Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biomaterials ; 297: 122121, 2023 06.
Article in English | MEDLINE | ID: mdl-37075613

ABSTRACT

Tumour-associated macrophages are linked with poor prognosis and resistance to therapy in Hodgkin lymphoma; however, there are no suitable preclinical models to identify macrophage-targeting therapeutics. We used primary human tumours to guide the development of a mimetic cryogel, wherein Hodgkin (but not Non-Hodgkin) lymphoma cells promoted primary human macrophage invasion. In an invasion inhibitor screen, we identified five drug hits that significantly reduced tumour-associated macrophage invasion: marimastat, batimastat, AS1517499, ruxolitinib, and PD-169316. Importantly, ruxolitinib has demonstrated recent success in Hodgkin lymphoma clinical trials. Both ruxolitinib and PD-169316 (a p38 mitogen-activated protein kinase (p38 MAPK) inhibitor) decreased the percent of M2-like macrophages; however, only PD-169316 enhanced the percentage of M1-like macrophages. We validated p38 MAPK as an anti-invasion drug target with five additional drugs using a high-content imaging platform. With our biomimetic cryogel, we modeled macrophage invasion in Hodgkin lymphoma and then used it for target discovery and drug screening, ultimately identifying potential future therapeutics.


Subject(s)
Hodgkin Disease , Tumor-Associated Macrophages , Humans , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Hodgkin Disease/drug therapy , Hodgkin Disease/pathology , Cryogels , p38 Mitogen-Activated Protein Kinases/metabolism , Extracellular Matrix/metabolism
2.
ACS Med Chem Lett ; 14(2): 199-210, 2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36793435

ABSTRACT

B cell lymphoma 6 (BCL6), a highly regulated transcriptional repressor, is deregulated in several forms of non-Hodgkin lymphoma (NHL), most notably in diffuse large B-cell lymphoma (DLBCL). The activities of BCL6 are dependent on protein-protein interactions with transcriptional co-repressors. To find new therapeutic interventions addressing the needs of patients with DLBCL, we initiated a program to identify BCL6 inhibitors that interfere with co-repressor binding. A virtual screen hit with binding activity in the high micromolar range was optimized by structure-guided methods, resulting in a novel and highly potent inhibitor series. Further optimization resulted in the lead candidate 58 (OICR12694/JNJ-65234637), a BCL6 inhibitor with low nanomolar DLBCL cell growth inhibition and an excellent oral pharmacokinetic profile. Based on its overall favorable preclinical profile, OICR12694 is a highly potent, orally bioavailable candidate for testing BCL6 inhibition in DLBCL and other neoplasms, particularly in combination with other therapies.

3.
J Mol Biol ; 433(23): 167294, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34662547

ABSTRACT

Activating mutations in the epidermal growth factor receptor (EGFR) are common driver mutations in non-small cell lung cancer (NSCLC). First, second and third generation EGFR tyrosine kinase inhibitors (TKIs) are effective at inhibiting mutant EGFR NSCLC, however, acquired resistance is a major issue, leading to disease relapse. Here, we characterize a small molecule, EMI66, an analog of a small molecule which we previously identified to inhibit mutant EGFR signalling via a novel mechanism of action. We show that EMI66 attenuates receptor tyrosine kinase (RTK) expression and signalling and alters the electrophoretic mobility of Coatomer Protein Complex Beta 2 (COPB2) protein in mutant EGFR NSCLC cells. Moreover, we demonstrate that EMI66 can alter the subcellular localization of EGFR and COPB2 within the early secretory pathway. Furthermore, we find that COPB2 knockdown reduces the growth of mutant EGFR lung cancer cells, alters the post-translational processing of RTKs, and alters the endoplasmic reticulum (ER) stress response pathway. Lastly, we show that EMI66 treatment also alters the ER stress response pathway and inhibits the growth of mutant EGFR lung cancer cells and organoids. Our results demonstrate that targeting of COPB2 with EMI66 presents a viable approach to attenuate mutant EGFR signalling and growth in NSCLC.


Subject(s)
Coatomer Protein/genetics , Coatomer Protein/metabolism , Drug Discovery , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Drug Discovery/methods , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects
4.
Nat Chem Biol ; 16(5): 577-586, 2020 05.
Article in English | MEDLINE | ID: mdl-32094923

ABSTRACT

Receptor tyrosine kinases (RTKs) are transmembrane receptors of great clinical interest due to their role in disease. Historically, therapeutics targeting RTKs have been identified using in vitro kinase assays. Due to frequent development of drug resistance, however, there is a need to identify more diverse compounds that inhibit mutated but not wild-type RTKs. Here, we describe MaMTH-DS (mammalian membrane two-hybrid drug screening), a live-cell platform for high-throughput identification of small molecules targeting functional protein-protein interactions of RTKs. We applied MaMTH-DS to an oncogenic epidermal growth factor receptor (EGFR) mutant resistant to the latest generation of clinically approved tyrosine kinase inhibitors (TKIs). We identified four mutant-specific compounds, including two that would not have been detected by conventional in vitro kinase assays. One of these targets mutant EGFR via a new mechanism of action, distinct from classical TKI inhibition. Our results demonstrate how MaMTH-DS is a powerful complement to traditional drug screening approaches.


Subject(s)
High-Throughput Screening Assays/methods , Protein Kinase Inhibitors/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line , Cell Line, Tumor , DNA Nucleotidyltransferases/genetics , Drug Discovery , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Genes, Reporter , Humans , Luciferases/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Phosphorylation/drug effects , Reproducibility of Results , Small Molecule Libraries/pharmacology , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
6.
J Ovarian Res ; 7: 123, 2014 Dec 21.
Article in English | MEDLINE | ID: mdl-25528264

ABSTRACT

OBJECTIVE: Previously, it has been shown that KIF14 mRNA is overexpressed in ovarian cancer (OvCa), regardless of histological subtype. KIF14 levels are independently predictive of poor outcome and increased rates of recurrence in serous OvCa patients. Furthermore, it has been shown that KIF14 also controls the in vivo tumorigenicity of OvCa cell lines. In this study, we evaluate the potential of KIF14 as a therapeutic target through selective inhibition of KIF14 in primary high-grade serous patient-derived OvCa cells. METHODS: To assess the dependence of primary serous OvCa cultures on KIF14, protein levels in 11 prospective high grade serous ovarian cancer samples were increased (KIF14 overexpression by transfection) or decreased (anti-KIF14 shRNA) in vitro, and proliferative capacity, anchorage independence and xenograft growth were assessed. RESULTS: Seven of eleven samples demonstrated increased/decreased in vitro proliferation in response to KIF14 overexpression/knockdown, respectively. When examining in vitro tumorigenicity (colony formation) and in vivo growth (subcutaneous xenografts) in response to KIF14 manipulation, none of the samples demonstrated growth in soft agar (11 samples), or xenograft growth (4 samples). CONCLUSIONS: Although primary high-grade serous OvCa cells may depend on KIF14 for in vitro proliferation we were unable to demonstrate a role for KIF14 on tumorigenicity or develop an in vivo model for assessment. We have, however developed an effective in vitro method to evaluate the effect of target gene manipulation on the proliferative capacity of primary OvCa cultures.


Subject(s)
Kinesins/physiology , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Oncogene Proteins/physiology , Ovarian Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Female , Gene Expression , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/pathology , Primary Cell Culture
7.
PLoS One ; 9(3): e91540, 2014.
Article in English | MEDLINE | ID: mdl-24626475

ABSTRACT

KIF14 (kinesin family member 14) is a mitotic kinesin and an important oncogene in several cancers. Tumor KIF14 expression levels are independently predictive of poor outcome, and in cancer cells KIF14 can modulate metastatic behavior by maintaining appropriate levels of cell adhesion and migration proteins at the cell membrane. Thus KIF14 is an exciting potential therapeutic target. Understanding KIF14's regulation in cancer cells is crucial to the development of effective and selective therapies to block its tumorigenic function(s). We previously determined that close to 30% of serous ovarian cancers (OvCa tumors) exhibit low-level genomic gain, indicating one mechanism of KIF14 overexpression in tumors. We now report on transcriptional and epigenetic regulation of KIF14. Through promoter deletion analyses, we identified one cis-regulatory region containing binding sites for Sp1, HSF1 and YY1. siRNA-mediated knockdown of these transcription factors demonstrated endogenous regulation of KIF14 overexpression by Sp1 and YY1, but not HSF1. ChIP experiments confirmed an enrichment of both Sp1 and YY1 binding to the endogenous KIF14 promoter in OvCa cell lines with high KIF14 expression. A strong correlation was seen in primary serous OvCa tumors between Sp1, YY1 and KIF14 expression, further evidence that these transcription factors are important players in KIF14 overexpression. Hypomethylation patterns were observed in primary serous OvCa tumors, suggesting a minor role for promoter methylation in the control of KIF14 gene expression. miRNA expression analysis determined that miR-93, miR-144 and miR-382 had significantly lower levels of expression in primary serous OvCa tumors than normal tissues; treatment of an OvCa cell line with miRNA mimics and inhibitors specifically modulated KIF14 mRNA levels, pointing to potential novel mechanisms of KIF14 overexpression in primary tumors. Our findings reveal multiple mechanisms of KIF14 upregulation in cancer cells, offering new targets for therapeutic interventions to reduce KIF14 in tumors, aiming at improved prognosis.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Kinesins/metabolism , Oncogene Proteins/metabolism , Ovarian Neoplasms/metabolism , Transcription, Genetic , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation , Female , Gene Deletion , HeLa Cells , Humans , Kinesins/genetics , Mitosis , Neoplasm Metastasis , Oncogene Proteins/genetics , Ovarian Neoplasms/genetics , Prognosis , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Sp1 Transcription Factor/metabolism , Up-Regulation , YY1 Transcription Factor/metabolism
8.
Clin Exp Ophthalmol ; 42(1): 33-52, 2014.
Article in English | MEDLINE | ID: mdl-24433356

ABSTRACT

Retinoblastoma is a paediatric ocular tumour that continues to reveal much about the genetic basis of cancer development. Study of genomic aberrations in retinoblastoma tumours has exposed important mechanisms of cancer development and identified oncogenes and tumour suppressors that offer potential points of therapeutic intervention. The recent development of next-generation genomic technologies has allowed further refinement of the genomic landscape of retinoblastoma at high resolution. In a relatively short period of time, a wealth of genetic and epigenetic data has emerged on a small number of tumour samples. These data highlight the inherent molecular complexity of this cancer despite the fact that most retinoblastomas are initiated by the inactivation of a single tumour suppressor gene. This review outlines the current understanding of the genomic, genetic and epigenetic changes in retinoblastoma, highlighting recent genome-wide analyses that have identified exciting candidate genes worthy of further validation as potential prognostic and therapeutic targets.


Subject(s)
Genomics , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Genes, Tumor Suppressor , Genome, Human/genetics , Humans , MicroRNAs/genetics , Polymorphism, Single Nucleotide
9.
Methods Mol Biol ; 1049: 323-36, 2013.
Article in English | MEDLINE | ID: mdl-23913227

ABSTRACT

We have refined the technique for isolating and propagating cultures of primary epithelial ovarian cancer (EOC) cells derived from solid tumors and ascites. Both protocols involve a simple yet rapid method for the growth and propagation of EOC tumor and ascites cells in a basal culture medium without the addition of growth factors. Isolation of tumor EOC cells involves the mechanical disruption of the tumor tissue with the help of a cell scraper, while ascites-derived EOC cells are mixed with growth medium and placed directly into culture with very little manipulation. We further describe a partial trypsinization method to eliminate fibroblast contamination from primary EOC cells derived from solid tumors. These methods allow for the direct application of many molecular, cellular, and functional analyses within a few weeks of initial isolation, with the added potential of retrospective analyses of archived cells and tissues. Thus, we have included steps for long-term cryopreservation of early-passage EOC cells. Initial isolation of EOC cells can be completed within 1 h, and primary cells are further expanded in culture for several weeks.


Subject(s)
Ascitic Fluid/pathology , Cell Culture Techniques/methods , Ovarian Neoplasms/pathology , Primary Cell Culture/methods , Cell Line, Tumor , Female , Humans , Ovarian Neoplasms/genetics
10.
Lancet Oncol ; 14(4): 327-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23498719

ABSTRACT

BACKGROUND: Retinoblastoma is the childhood retinal cancer that defined tumour-suppressor genes. Previous work shows that mutation of both alleles of the RB1 retinoblastoma suppressor gene initiates disease. We aimed to characterise non-familial retinoblastoma tumours with no detectable RB1 mutations. METHODS: Of 1068 unilateral non-familial retinoblastoma tumours, we compared those with no evidence of RB1 mutations (RB1(+/+)) with tumours carrying a mutation in both alleles (RB1(-/-)). We analysed genomic copy number, RB1 gene expression and protein function, retinal gene expression, histological features, and clinical data. FINDINGS: No RB1 mutations (RB1(+/+)) were reported in 29 (2·7%) of 1068 unilateral retinoblastoma tumours. 15 of the 29 RB1(+/+) tumours had high-level MYCN oncogene amplification (28-121 copies; RB1(+/+)MYCN(A)), whereas none of 93 RB1(-/-) primary tumours tested showed MYCN amplification (p<0·0001). RB1(+/+)MYCN(A) tumours expressed functional RB1 protein, had fewer overall genomic copy-number changes in genes characteristic of retinoblastoma than did RB1(-/-) tumours, and showed distinct aggressive histological features. MYCN amplification was the sole copy-number change in one RB1(+/+)MYCN(A) retinoblastoma. One additional MYCN(A) tumour was discovered after the initial frequencies were determined, and this is included in further analyses. Median age at diagnosis of the 17 children with RB1(+/+)MYCN(A) tumours was 4·5 months (IQR 3·5-10), compared with 24 months (15-37) for 79 children with non-familial unilateral RB1(-/-) retinoblastoma. INTERPRETATION: Amplification of the MYCN oncogene might initiate retinoblastoma in the presence of non-mutated RB1 genes. These unilateral RB1(+/+)MYCN(A) retinoblastomas are characterised by distinct histological features, only a few of the genomic copy-number changes that are characteristic of retinoblastoma, and very early age of diagnosis. FUNDING: National Cancer Institute-National Institutes of Health, Canadian Institutes of Health Research, German Research Foundation, Canadian Retinoblastoma Society, Hyland Foundation, Toronto Netralaya and Doctors Lions Clubs, Ontario Ministry of Health and Long Term Care, UK-Essen, and Foundations Avanti-STR and KiKa.


Subject(s)
Gene Dosage , Nuclear Proteins , Oncogene Proteins , Retinoblastoma Protein , Retinoblastoma , Alleles , Cell Line, Tumor , Child , Child, Preschool , Female , Gene Amplification , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Infant , Mutation , N-Myc Proto-Oncogene Protein , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Polymorphism, Single Nucleotide , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retinoblastoma/pathology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
11.
J Cell Biol ; 199(6): 951-67, 2012 Dec 10.
Article in English | MEDLINE | ID: mdl-23209302

ABSTRACT

The small GTPase Rap1 regulates inside-out integrin activation and thereby influences cell adhesion, migration, and polarity. Several Rap1 effectors have been described to mediate the cellular effects of Rap1 in a context-dependent manner. Radil is emerging as an important Rap effector implicated in cell spreading and migration, but the molecular mechanisms underlying its functions are unclear. We report here that the kinesin KIF14 associates with the PDZ domain of Radil and negatively regulates Rap1-mediated inside-out integrin activation by tethering Radil on microtubules. The depletion of KIF14 led to increased cell spreading, altered focal adhesion dynamics, and inhibition of cell migration and invasion. We also show that Radil is important for breast cancer cell proliferation and for metastasis in mice. Our findings provide evidence that the concurrent up-regulation of Rap1 activity and increased KIF14 levels in several cancers is needed to reach optimal levels of Rap1-Radil signaling, integrin activation, and cell-matrix adhesiveness required for tumor progression.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Kinesins/metabolism , Oncogene Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction , Transplantation, Heterologous
12.
BMC Cancer ; 12: 69, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22336108

ABSTRACT

BACKGROUND: Most human cancers show inactivation of both pRB- and p53-pathways. While retinoblastomas are initiated by loss of the RB1 tumor suppressor gene, TP53 mutations have not been found. High expression of the p53-antagonist MDM2 in human retinoblastomas may compromise p53 tumor surveillance so that TP53 mutations are not selected for in retinoblastoma tumorigenesis. We previously showed that p14ARF protein, which activates p53 by inhibiting MDM2, is low in retinoblastomas despite high mRNA expression. METHODS: In human fetal retinas, adult retinas, and retinoblastoma cells, we determined endogenous p14ARF mRNA, ARF protein, and miR-24 expression, while integrity of p53 signalling in WERI-Rb1 cells was tested using an adenovirus vector expressing p14ARF. To study p14ARF biogenesis, retinoblastoma cells were treated with the proteasome inhibitor, MG132, and siRNA against miR-24. RESULTS: In human retinoblastoma cell lines, p14ARF mRNA was disproportionally high relative to the level of p14ARF protein expression, suggesting a perturbation of p14ARF regulation. When p14ARF was over-expressed by an adenovirus vector, expression of p53 and downstream targets increased and cell growth was inhibited indicating an intact p14ARF-p53 axis. To investigate the discrepancy between p14ARF mRNA and protein in retinoblastoma, we examined p14ARF biogenesis. The proteasome inhibitor, MG132, did not cause p14ARF accumulation, although p14ARF normally is degraded by proteasomes. miR-24, a microRNA that represses p14ARF expression, is expressed in retinoblastoma cell lines and correlates with lower protein expression when compared to other cell lines with high p14ARF mRNA. Transient over-expression of siRNA against miR-24 led to elevated p14ARF protein in retinoblastoma cells. CONCLUSIONS: In retinoblastoma cells where high levels of p14ARF mRNA are not accompanied by high p14ARF protein, we found a correlation between miR-24 expression and low p14ARF protein. p14ARF protein levels were restored without change in mRNA abundance upon miR-24 inhibition suggesting that miR-24 could functionally repress expression, effectively blocking p53 tumor surveillance. During retinal tumorigenesis, miR-24 may intrinsically compromise the p53 response to RB1 loss.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Retinal Neoplasms/metabolism , Retinoblastoma/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/physiology , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Humans , Leupeptins/pharmacology , RNA/metabolism , RNA, Mitochondrial , RNA, Small Interfering/metabolism , Retina/embryology , Retina/metabolism , Retinal Neoplasms/genetics , Tumor Suppressor Protein p14ARF/drug effects , Tumor Suppressor Protein p14ARF/genetics
14.
Int J Cancer ; 130(8): 1844-54, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-21618518

ABSTRACT

The novel oncogene KIF14 (kinesin family member 14) shows genomic gain and overexpression in many cancers including OvCa (ovarian cancer). We discovered that expression of the mitotic kinesin KIF14 is predictive of poor outcome in breast and lung cancers. We now determine the prognostic significance of KIF14 expression in primary OvCa tumors, and evaluate KIF14 action on OvCa cell tumorigenicity in vitro. Gene-specific multiplex PCR and real-time QPCR were used to measure KIF14 genomic (109 samples) and mRNA levels (122 samples) in OvCa tumors. Association of KIF14 with clinical variables was studied using Kaplan-Meier survival and Cox regression analyses. Cellular effects of KIF14 overexpression (stable transfection) and inhibition (stable shRNA knockdown) were studied by proliferation (cell counts), survival (Annexin V immunocytochemistry) and colony formation (soft-agar growth). KIF14 genomic gain (>2.6 copies) was present in 30% of serous OvCas, and KIF14 mRNA was elevated in 91% of tumors versus normal epithelium. High KIF14 in tumors independently predicted for worse outcome (p = 0.03) with loss of correlation with proliferation marker expression and increased rates of recurrence. Overexpression of KIF14 in OvCa cell lines increased proliferation and colony formation (p < 0.01), whereas KIF14 knockdown induced apoptosis and dramatically reduced colony formation (p < 0.05). Our findings indicate that KIF14 mRNA is an independent prognostic marker in serous OvCa. Dependence of OvCa cells on KIF14 for maintenance of in vitro colony formation suggests a role of KIF14 in promoting a tumorigenic phenotype, beyond its known role in proliferation.


Subject(s)
Biomarkers, Tumor/genetics , Kinesins/genetics , Oncogene Proteins/genetics , Ovarian Neoplasms/genetics , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/therapy , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/therapy , DNA Copy Number Variations , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Kinesins/metabolism , Oncogene Proteins/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Prognosis , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proportional Hazards Models , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction/statistics & numerical data
15.
Cancer Lett ; 313(1): 108-21, 2011 Dec 26.
Article in English | MEDLINE | ID: mdl-21945631

ABSTRACT

TGFß superfamily signalling participates in normal and pathophysiologic cellular processes. Despite several reports demonstrating active TGFß superfamily signalling pathways in OvCa cell lines and primary cultures, few studies examine their functional outcome. Herein we show that primary human ovarian cancer cells possess intact autocrine BMP, TGFß and activin signalling. Blocking autocrine signalling resulted in differential cellular responses affecting cellular morphology, motility and proliferation. Additionally, BMP4-induced alterations in morphology and motility are dependent on Smad signalling. These results suggest that a balance between BMP and TGFß/activin signalling may be altered to favour BMP signalling during ovarian cancer metastatic progression.


Subject(s)
Activins/pharmacology , Bone Morphogenetic Proteins/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Transforming Growth Factor beta/pharmacology , Activins/genetics , Autocrine Communication , Blotting, Western , Bone Morphogenetic Proteins/genetics , Cell Line , Cell Shape/drug effects , Female , Gene Expression/drug effects , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Molecular Sequence Data , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA Interference , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins/genetics , Smad Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
16.
Invest Ophthalmol Vis Sci ; 52(10): 7618-24, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21862643

ABSTRACT

PURPOSE: Human retinoblastoma arises from an undefined developing retinal cell after inactivation of RB1. This is emulated in a murine retinoblastoma model by inactivation of pRB by retinal-specific expression of simian virus 40 large T-antigen (TAg-RB). Some mutational events after RB1 loss in humans are recapitulated at the expression level in TAg-RB, supporting preclinical evidence that this model is useful for comparative studies between mouse and human. Here, the characteristics of the TAg-RB cell of origin are defined. METHODS: TAg-RB mice were killed at ages from embryonic day (E)18 to postnatal day (P)35. Tumors were analyzed by immunostaining, DNA copy number PCR, or real-time quantitative RT-PCR for TAg protein, retinal cell type markers, and retinoblastoma-relevant genes. RESULTS: TAg expression began at P8 in a row of inner nuclear layer cells that increased in number through P21 to P28, when clusters reminiscent of small tumors emerged from cells that escaped a wave of apoptosis. Early TAg-expressing cells coexpressed the developmental marker Chx10 and glial markers CRALBP, clusterin, and carbonic anhydrase II (Car2), but not TuJ1, an early neuronal marker. Emerging tumors retained expression of only Chx10 and carbonic anhydrase II. As with human retinoblastoma, TAg-RB tumors showed decreased Cdh11 DNA copy number and gain of Kif14 and Mycn. It was confirmed that TAg-RB tumors lose expression of tumor suppressor cadherin-11 and overexpress oncogenes Kif14, Dek, and E2f3. CONCLUSIONS: TAg-RB tumors displayed molecular similarity to human retinoblastoma and origin in a cell with features of differentiated Müller glia with progenitor properties.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Gene Expression Regulation/physiology , Neoplastic Stem Cells/pathology , Neuroglia/pathology , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Animals , Apoptosis , Biomarkers, Tumor , Caspase 3/metabolism , Cell Differentiation/genetics , Cell Lineage , Cell Proliferation , DNA, Neoplasm/genetics , Fluorescent Antibody Technique, Indirect , Gene Silencing/physiology , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retinoblastoma Protein/genetics , Tubulin/metabolism
17.
J Ovarian Res ; 4: 10, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21699706

ABSTRACT

Ovarian cancer continues to be the most lethal of the gynaecologic malignancies due to the lack of early detection, screening strategies and ineffective therapeutics for late-stage metastatic disease, particularly in the recurrent setting. The gathering of researchers investigating fundamental pathobiology of ovarian cancer and the clinicians who treat patients with this insidious disease is paramount to meeting the challenges we face. Since 2002, the Canadian Conference on Ovarian Cancer Research, held every two years, has served this essential purpose. The objectives of this conference have been to disseminate new information arising from the most recent ovarian cancer research and identify the most pressing challenges we still face as scientists and clinicians. This is best accomplished through direct encounters and exchanges of innovative ideas among colleagues and trainees from the realms of basic science and clinical disciplines. This meeting has and continues to successfully facilitate rapid networking and establish new collaborations from across Canada. This year, more guest speakers and participants from other countries have extended the breadth of the research on ovarian cancer that was discussed at the meeting. This report summarizes the key findings presented at the fifth biennial Canadian Conference on Ovarian Cancer Research held in Toronto, Ontario, and includes the important issues and challenges we still face in the years ahead to make a significant impact on this devastating disease.

18.
Gene ; 414(1-2): 95-105, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18372118

ABSTRACT

Bone morphogenetic protein (BMP)-4 signalling leads to the direct upregulation of ID3 proto-oncogene expression in human ovarian cancer cells. An upstream BMP4-responsive enhancer element consisting of a palindromic BMP response element (BRE) site and CAGA box was identified ~3.0 kb upstream of the human ID3 gene, and a nearly-identical element exists in the second intron of the ID3 gene. BMP4 stimulation leads to the direct binding of Smads 1/5 and Smad4 to the upstream and intronic enhancers, and together both enhancers cooperate to yield heightened BMP4-mediated ID3 promoter activity. We further demonstrate that ID3 is overexpressed in human ovarian cancer cells when compared to normal ovarian surface epithelial cells, and treatment of ovarian cancer cells with the BMP4 antagonist Noggin abrogates endogenous ID3 gene expression. Our findings define the mechanism of BMP4-mediated ID3 gene expression, and support the notion that ovarian cancer cells possess autocrine BMP4 signalling required to sustain ID3 overexpression which may contribute to human ovarian cancer pathogenesis.


Subject(s)
Autocrine Communication , Bone Morphogenetic Proteins/metabolism , Inhibitor of Differentiation Proteins/genetics , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Signal Transduction , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , COS Cells , Cells, Cultured , Chlorocebus aethiops , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , Female , Humans , Inhibitor of Differentiation Proteins/metabolism , Luciferases , Molecular Sequence Data , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , Promoter Regions, Genetic/genetics , Proto-Oncogene Mas , Response Elements , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection
19.
Carcinogenesis ; 28(6): 1153-62, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17272306

ABSTRACT

We identified previously an autocrine bone morphogenetic protein-4 (BMP4) signalling pathway in primary human normal ovarian surface epithelial (OSE) and epithelial ovarian cancer (OvCa) cells. Herein we show that treatment of OvCa cells with BMP4 produced morphological alterations and increased cellular adhesion, motility and invasion. The BMP4 inhibitor noggin blocked the BMP4-induced phenotype, and decreased autocrine BMP4-mediated OvCa cell motility and adherence. In response to exogenous BMP4, the epithelial-mesenchymal transition (EMT) markers Snail and Slug mRNA and protein were up-regulated, E-cadherin mRNA and protein were down-regulated and the network of alpha smooth muscle actin changed to resemble a mesenchymal cell. We also observed changes in the level of activated Rho GTPases in OvCa cells treated with BMP4, strongly suggesting that the changes in morphology, adhesion, motility and invasion are probably mediated through the activation of these molecules. Strikingly, treatment of normal OSE cells with BMP4 or noggin failed to alter cell motility, providing evidence that OSE and OvCa cells possess a distinct capability to respond to BMP4. Overall, our studies suggest a link between autocrine BMP signalling mediated through the Rho GTPase family and Snail- and Slug-induced EMT that may collectively contribute to aggressive OvCa behaviour.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Morphogenetic Proteins/physiology , Mesoderm/enzymology , Ovarian Neoplasms/enzymology , rho GTP-Binding Proteins/metabolism , Bone Morphogenetic Protein 4 , Disease Progression , Enzyme Activation/physiology , Epithelium/enzymology , Epithelium/pathology , Female , Humans , Mesoderm/pathology , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
20.
Nat Protoc ; 1(6): 2643-9, 2006.
Article in English | MEDLINE | ID: mdl-17406520

ABSTRACT

Our laboratory has refined the technique for isolating primary cultures of normal human ovarian surface epithelial (OSE) cells by combining two different protocols involving the enzymatic and mechanical removal of OSE cells from ovarian biopsies. A simple protocol of obtaining primary epithelial ovarian cancer (EOC) cells from the ascites fluid removed from patients with high-grade ovarian cancer is also described. These methods allow for the direct application of many molecular and cellular analyses of normal versus cancer cells isolated freshly from patients, with the added potential for retrospective analyses of archived cells and tissues. Thus, we have included optional steps for the immediate preparation of ascites-derived EOC cells to be used for subsequent cytological analyses. Initial isolation of OSE or EOC cells can be completed in 1 h, and primary cells are further expanded in culture for several weeks.


Subject(s)
Cell Culture Techniques , Epithelial Cells , Ovarian Neoplasms , Tumor Cells, Cultured , Ascites , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...