba virus oncolytic potential
- Author
-
John C. Bell, David F. Stojdl, Trevor G. Shepherd, Milani Sivapragasam, John W. Barrett, Gabriel E. DiMattia, Yudith Ramos Valdes, and Jessica Tong
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Oncolytic virus ,Tumour heterogeneity ,Population ,Resistance ,Context (language use) ,Carcinoma, Ovarian Epithelial ,lcsh:RC254-282 ,Virus ,Maraba virus ,03 medical and health sciences ,Genetic Heterogeneity ,Cell and Developmental Biology ,0302 clinical medicine ,Interferon ,Internal medicine ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Neoplasms, Glandular and Epithelial ,education ,Oncolytic Virotherapy ,Ovarian Neoplasms ,education.field_of_study ,Genetic heterogeneity ,business.industry ,Cancer ,Ascites ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Coculture Techniques ,3. Good health ,Oncolytic Viruses ,030104 developmental biology ,030220 oncology & carcinogenesis ,Anatomy ,business ,High-grade serous ovarian cancer ,medicine.drug ,Research Article - Abstract
Background Epithelial ovarian cancer exhibits extensive interpatient and intratumoral heterogeneity, which can hinder successful treatment strategies. Herein, we investigated the efficacy of an emerging oncolytic, Maraba virus (MRBV), in an in vitro model of ovarian tumour heterogeneity. Methods Four ovarian high-grade serous cancer (HGSC) cell lines were isolated and established from a single patient at four points during disease progression. Limiting-dilution subcloning generated seven additional subclone lines to assess intratumoral heterogeneity. MRBV entry and oncolytic efficacy were assessed among all 11 cell lines. Low-density receptor (LDLR) expression, conditioned media treatments and co-cultures were performed to determine factors impacting MRBV oncolysis. Results Temporal and intratumoral heterogeneity identified two subpopulations of cells: one that was highly sensitive to MRBV, and another set which exhibited 1000-fold reduced susceptibility to MRBV-mediated oncolysis. We explored both intracellular and extracellular mechanisms influencing sensitivity to MRBV and identified that LDLR can partially mediate MRBV infection. LDLR expression, however, was not the singular determinant of sensitivity to MRBV among the HGSC cell lines and subclones. We verified that there were no apparent extracellular factors, such as type I interferon responses, contributing to MRBV resistance. However, direct cell-cell contact by co-culture of MRBV-resistant subclones with sensitive cells restored virus infection and oncolytic killing of mixed population. Conclusions Our data is the first to demonstrate differential efficacy of an oncolytic virus in the context of both spatial and temporal heterogeneity of HGSC cells and to evaluate whether it will constitute a barrier to effective viral oncolytic therapy. Electronic supplementary material The online version of this article (10.1186/s12885-017-3600-2) contains supplementary material, which is available to authorized users.
- Published
- 2017
20. A Systematic Analysis of Negative Growth Control Implicates the DREAM Complex in Cancer Cell Dormancy
- Author
-
Yudith Ramos-Valdes, Pirunthan Perampalam, Gabriel E. DiMattia, James I. MacDonald, Larissa Litovchick, and Frederick A. Dick
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Cell Survival ,Cell Cycle Proteins ,Biology ,Carcinoma, Ovarian Epithelial ,Protein Serine-Threonine Kinases ,Carboplatin ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Tumor Cells, Cultured ,Humans ,DREAM complex ,Viability assay ,Neoplasms, Glandular and Epithelial ,Molecular Biology ,Cell Proliferation ,Ovarian Neoplasms ,Cell Cycle ,Cancer ,Kv Channel-Interacting Proteins ,Cell cycle ,Protein-Tyrosine Kinases ,medicine.disease ,Cell biology ,Repressor Proteins ,Harmine ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Trans-Activators ,Ovarian cancer ,Cancer dormancy - Abstract
Epithelial ovarian cancer (EOC) generates multicellular aggregates called spheroids that detach from the primary tumor and disseminate through ascites. Spheroids possess a number of characteristics of tumor dormancy including withdrawal from the cell cycle and resistance to chemotherapeutics. This report systematically analyzes the effects of RNAi depletion of 21 genes that are known to contribute to negative regulation of the cell cycle in 10 ovarian cancer cell lines. Interestingly, spheroid cell viability was compromised by loss of some cyclin-dependent kinase inhibitors such as p57Kip2, as well as Dyrk1A, Lin52, and E2F5 in most cell lines tested. Many genes essential for EOC spheroid viability are pertinent to the mammalian DREAM repressor complex. Mechanistically, the data demonstrate that DREAM is assembled upon the induction of spheroid formation, which is dependent upon Dyrk1A. Loss of Dyrk1A results in retention of the b-Myb–MuvB complex, elevated expression of DREAM target genes, and increased DNA synthesis that is coincident with cell death. Inhibition of Dyrk1A activity using pharmacologic agents Harmine and INDY compromises viability of spheroids and blocks DREAM assembly. In addition, INDY treatment improves the response to carboplatin, suggesting this is a therapeutic target for EOC treatment. Implications: Loss of negative growth control mechanisms in cancer dormancy lead to cell death and not proliferation, suggesting they are an attractive therapeutic approach. Mol Cancer Res; 15(4); 371–81. ©2016 AACR.
- Published
- 2016
21. Modulation of AKT activity is associated with reversible dormancy in ascites-derived epithelial ovarian cancer spheroids
- Author
-
Teresa Peart, Trevor G. Shepherd, Yudith Ramos Valdes, Rohann J.M. Correa, and Gabriel E. DiMattia
- Subjects
Cancer Research ,endocrine system diseases ,Cell division ,Cell ,Carcinoma, Ovarian Epithelial ,Metastasis ,Peritoneal cavity ,Cell Line, Tumor ,Spheroids, Cellular ,Cell Adhesion ,medicine ,Humans ,Neoplasms, Glandular and Epithelial ,Phosphorylation ,Protein kinase B ,Cell Proliferation ,Ovarian Neoplasms ,Cell growth ,Chemistry ,Spheroid ,Ascites ,General Medicine ,medicine.disease ,female genital diseases and pregnancy complications ,Cell biology ,medicine.anatomical_structure ,Cell culture ,embryonic structures ,Female ,Proto-Oncogene Proteins c-akt - Abstract
Epithelial ovarian cancer (EOC) metastasis is a direct contributor to high recurrence and low survival for patients with this disease. Metastasis in EOC occurs by cell exfoliation from the primary tumor into the fluid-filled peritoneal cavity, persistence of these cells as non-adherent multicellular aggregates or spheroids and reattachment of spheroids to form secondary lesions. We have recovered native spheroids from ascites fluid and demonstrated that EOC cells within these structures exhibit reduced proliferation, yet regain the capacity to attach and reinitiate cell division. To model this process in vitro for further investigation, primary EOC cells from patient peritoneal fluid were cultured under non-adherent conditions. Here we show that these cells naturally form spheroids resembling those observed in ascites. Spheroids exhibit reduced cell proliferation and a protein expression pattern consistent with cellular quiescence: specifically, decreased phospho-AKT and p45/SKP2 with a concomitant increase in p130/RBL2 and p27(Kip1). However, when spheroids are seeded to an adherent surface, reattachment occurs rapidly and is followed by reinitiation of AKT-dependent cell proliferation. These results were strikingly consistent among numerous clinical specimens and were corroborated in the EOC cell line OVCAR3. Therefore, our data reveal that EOC cells become quiescent when forming spheroids, but reactivate proliferative mechanisms upon attachment to a permissive substratum. Overall, this work utilizes a novel in vitro model of EOC metastasis that employs primary human EOC cells and introduces the important concept of reversible dormancy in EOC pathogenesis.
- Published
- 2011
22. Abstract A17: NUAK1 acts as a growth suppressor in epithelial ovarian cancer
- Author
-
Yudith Ramos Valdes, Parima Saxena, Trevor G. Shepherd, Anne-Claude Gingras, Kevin R. Brown, Karen Colwill, Robert Rottapel, Kyle E. Francis, Gabriel E. DiMattia, Olga Collins, and Adrian Buensuceso
- Subjects
Cancer Research ,Cell cycle checkpoint ,Kinase ,Cell growth ,Cell ,NUAK1 ,Biology ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Ovarian carcinoma ,medicine ,Cancer research ,Protein kinase A ,Ovarian cancer - Abstract
Epithelial ovarian cancer (EOC) cells form multicellular aggregates, or spheroids, and enter a dormant state during intraperitoneal metastasis. Dormant spheroids reduce anabolic metabolism and cell proliferation, which are linked to chemo-resistance. Liver kinase B1 (LKB1), encoded by the STK11 gene, is a critical regulator of stress metabolic signaling; LKB1 phosphorylates the downstream substrates AMP-activated protein kinase (AMPK) and AMPK-related kinases (ARKs) to mediate stress signaling. We have demonstrated that LKB1 expression and activity is required for EOC spheroid cell survival, yet knockdown of AMPKα1/α2 has no effect on viability. In addition, we have preliminary data demonstrating that phosphorylated AMPK is still maintained in EOC spheroids generated from CRISPR-mediated STK11-deleted cells, which completely lack LKB1 expression. Taken together, these results implicate the importance of other AMPK-independent effectors of LKB1 signaling in spheroid cell viability. Therefore, to identify critical downstream targets of LKB1-mediated signaling in EOC spheroids, we employed multiplex inhibitor beads/mass spectrometry (MIB/MS) using OVCAR8 STK11-knockout cells and OVCAR8 control cells grown in adherent and spheroid culture conditions. Using this proteomic approach, we identified NUAK1 as the sole ARK out of 12 different family members that is negatively affected by LKB1 loss. In fact, both phosphorylated and total NUAK1 protein expression are decreased in STK11-knockout cells and spheroids. NUAK1 can negatively control cell growth and proliferation by direct regulation of cell cycle checkpoint proteins in several cell systems; however, it has been relatively understudied in EOC. In analysis of the serous ovarian carcinoma data from TCGA, NUAK1 is infrequently altered ( Citation Format: Parima Saxena, Olga Collins, Yudith Ramos Valdes, Adrian Buensuceso, Kyle Francis, Kevin Brown, Karen Colwill, Anne-Claude Gingras, Robert Rottapel, Gabriel E. DiMattia, Trevor G. Shepherd. NUAK1 acts as a growth suppressor in epithelial ovarian cancer. [abstract]. In: Proceedings of the AACR Conference: Addressing Critical Questions in Ovarian Cancer Research and Treatment; Oct 1-4, 2017; Pittsburgh, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(15_Suppl):Abstract nr A17.
- Published
- 2018
23. Abstract A12: The metabolic stress mediator LKB1 is required for ovarian cancer metastasis
- Author
-
Trevor G. Shepherd, Adrian Buensuceso, Gabriel E. DiMattia, Rene Figueredo, and Yudith Ramos Valdes
- Subjects
Cancer Research ,Mediator ,Oncology ,business.industry ,medicine ,Cancer research ,Metabolic Stress ,medicine.disease ,Ovarian cancer ,business ,Metastasis - Abstract
Most epithelial ovarian cancer (EOC) patients with metastatic disease initially respond to cytotoxic chemotherapy, yet almost all will relapse with resistant disease. Thus, improving patient outcomes will require novel approaches to limit metastasis and overcome chemo-resistance. Liver Kinase B1 (LKB1), encoded by the STK11 gene, is a key intracellular regulator of metabolic stress and is considered a putative tumor suppressor in some cancers. However, we have demonstrated that LKB1 is intact and required for EOC cell viability and growth in an in vitro spheroid model of ovarian cancer metastasis. We propose that LKB1 signaling enables malignant EOC cells to maintain viability and survive in metabolically challenging environments like that encountered during intraperitoneal metastasis. To further investigate the therapeutic potential of targeting LKB1 activity in metastatic EOC, we generated STK11-knockout cell lines—normal FT190 cells, and EOC cell lines OVCAR8, HeyA8, and iOvCa147—using CRISPR technology. STK11KO resulted in decreased malignant properties of EOC cells in vitro, including clonogenicity and anchorage-independent growth; however, loss of LKB1 in FT190 cells had no effect on cell proliferation, clonogenicity, or anchorage-independent growth, indicating LKB1 does not likely act as a tumor suppressor in EOC. Loss of LKB1 sensitized EOC cells to the growth-inhibiting effects of specific metabolic stresses. OVCAR8-STK11KO cells were more sensitive to nutrient deprivation in adherent culture, and to carboplatin and paclitaxel treatment in spheroid culture, as compared with OVCAR8 cells. Among the three EOC cell lines, STK11KO yielded variable sensitivity to inhibition of mitochondrial ATP production via oligomycin treatment. Interestingly, STK11KO did not affect induction of AMP-activated protein kinase (AMPK) phosphorylation in EOC spheroids, indicating that metabolic stress signaling to support EOC cell survival in spheroids during metastasis may occur via alternative pathways. In support of our previous knockdown results, EOC spheroids completely lacking LKB1 had markedly impaired growth in suspension culture compared to parental cell controls. In contrast, FT190 spheroids exhibited rapid cell attrition in spheroid culture regardless of LKB1 status. These results indicate that LKB1 may be specifically required in EOC cells to evade anoikis during metastatic spread. Finally, to test directly whether loss of LKB1 activity affects the metastatic potential of EOC cells, we performed intraperitoneal injections of OVCAR8-STK11KO and HeyA8-STK11KO cells with their respective parental cell controls. Loss of LKB1 in both aggressive EOC cell lines exhibited a dramatic reduction on tumor burden. STK11KO significantly decreased the establishment of large, solid tumor masses, reduced adhesion of OVCAR8 tumor nodules, and even changed the metastatic trajectory of HeyA8 cells with evidence of tumor cell growth only as a thin layer on peritoneal walls. Histologic analysis revealed evidence of extensive necrosis in STK11KO tumors, which was likely the major contributor to reduced tumor burden. These results strongly indicate that loss of LKB1 activity abrogates the metabolic stress response necessary during EOC metastasis both in spheroids and establishment of intraperitoneal tumors. Overall, LKB1 or its AMPK-independent signaling mediators represent unique yet very potent therapeutic vulnerabilities in metastatic EOC. Citation Format: Adrian Buensuceso, Yudith R. Valdes, Rene Figueredo, Gabriel E. DiMattia, Trevor G. Shepherd. The metabolic stress mediator LKB1 is required for ovarian cancer metastasis. [abstract]. In: Proceedings of the AACR Conference: Addressing Critical Questions in Ovarian Cancer Research and Treatment; Oct 1-4, 2017; Pittsburgh, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(15_Suppl):Abstract nr A12.
- Published
- 2018
24. Evidence for differential viral oncolytic efficacy in an in vitro model of epithelial ovarian cancer metastasis
- Author
-
Yudith Ramos Valdes, John C. Bell, J. Andrea McCart, Jessica Tong, John W. Barrett, Trevor G. Shepherd, Grant McFadden, David F. Stojdl, and Gabriel E. DiMattia
- Subjects
Cancer Research ,Cancer therapy ,Receptor expression ,viruses ,Myxoma virus ,Biology ,lcsh:RC254-282 ,Virus ,Article ,Metastasis ,03 medical and health sciences ,chemistry.chemical_compound ,Cell and Developmental Biology ,0302 clinical medicine ,medicine ,Pharmacology (medical) ,Cancer models ,030304 developmental biology ,Gynaecological cancer ,0303 health sciences ,Obstetrics and Gynecology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,biology.organism_classification ,Virology ,3. Good health ,Oncolytic virus ,Cell killing ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,embryonic structures ,Molecular Medicine ,Vaccinia ,Anatomy ,Ovarian cancer - Abstract
Epithelial ovarian cancer is unique among most carcinomas in that metastasis occurs by direct dissemination of malignant cells traversing throughout the intraperitoneal fluid. Accordingly, we test new therapeutic strategies using an in vitro three-dimensional spheroid suspension culture model that mimics key steps of this metastatic process. In the present study, we sought to uncover the differential oncolytic efficacy among three different virusesâMyxoma virus, double-deleted vaccinia virus, and Maraba virusâusing three ovarian cancer cell lines in our metastasis model system. Herein, we demonstrate that Maraba virus effectively infects, replicates, and kills epithelial ovarian cancer (EOC) cells in proliferating adherent cells and with slightly slower kinetics in tumor spheroids. Myxoma virus and vaccinia viruses infect and kill adherent cells to a much lesser extent than Maraba virus, and their oncolytic potential is almost completely attenuated in spheroids. Myxoma virus and vaccinia are able to infect and spread throughout spheroids, but are blocked in the final stages of the lytic cycle, and oncolytic-mediated cell killing is reactivated upon spheroid reattachment. Alternatively, Maraba virus has a remarkably reduced ability to initially enter spheroid cells, yet rapidly infects and spreads throughout spheroids generating significant cell killing effects. We show that low-density lipoprotein receptor expression in ovarian cancer spheroids is reduced and this controls efficient Maraba virus binding and entry into infected cells. Taken together, these results are the first to implicate the potential impact of differential viral oncolytic properties at key steps of ovarian cancer metastasis.
- Published
- 2015
25. Beclin-1 Expression is Retained in High-Grade Serous Ovarian Cancer yet is Not Essential for Autophagy Induction In Vitro
- Author
-
Rohann J.M. Correa, Gabriel E. DiMattia, Yudith Ramos Valdes, and Trevor G. Shepherd
- Subjects
Cell Survival ,Apoptosis ,Biology ,medicine.disease_cause ,Biochemistry ,Ovarian tumor ,Cell and Developmental Biology ,Cell Line, Tumor ,Spheroids, Cellular ,Obstetrics and Gynaecology ,medicine ,Autophagy ,Humans ,Ovarian Neoplasms ,Regulation of gene expression ,Gene knockdown ,Research ,Membrane Proteins ,Obstetrics and Gynecology ,Cancer ,BECN1 ,medicine.disease ,Cystadenocarcinoma, Serous ,3. Good health ,Cell biology ,Gene Expression Regulation, Neoplastic ,Spheroid ,Oncology ,Female ,Beclin-1 ,Anatomy ,Apoptosis Regulatory Proteins ,Ovarian cancer ,Carcinogenesis ,High-grade serous ovarian cancer - Abstract
Background Autophagy is a conserved cellular self-digestion mechanism that can either suppress or promote cancer in a context-dependent manner. In ovarian cancer, prevalent mono-allelic deletion of BECN1 (a canonical autophagy-inducer) suggests that autophagy is impaired to promote carcinogenesis and that Beclin-1 is a haploinsufficient tumor suppressor. Nonetheless, autophagy is known to be readily inducible in ovarian cancer cells. We sought to clarify whether Beclin-1 expression is in fact disrupted in ovarian cancer and whether this impacts autophagy regulation. Methods BECN1 expression levels were assessed using The Cancer Genome Atlas (TCGA) datasets from 398 ovarian high-grade serous cystadenocarcinomas (HGSC) and protein immunoblot data from HGSC samples obtained at our institution. Knockdown of BECN1 and other autophagy-related gene expression was achieved using siRNA in established human ovarian cancer cell lines (CaOV3, OVCAR8, SKOV3, and HeyA8) and a novel early-passage, ascites-derived cell line (iOvCa147-E2). LC3 immunoblot, autophagic flux assays, transmission electron microscopy and fluorescence microscopy were used to assess autophagy. Results We observed prevalent mono-allelic BECN1 gene deletion (76 %) in TCGA tumors, yet demonstrate for the first time that Beclin-1 protein expression remains relatively unaltered in these and additional samples generated at our institution. Surprisingly, efficient siRNA-mediated Beclin-1 knockdown did not attenuate autophagy induction, whereas knockdown of other autophagy-related genes blocked the process. Beclin-1 knockdown instead decreased cell viability without inducing apoptosis. Conclusions Taken together, these data demonstrate that despite its sustained expression, Beclin-1 is dispensable for autophagy induction in ovarian tumor cells in vitro, yet may be retained to promote cell viability by a mechanism independent of autophagy or apoptosis regulation. Overall, this work makes novel observations about tumor expression of Beclin-1 and challenges the accepted understanding of its role in regulating autophagy in ovarian cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13048-015-0182-y) contains supplementary material, which is available to authorized users.
- Published
- 2015
26. Intact LKB1 activity is required for survival of dormant ovarian cancer spheroids
- Author
-
Gabriel E. DiMattia, Rohann J.M. Correa, Yudith Ramos Valdes, Jacob McGee, Michel Prefontaine, Trevor G. Shepherd, Akira Sugimoto, Monique Bertrand, Elena N. Fazio, and Teresa Peart
- Subjects
AMPK ,Pathology ,medicine.medical_specialty ,LKB1 ,endocrine system diseases ,Cell Survival ,Cell ,STK11 ,AMP-Activated Protein Kinases ,Carcinoma, Ovarian Epithelial ,Protein Serine-Threonine Kinases ,Transfection ,Metastasis ,03 medical and health sciences ,Cell and Developmental Biology ,0302 clinical medicine ,AMP-Activated Protein Kinase Kinases ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Humans ,Neoplasms, Glandular and Epithelial ,Phosphorylation ,Protein kinase A ,030304 developmental biology ,Ovarian Neoplasms ,0303 health sciences ,tumour cell dormancy ,business.industry ,Kinase ,medicine.disease ,3. Good health ,medicine.anatomical_structure ,ovarian cancer ,Oncology ,spheroid ,030220 oncology & carcinogenesis ,Cancer cell ,embryonic structures ,Cancer research ,Female ,Anatomy ,business ,Ovarian cancer ,Signal Transduction ,Research Paper - Abstract
// Teresa Peart 1, 2 , Yudith Ramos Valdes 1 , Rohann J. M. Correa 1, 3 , Elena Fazio 1, 2 , Monique Bertrand 1, 4, 5 , Jacob McGee 1, 4 , Michel Prefontaine 1, 4 , Akira Sugimoto 1, 4, 5 , Gabriel E. DiMattia 1, 3, 4, 5 , Trevor G. Shepherd 1, 2, 4, 5 1 Translational Ovarian Cancer Research Program, London Regional Cancer Program, London, Ontario, Canada 2 Departments of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada 3 Departments of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada 4 Departments of Obstetrics & Gynaecology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada 5 Departments of Oncology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada Correspondence to: Trevor G. Shepherd, e-mail: tshephe6@uwo.ca Keywords: ovarian cancer, spheroid, LKB1, AMPK, tumour cell dormancy Received: May 14, 2015 Accepted: May 23, 2015 Published: June 05, 2015 ABSTRACT Metastatic epithelial ovarian cancer (EOC) cells can form multicellular spheroids while in suspension and disperse directly throughout the peritoneum to seed secondary lesions. There is growing evidence that EOC spheroids are key mediators of metastasis, and they use specific intracellular signalling pathways to control cancer cell growth and metabolism for increased survival. Our laboratory discovered that AKT signalling is reduced during spheroid formation leading to cellular quiescence and autophagy, and these may be defining features of tumour cell dormancy. To further define the phenotype of EOC spheroids, we have initiated studies of the Liver kinase B1 (LKB1)-5′-AMP-activated protein kinase (AMPK) pathway as a master controller of the metabolic stress response. We demonstrate that activity of AMPK and its upstream kinase LKB1 are increased in quiescent EOC spheroids as compared with proliferating adherent EOC cells. We also show elevated AMPK activity in spheroids isolated directly from patient ascites. Functional studies reveal that treatment with the AMP mimetic AICAR or allosteric AMPK activator A-769662 led to a cytostatic response in proliferative adherent ovarian cancer cells, but they fail to elicit an effect in spheroids. Targeted knockdown of STK11 by RNAi to reduce LKB1 expression led to reduced viability and increased sensitivity to carboplatin treatment in spheroids only, a phenomenon which was AMPK-independent. Thus, our results demonstrate a direct impact of altered LKB1-AMPK signalling function in EOC. In addition, this is the first evidence in cancer cells demonstrating a pro-survival function for LKB1, a kinase traditionally thought to act as a tumour suppressor.
- Published
- 2015
27. Combination of AKT inhibition with autophagy blockade effectively reduces ascites-derived ovarian cancer cell viability
- Author
-
Rohann J.M. Correa, Gabriel E. DiMattia, Michel Prefontaine, Akira Sugimoto, Monique Bertrand, Elena N. Fazio, Teresa Peart, Trevor G. Shepherd, Yudith Ramos Valdes, and Jacob McGee
- Subjects
Cancer Research ,Programmed cell death ,Benzylamines ,endocrine system diseases ,Cell Survival ,Original Manuscript ,Antineoplastic Agents ,Pharmacology ,Biology ,Inhibitory Concentration 50 ,Downregulation and upregulation ,Allosteric Regulation ,Cell Line, Tumor ,Quinoxalines ,Spheroids, Cellular ,medicine ,Autophagy ,Humans ,Viability assay ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Ovarian Neoplasms ,Ascites ,Chloroquine ,Drug Synergism ,General Medicine ,medicine.disease ,female genital diseases and pregnancy complications ,Cell culture ,Drug Resistance, Neoplasm ,Quinazolines ,Female ,Drug Screening Assays, Antitumor ,Ovarian cancer ,Proto-Oncogene Proteins c-akt - Abstract
Recent genomics analysis of the high-grade serous subtype of epithelial ovarian cancer (EOC) show aberrations in the phosphatidylinositol 3-kinase (PI3K)/AKT pathway that result in upregulated signaling activity. Thus, the PI3K/AKT pathway represents a potential therapeutic target for aggressive high-grade EOC. We previously demonstrated that treatment of malignant ascites-derived primary human EOC cells and ovarian cancer cell lines with the allosteric AKT inhibitor Akti-1/2 induces a dormancy-like cytostatic response but does not reduce cell viability. In this report, we show that allosteric AKT inhibition in these cells induces cytoprotective autophagy. Inhibition of autophagy using chloroquine (CQ) alone or in combination with Akti-1/2 leads to a significant decrease in viable cell number. In fact, Akti-1/2 sensitizes EOC cells to CQ-induced cell death by exhibiting markedly reduced EC50 values in combination-treated cells compared with CQ alone. In addition, we evaluated the effects of the novel specific and potent autophagy inhibitor-1 (Spautin-1) and demonstrate that Spautin-1 inhibits autophagy in a Beclin-1-independent manner in primary EOC cells and cell lines. Multicellular EOC spheroids are highly sensitive to Akti-1/2 and CQ/Spautin-1 cotreatments, but resistant to each agent alone. Indeed, combination index analysis revealed strong synergy between Akti-1/2 and Spautin-1 when both agents were used to affect cell viability; Akti-1/2 and CQ cotreatment also displayed synergy in most samples. Taken together, we propose that combination AKT inhibition and autophagy blockade would prove efficacious to reduce residual EOC cells for supplying ovarian cancer recurrence. © The Author 2014. Published by Oxford University Press. All rights reserved.
- Published
- 2014
28. Abstract B51: Stress management: LKB1 controls growth and survival of dormant epithelial ovarian cancer spheroid cells
- Author
-
Gabriel E. DiMattia, Meera Shah, Yudith Ramos Valdes, Teresa Peart, and Trevor G. Shepherd
- Subjects
Cancer Research ,Programmed cell death ,endocrine system diseases ,Cell growth ,Biology ,medicine.disease ,Metastasis ,Oncology ,Cell culture ,Cancer cell ,Immunology ,medicine ,Cancer research ,Viability assay ,Signal transduction ,Ovarian cancer - Abstract
Aberrant cell metabolism is rapidly establishing itself as a critical hallmark of human malignancies. Cancer cells are faced with huge metabolic demands to support rapid tumor growth, yet are commonly starved for nutrients. In response, cancer cells hijack alternative signaling pathways during these times of energy and metabolic stress to sustain viability. Metastatic epithelial ovarian cancer (EOC) cells are faced with additional stressors during transcoelomic spread, such as detachment from a matrix substratum and inhospitable conditions in peritoneal fluid. However, EOC cells have a natural ability to aggregate when in suspension to form multicellular aggregates, or spheroids, which supports a survival advantage for cells when transiting the peritoneal space during metastatic progression. Therefore, we utilize an in vitro spheroid culture model system to investigate signaling pathways altered in EOC cells that may be implicated in ovarian cancer pathobiology and promote metastasis. For example, we have shown that spheroid formation induces cellular quiescence and autophagy, two disparate processes which promote EOC cell survival and resistance to platinum-based chemotherapeutics. In addition, we discovered that EOC spheroids have significantly reduced mitochondrial activity and ATP levels compared with matched proliferating adherent cells. Liver kinase B1 (LKB1) acts as a chief responder to intracellular stress due to reduced energy and nutrients by eliciting general growth suppression during these starvation-like conditions. Thus, we hypothesized that LKB1 activity is increased in EOC spheroids to promote tumor cell dormancy and cell survival. Although the STK11 gene encoding LKB1 is heterozygously deleted in 84% of serous ovarian tumors, we demonstrate that almost all ovarian tumor cells and established EOC cell lines retain intact LKB1 expression. In fact, LKB1 protein expression increases when EOC cells form spheroids and this coincides with induced serine-428 phosphorylation, a modification that is required for LKB1 growth suppressive activity. To address the potential functional requirement of LKB1 in EOC spheroids, we first performed transient knockdown of STK11 to block LKB1 expression. Indeed, reduced LKB1 renders spheroids susceptible to cell death and increases sensitivity to carboplatin. Next, we utilized Cas9-mediated genome editing of the STK11 locus to completely ablate LKB1 expression in HEYA8 and OVCAR8 cells. STK11-knockout HEYA8 and OVCAR8 cells yielded significantly decreased spheroid number and viability compared with parental cell lines. In a reciprocal fashion, forced re-expression of LKB1 in CaOV3 and TOV21G cells—two EOC cell lines which harbor inactivating STK11 mutations—reduces cell growth and colony formation in proliferating adherent culture. Proliferation and dispersion of CaOV3-LKB1 and TOV21G-LKB1 cells from re-attached spheroids are also reduced. Taken together, LKB1 has growth suppressive effects in EOC cells, yet it serves the additional purpose to promote cell survival in spheroids. The canonical target of LKB1 is AMP-activated protein kinase (AMPK), which is thought to elicit the majority of LKB1 growth suppressive effects during stress metabolism signaling. As expected, AMPK phosphorylation at threonine-172 is also induced upon spheroid formation. Using STK11-knockout EOC cells, however, we demonstrate that LKB1 is not required to phosphorylate AMPK in spheroids; more importantly, knockdown of PRKAA1 encoding the catalytic alpha-subunit of AMPK has no effect on spheroid cell viability. Thus, our results suggest that LKB1 utilizes alternative mechanisms to regulate the dormancy phenotype in EOC spheroids. Future work will entail direct experiments testing whether LKB1-mediated stress metabolism signaling has the potential to promote EOC metastasis and recurrence of chemo-resistant disease. Citation Format: Trevor G. Shepherd, Yudith Ramos Valdes, Teresa Peart, Meera Shah, Gabriel E. DiMattia. Stress management: LKB1 controls growth and survival of dormant epithelial ovarian cancer spheroid cells. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: Exploiting Vulnerabilities; Oct 17-20, 2015; Orlando, FL. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(2 Suppl):Abstract nr B51.
- Published
- 2016
29. BMP signalling controls the malignant potential of ascites-derived human epithelial ovarian cancer spheroids via AKT kinase activation
- Author
-
Trevor G. Shepherd, Teresa Peart, Yudith Ramos Valdes, Rohann J.M. Correa, and Gabriel E. DiMattia
- Subjects
Cancer Research ,Cellular pathology ,animal structures ,Receptor expression ,Biology ,Carcinoma, Ovarian Epithelial ,Bone morphogenetic protein ,Spheroids, Cellular ,Cell Adhesion ,Tumor Cells, Cultured ,Humans ,Neoplasms, Glandular and Epithelial ,Phosphorylation ,Cell adhesion ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Glycoproteins ,Ovarian Neoplasms ,Ascites ,General Medicine ,BMPR2 ,Cell biology ,Enzyme Activation ,Pyrimidines ,Oncology ,embryonic structures ,Bone Morphogenetic Proteins ,Intercellular Signaling Peptides and Proteins ,Pyrazoles ,Signal transduction ,Carrier Proteins ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Epithelial ovarian cancer (EOC) cells have the ability to form multi-cellular aggregates in malignant ascites which dramatically alters cell signalling, survival, and metastatic potential. Herein, we demonstrate that patient ascites-derived EOC cells down-regulate endogenous bone morphogenetic protein (BMP) signalling by decreasing BMP ligand expression when grown in suspension culture to form spheroids. Enforced BMP signalling in these cells via constitutively-active BMP type I ALK3(QD) receptor expression causes the formation of smaller, more loosely-aggregated spheroids. Additionally, ALK3(QD)-expressing spheroids have an increased rate of adhesion and dispersion upon reattachment to substratum. Inhibition of endogenous BMP signalling using recombinant Noggin or small molecule inhibitor LDN-193189, on the other hand, opposed these phenotypic changes. To identify potential targets that impact the phenotype of EOC spheroids due to activated BMP signalling, we performed genome-wide expression analyses using Affymetrix arrays. Using the online Connectivity Map resource, the BMP signalling gene expression signature revealed that the AKT pathway is induced by activated BMP signalling in EOC cells; this finding was further validated by phospho-AKT immuno-blotting. In fact, treatment of EOC spheroids with an AKT inhibitor, Akti-1/2, reduced BMP-stimulated cell dispersion during reattachment as compared to controls. Thus, we have identified AKT as being one important downstream component of activated BMP signalling on EOC spheroid pathobiology, which may have important implications on the metastatic potential of this malignancy.
- Published
- 2011
30. Human stanniocalcin-1 or -2 expressed in mice reduces bone size and severely inhibits cranial intramembranous bone growth
- Author
-
Jennifer L. Johnston, Frank Beier, Yudith Ramos-Valdes, Gabriel E. DiMattia, Lee-Anne Stanton, and Sadia Ladhani
- Subjects
Genetically modified mouse ,Male ,medicine.medical_specialty ,Transgene ,Gene Expression ,Calvaria ,Mice, Transgenic ,Biology ,Mice ,Osteogenesis ,Pregnancy ,Internal medicine ,Genetics ,medicine ,STC1 ,Animals ,Humans ,Phosphate Transport Proteins ,Growth Plate ,Intramembranous bone growth ,DNA Primers ,Glycoproteins ,Bone Development ,Base Sequence ,Skull ,Cranial Sutures ,Chondrogenesis ,Recombinant Proteins ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,Intramembranous ossification ,MEPE ,Mice, Inbred CBA ,Intercellular Signaling Peptides and Proteins ,Animal Science and Zoology ,Female ,Agronomy and Crop Science ,Biotechnology - Abstract
Stanniocalcin-1 (STC1) and -2 (STC2) are highly related, secreted, homodimeric glycoproteins that are significantly upregulated by different forms of stress including high phosphate levels. Transgenic mice that constitutively express either human STC1 or STC2 exhibit intra-uterine growth restriction and permanent post-natal growth retardation. STC1 is expressed in chondrocytic and osteoblastic cells during murine development and can enhance differentiation of calvarial cells in culture. Therefore, there is mounting evidence that stanniocalcins (STCs) modulate bone development in vivo. To further define the effects of stanniocalcins on skeletal development, we performed a series of measurements on components of the axial, appendicular, and cranial skeleton in transgenic and wildtype mice. We show that skeletal growth is retarded and that the intramembranous bones of the cranium exhibit a particularly severe delay in suture closure. The posterior frontal suture remains patent throughout the lifetime of human STC1 and STC2 transgenic mice. We did not observe significant effects on chondrogenesis: however, calvarial cells exhibited reduced viability, proliferation and delayed differentiation, indicating that developing osteoblasts are particularly sensitive to the levels of STCs. Given the evidence linking STC1 to cellular phosphate homeostasis, we assessed the expression of a variety of phosphate regulators in transgenic and wildtype calvarial cells and found significantly lower levels of Mepe, Dmp1, Sfrp4 in transgenic cells without a change in Pit1 or Pit2. Collectively these data support a direct regulatory role for STCs in osteoblasts and suggest that overexposure to these factors inhibits normal skeletal development without significant changes in patterning.
- Published
- 2009
31. Stanniocalcin 2 expression is regulated by hormone signalling and negatively affects breast cancer cell viability in vitro
- Author
-
Gabriel E. DiMattia, Sanda Raulic, and Yudith Ramos-Valdes
- Subjects
medicine.medical_specialty ,Cell Survival ,Endocrinology, Diabetes and Metabolism ,Breast Neoplasms ,Tretinoin ,Biology ,Paracrine signalling ,Mice ,Endocrinology ,Breast cancer ,Downregulation and upregulation ,Internal medicine ,Cell Line, Tumor ,medicine ,STC1 ,Animals ,Humans ,Autocrine signalling ,Transcription factor ,Cell Proliferation ,Glycoproteins ,Estradiol ,Cell growth ,Mammary Neoplasms, Experimental ,medicine.disease ,Gene Expression Regulation ,Hormone receptor ,Intercellular Signaling Peptides and Proteins ,Female ,Signal Transduction - Abstract
Stanniocalcin 1 (STC1) and STC2 are secreted, homodimeric glycoproteins that share 30% amino acid sequence identity. Breast tumour gene profiling studies have demonstrated significantly upregulated STC2 expression in hormone-responsive positive breast tumours; therefore, the purpose of this study was to investigate STC2 hormonal regulation and function in breast cancer cells. Here we report that STC2 is expressed in a number of human breast cancer cell lines, regardless of their oestrogen (E2) and progesterone (P4) receptor status, and its expression is readily detectable in human and mouse mammary gland tumours. Besides E2, retinoic acid (RA) and P4 play an important role in the regulation of STC2 expression, not only in MCF-7 but also in other breast cancer and non-breast cell lines. The expression of the related hormone, STC1, is not affected by the above hormones in breast and endometrial cancer cell lines implying a fundamental difference in regulation in cancer cell lines. The induction of STC2 expression by E2 and RA occurs at the transcriptional level but through intermediary transcription factors. The STC2 proximal promoter region is not responsible for hormonal induction, but exhibits a high basal transcriptional activity. Constitutive STC2 expression in human breast cancer cell lines resulted in significant impairment of cell growth, migration and cell viability after serum withdrawal. In conclusion, STC2 is a downstream target of E2, P4 and RA signalling pathways. In hormone receptor negative cell lines it can function in a paracrine/autocrine fashion to reduce cell proliferation.
- Published
- 2008
32. Abstract A45: Beclin1-independent autophagy promotes the survival of metastatic ovarian cancer cells
- Author
-
Teresa Peart, Jacob McGee, Rohann J.M. Correa, Akira Sugimoto, Michel Prefontaine, Monique Bertrand, Trevor G. Shepherd, Elena N. Fazio, Yudith Ramos-Valdes, and Gabriel E. DiMattia
- Subjects
Cancer Research ,ATG5 ,Autophagy ,Biology ,medicine.disease ,Metastasis ,Cell biology ,Oncology ,Downregulation and upregulation ,medicine ,Ovarian cancer ,Protein kinase B ,MAP1LC3B ,PI3K/AKT/mTOR pathway - Abstract
Epithelial ovarian cancer (EOC) metastasis proceeds by an intra-peritoneal route, involving detachment of cells from the primary tumor and dissemination throughout the peritoneal cavity as multicellular aggregates, or spheroids. Herein, we demonstrate that ascites-derived cells upregulate the process of macroautophagy (autophagy), a highly-conserved self-digestion mechanism that functions to mitigate cellular stresses. Both non-adherent culture of these cells to form in vitro spheroids as well as exposure to an allosteric AKT inhibitor stimulated this process, as evidenced by Western blot and immunofluorescence staining for the autophagy marker MAP1LC3B (LC3). Likewise, expression of an eGFP-labeled LC3 protein revealed a shift from diffuse to ‘punctate’ cytoplasmic localization, demonstrating the formation of intracellular autophagic vesicles (autophagosomes). Additionally, we performed transmission electron microscopy and autophagy flux assays, confirming that autophagic degradation proceeded to completion. To determine the role of autophagy upregulation, we attempted to block this process using siRNAs targeting critical autophagy-related (ATG) genes. Interestingly, we found that depletion of Beclin1/ATG6 had no effect on autophagy, despite its role as a canonical inducer of the process. Conversely, depletion of the autophagy regulators ATG5 & ATG7 efficiently blocked LC3 processing and autophagosome formation. Likewise, autophagy was also blocked pharmacologically using the classical inhibitor Chloroquine and the novel agent Spautin-1 (the first targeted autophagy inhibitor to be published). Autophagy blockade by any of these mechanisms reduced EOC cell viability in both adherent and spheroid cultures, suggesting that the upregulation of this process serves to promote cell survival. Moreover, when autophagy blockade was combined with AKT inhibition, a synergistic reduction in cell survival was observed based on combination index analysis. Taken together, this work describes spheroid formation as a stimulus for a non-canonical, Beclin1-independent autophagy program, but also reveals autophagy to be a promoter of tumor cell survival during ovarian cancer metastasis, particularly in cells subjected to AKT/mTOR inhibition. In light of these findings, we propose that clinical autophagy inhibition could enhance the effectiveness of current or investigational ovarian cancer therapies. Citation Format: Rohann Correa, Yudith Ramos-Valdes, Teresa Peart, Elena Fazio, Monique Bertrand, Jacob McGee, Michel Préfontaine, Akira Sugimoto, Trevor G. Shepherd, Gabriel E. DiMattia. Beclin1-independent autophagy promotes the survival of metastatic ovarian cancer cells. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: From Concept to Clinic; Sep 18-21, 2013; Miami, FL. Philadelphia (PA): AACR; Clin Cancer Res 2013;19(19 Suppl):Abstract nr A45.
- Published
- 2013
33. Abstract A47: The LKB1-AMPK pathway mediates the metabolic stress response of dormant ovarian cancer spheroids
- Author
-
Akira Sugimoto, Monique Bertrand, Gabriel E. DiMattia, Yudith Ramos-Valdes, Michel Prefontaine, Teresa Peart, Elena N. Fazio, Trevor G. Shepherd, and Jacob McGee
- Subjects
Cancer Research ,Programmed cell death ,endocrine system diseases ,Cell ,Cancer ,AMPK ,Biology ,medicine.disease ,Cytostasis ,female genital diseases and pregnancy complications ,Metastasis ,Cell biology ,medicine.anatomical_structure ,Oncology ,embryonic structures ,medicine ,Ovarian cancer ,Protein kinase B - Abstract
Metastatic epithelial ovarian cancer (EOC) cells can form multicellular spheroids while in suspension and disperse throughout the peritoneum via ascites to seed secondary lesions. We hypothesize that EOC spheroids are key mediators of metastasis, which use specific signaling pathways to alter cell metabolism for increased survival. Our lab discovered that AKT signaling is reduced during spheroid formation leading to cellular quiescence and autophagy. Given the induction of quiescence and autophagy in EOC spheroids, we are studying the 5′-AMP-activated protein kinase (AMPK) pathway as a master controller of the metabolic stress response and dormant phenotype of EOC spheroids. We demonstrate AMPK activity and its upstream kinase LKB1 are increased in quiescent EOC spheroids compared with proliferating adherent EOC cells. We also show elevated AMPK activity in spheroids isolated directly from patient ascites. Targeted knockdown of STK11, encoding LKB1, reduces cell viability in ovarian cancer cell line spheroids; PRKAA1 (AMPKα1) knockdown has little to no effect on EOC cell or spheroid viability. Combination of STK11 knockdown with carboplatin treatment leads to a synergistic enhancement in EOC spheroid cell death. In contrast, AICAR treatment of proliferating adherent ovarian cancer cell lines and primary EOC cells induces AMPK activity and causes either cytostasis or cell death. In addition, AICAR treatment of spheroids during reattachment decreases the dispersion capacity of migrating EOC cells. These results offer a glimpse of the potential important contributions of LKB1-AMPK pathway in stress signaling related to EOC cell survival during metastasis. In addition, downstream effectors of upregulated LKB1-AMPK signalling may provide additional mechanisms by which EOC evades chemotherapy. It is foreseeable that these findings will allow us to identify new therapeutic targets within this pathway critical to EOC progression for ultimate translation to the clinic. Citation Format: Teresa Peart, Elena Fazio, Yudith Ramos-Valdes, Monique Bertrand, Jacob McGee, Michel Prefontaine, Akira Sugimoto, Gabriel E. DiMattia, Trevor G. Shepherd. The LKB1-AMPK pathway mediates the metabolic stress response of dormant ovarian cancer spheroids. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: From Concept to Clinic; Sep 18-21, 2013; Miami, FL. Philadelphia (PA): AACR; Clin Cancer Res 2013;19(19 Suppl):Abstract nr A47.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.