1
|
LaFargue CJ, Amero P, Noh K, Mangala LS, Wen Y, Bayraktar E, Umamaheswaran S, Stur E, Dasari SK, Ivan C, Pradeep S, Yoo W, Lu C, Jennings NB, Vathipadiekal V, Hu W, Chelariu-Raicu A, Ku Z, Deng H, Xiong W, Choi HJ, Hu M, Kiyama T, Mao CA, Ali-Fehmi R, Birrer MJ, Liu J, Zhang N, Lopez-Berestein G, de Franciscis V, An Z, Sood AK. Overcoming adaptive resistance to anti-VEGF therapy by targeting CD5L. Nat Commun 2023; 14:2407. [PMID: 37100807 PMCID: PMC10133315 DOI: 10.1038/s41467-023-36910-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 02/22/2023] [Indexed: 04/28/2023] Open
Abstract
Antiangiogenic treatment targeting the vascular endothelial growth factor (VEGF) pathway is a powerful tool to combat tumor growth and progression; however, drug resistance frequently emerges. We identify CD5L (CD5 antigen-like precursor) as an important gene upregulated in response to antiangiogenic therapy leading to the emergence of adaptive resistance. By using both an RNA-aptamer and a monoclonal antibody targeting CD5L, we are able to abate the pro-angiogenic effects of CD5L overexpression in both in vitro and in vivo settings. In addition, we find that increased expression of vascular CD5L in cancer patients is associated with bevacizumab resistance and worse overall survival. These findings implicate CD5L as an important factor in adaptive resistance to antiangiogenic therapy and suggest that modalities to target CD5L have potentially important clinical utility.
Collapse
Affiliation(s)
- Christopher J LaFargue
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Istituto di Endocrinologia ed Oncologia Sperimentale, CNR, Naples, Italy
| | - Kyunghee Noh
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Laboratory of Disease Modeling and Therapeutics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yunfei Wen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA.
| | - Emine Bayraktar
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Sujanitha Umamaheswaran
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Elaine Stur
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Santosh K Dasari
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sunila Pradeep
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Wonbeak Yoo
- Department of Molecular & Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chunhua Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Nicholas B Jennings
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Vinod Vathipadiekal
- Wave Life Sciences, 733 Concord Avenue, Cambridge, MA, 02138, USA
- Department of Genetic Medicines, Alloy Therapeutics, Waltham, USA
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Anca Chelariu-Raicu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Obstetrics and Gynecology, Ludwig Maximilians University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Munich, Germany
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hyun-Jin Choi
- Department of Obstetrics and Gynecology, Chung-Ang University, College of Medicine, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Chung-Ang University Gwangmyeong Hospital, College of Medicine Chung-Ang University, Seoul, South Korea
| | - Min Hu
- CPRIT Single Core, Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Takae Kiyama
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, 77030, USA
| | - Chai-An Mao
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, 77030, USA
- The MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Rouba Ali-Fehmi
- Department of Pathology, Wayne State University, Detroit, MI, 48201, USA
| | - Michael J Birrer
- Winthrop P. Rockefeller Cancer Institute at the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vittorio de Franciscis
- National Research Council (CNR), Institute of Genetic and Biomedical Research (IRGB)-UOS Milan via Rita Levi Montalcini, 20090, Pieve Emanuele, MI, Italy
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA.
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Mitok KA, Keller MP, Attie AD. Sorting through the extensive and confusing roles of sortilin in metabolic disease. J Lipid Res 2022; 63:100243. [PMID: 35724703 PMCID: PMC9356209 DOI: 10.1016/j.jlr.2022.100243] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 01/06/2023] Open
Abstract
Sortilin is a post-Golgi trafficking receptor homologous to the yeast vacuolar protein sorting receptor 10 (VPS10). The VPS10 motif on sortilin is a 10-bladed β-propeller structure capable of binding more than 50 proteins, covering a wide range of biological functions including lipid and lipoprotein metabolism, neuronal growth and death, inflammation, and lysosomal degradation. Sortilin has a complex cellular trafficking itinerary, where it functions as a receptor in the trans-Golgi network, endosomes, secretory vesicles, multivesicular bodies, and at the cell surface. In addition, sortilin is associated with hypercholesterolemia, Alzheimer's disease, prion diseases, Parkinson's disease, and inflammation syndromes. The 1p13.3 locus containing SORT1, the gene encoding sortilin, carries the strongest association with LDL-C of all loci in human genome-wide association studies. However, the mechanism by which sortilin influences LDL-C is unclear. Here, we review the role sortilin plays in cardiovascular and metabolic diseases and describe in detail the large and often contradictory literature on the role of sortilin in the regulation of LDL-C levels.
Collapse
Affiliation(s)
- Kelly A Mitok
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
3
|
Sortilin 1 regulates hepatocellular carcinoma progression by activating the PI3K/AKT signaling. Hum Exp Toxicol 2022; 41:9603271221140111. [DOI: 10.1177/09603271221140111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background Sortilin 1 (SORT1) has been reported as an oncogene in several human tumors. Nonetheless, the biological functions of SORT1 in hepatocellular carcinoma (HCC) remain poorly understood. Methods Western blotting was employed for the determination of protein expression. Hepatocellular carcinoma cell viability, apoptosis, migration, and invasion were measured via CCK-8, flow cytometry, wound healing, and Transwell assays. Results Sortilin 1 was upregulated in HCC and closely associated with unsatisfactory outcomes of HCC patients. Furthermore, in vitro and in vivo assays revealed that SORT1 knockdown significantly diminished HCC cell proliferation and metastasis but accelerated HCC cell apoptosis; moreover, SORT1 depletion also restrained the growth of xenografted HCC tumors. Mechanistically, SORT1 activated PI3K/AKT signaling in HCC cells, thereby promoting the malignant behaviors of HCC cells. Conclusion This study demonstrated that SORT1 might promote HCC progression by activating PI3K/AKT signaling, indicating that SORT1 might be a promising target and biomarker for HCC treatment and prognosis.
Collapse
|
4
|
Facchinello N, Astone M, Audano M, Oberkersch RE, Spizzotin M, Calura E, Marques M, Crisan M, Mitro N, Santoro MM. Oxidative pentose phosphate pathway controls vascular mural cell coverage by regulating extracellular matrix composition. Nat Metab 2022; 4:123-140. [PMID: 35102339 PMCID: PMC7612297 DOI: 10.1038/s42255-021-00514-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/06/2021] [Indexed: 12/11/2022]
Abstract
Vascular mural cells (vMCs) play an essential role in the development and maturation of the vasculature by promoting vessel stabilization through their interactions with endothelial cells. Whether endothelial metabolism influences mural cell recruitment and differentiation is unknown. Here, we show that the oxidative pentose phosphate pathway (oxPPP) in endothelial cells is required for establishing vMC coverage of the dorsal aorta during early vertebrate development in zebrafish and mice. We demonstrate that laminar shear stress and blood flow maintain oxPPP activity, which in turn, promotes elastin expression in blood vessels through production of ribose-5-phosphate. Elastin is both necessary and sufficient to drive vMC recruitment and maintenance when the oxPPP is active. In summary, our work demonstrates that endothelial cell metabolism regulates blood vessel maturation by controlling vascular matrix composition and vMC recruitment.
Collapse
Affiliation(s)
- Nicola Facchinello
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Matteo Astone
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | - Roxana E Oberkersch
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Marianna Spizzotin
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy
| | - Enrica Calura
- Department of Biology, University of Padua, Padua, Italy
| | - Madalena Marques
- Centre for Cardiovascular Science and Centre for Regenerative Medicine/Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Mihaela Crisan
- Centre for Cardiovascular Science and Centre for Regenerative Medicine/Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | - Massimo M Santoro
- Laboratory of Angiogenesis and Cancer Metabolism, DiBio, University of Padua, Padua, Italy.
| |
Collapse
|
5
|
Zhang H, Wu Y, Li H, Sun L, Meng X. Model constructions of chemosensitivity and prognosis of high grade serous ovarian cancer based on evaluation of immune microenvironment and immune response. Cancer Cell Int 2021; 21:593. [PMID: 34736480 PMCID: PMC8567582 DOI: 10.1186/s12935-021-02295-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/21/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The prognosis of high grade serous ovarian cancer (HGSOC) patients is closely related to the immune microenvironment and immune response. Based on this, the purpose of this study was to construct a model to predict chemosensitivity and prognosis, and provide novel biomarkers for immunotherapy and prognosis evaluation of HGSOC. METHODS GSE40595 (38 samples), GSE18520 (63 samples), GSE26712 (195 samples), TCGA (321 samples) and GTEx (88 samples) were integrated to screen differential expressed genes (DEGs) of HGSOC. The prognosis related DEGs (DEPGs) were screened through overall survival analysis. The DEGs-encoded protein-protein interaction network was constructed and hub genes of DEPGs (DEPHGs) were generated by STRING. Immune characteristics of the samples were judged by ssGSEA, ESTIMATE and CYBERSORT. TIMER was used to analyze the relationship between DEPHGs and tumor-infiltrating immunocytes, as well as the immune checkpoint genes, finally immune-related DEPHGs (IDEPHGs) were determined, and whose expression in 12 pairs of HGSOC tissues and tumor-adjacent tissues were analyzed by histological verification. Furthermore, the chemosensitivity genes in IDEPHGs were screened according to GSE15622 (n = 65). Finally, two prediction models of paclitaxel sensitivity score (PTX score) and carboplatin sensitivity score (CBP score) were constructed by lasso algorithm. The area under curve was calculated to estimate the accuracy of candidate gene models in evaluating chemotherapy sensitivity. RESULTS 491 DEGs were screened and 37 DEGs were identified as DEPGs, and 11 DEPHGs were further identified. Among them, CXCL13, IDO1, PI3, SPP1 and TRIM22 were screened as IDEPHGs and verified in the human tissues. Further analysis showed that IDO1, PI3 and TRIM22 could independently affect the chemotherapy sensitivity of HGSOC patients. The PTX score was significantly better than TRIM22, PI3, SPP1, IDO1 and CXCL13 in predicting paclitaxel sensitivity, so was CBP score in predicting carboplatin sensitivity. What's more, both of the HGSOC patients with high PTX score or high CBP score had longer survival time. CONCLUSIONS Five IDEPHGs identified through comprehensive bioinformatics analysis were closely related with the prognosis, immune microenvironment and chemotherapy sensitivity of HGSOC. Two prediction models based on IDEPHGs might have potential application of chemotherapy sensitivity and prognosis for patients with HGSOC.
Collapse
Affiliation(s)
- Han Zhang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yijun Wu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Hao Li
- Department of Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiangkai Meng
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
6
|
Dong B, Liang J, Li D, Song W, Zhao S, Ma Y, Song J, Zhu M, Yang T. Tumor Expression Profile Analysis Developed and Validated a Prognostic Model Based on Immune-Related Genes in Bladder Cancer. Front Genet 2021; 12:696912. [PMID: 34512722 PMCID: PMC8429908 DOI: 10.3389/fgene.2021.696912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Bladder cancer (BLCA) ranks 10th in incidence among malignant tumors and 6th in incidence among malignant tumors in males. With the application of immune therapy, the overall survival (OS) rate of BLCA patients has greatly improved, but the 5-year survival rate of BLCA patients is still low. Furthermore, not every BLCA patient benefits from immunotherapy, and there are a limited number of biomarkers for predicting the immunotherapy response. Therefore, novel biomarkers for predicting the immunotherapy response and prognosis of BLCA are urgently needed. Methods: The RNA sequencing (RNA-seq) data, clinical data and gene annotation files for The Cancer Genome Atlas (TCGA) BLCA cohort were extracted from the University of California, Santa Cruz (UCSC) Xena Browser. The BLCA datasets GSE31684 and GSE32894 from the Gene Expression Omnibus (GEO) database were extracted for external validation. Immune-related genes were extracted from InnateDB. Significant differentially expressed genes (DEGs) were identified using the R package “limma,” and Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis for the DEGs were performed using R package “clusterProfiler.” Least absolute shrinkage and selection operator (LASSO) regression analysis were used to construct the signature model. The infiltration level of each immune cell type was estimated using the single-sample gene set enrichment analysis (ssGSEA) algorithm. The performance of the model was evaluated with receiver operating characteristic (ROC) curves and calibration curves. Results: In total, 1,040 immune-related DEGs were identified, and eight signature genes were selected to construct a model using LASSO regression analysis. The risk score of BLCA patients based on the signature model was negatively correlated with OS and the immunotherapy response. The ROC curve for OS revealed that the model had good accuracy. The calibration curve showed good agreement between the predictions and actual observations. Conclusions: Herein, we constructed an immune-related eight-gene signature that could be a potential biomarker to predict the immunotherapy response and prognosis of BLCA patients.
Collapse
Affiliation(s)
- Bingqi Dong
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jiaming Liang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Ding Li
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenping Song
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Shiming Zhao
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yongkang Ma
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jinbo Song
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Mingkai Zhu
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Tiejun Yang
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
7
|
Torres Crigna A, Link B, Samec M, Giordano FA, Kubatka P, Golubnitschaja O. Endothelin-1 axes in the framework of predictive, preventive and personalised (3P) medicine. EPMA J 2021; 12:265-305. [PMID: 34367381 PMCID: PMC8334338 DOI: 10.1007/s13167-021-00248-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
Endothelin-1 (ET-1) is involved in the regulation of a myriad of processes highly relevant for physical and mental well-being; female and male health; in the modulation of senses, pain, stress reactions and drug sensitivity as well as healing processes, amongst others. Shifted ET-1 homeostasis may influence and predict the development and progression of suboptimal health conditions, metabolic impairments with cascading complications, ageing and related pathologies, cardiovascular diseases, neurodegenerative pathologies, aggressive malignancies, modulating, therefore, individual outcomes of both non-communicable and infectious diseases such as COVID-19. This article provides an in-depth analysis of the involvement of ET-1 and related regulatory pathways in physiological and pathophysiological processes and estimates its capacity as a predictor of ageing and related pathologies,a sensor of lifestyle quality and progression of suboptimal health conditions to diseases for their targeted preventionand as a potent target for cost-effective treatments tailored to the person.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Barbara Link
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
8
|
Yan M, Zhu X, Qiao H, Zhang H, Xie W, Cai J. Downregulated CMTM8 Correlates with Poor Prognosis in Gastric Cancer Patients. DNA Cell Biol 2021; 40:791-797. [PMID: 33978454 DOI: 10.1089/dna.2021.0110] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
This study aimed to examine the expression and clinical significance of chemokine-like factor-like MARVEL transmembrane domain-containing family member 8 (CMTM8) in gastric cancer (GC). The mRNA and protein expression of CMTM8 were detected by bioinformatics analysis and immunohistochemistry (IHC), respectively. Bioinformatics analysis found that there was a high mRNA expression of CMTM8 in GC tissues, but failed to identify a significant relationship with the clinicopathological features or prognosis of GC patients. However, IHC results showed that the positive expression of CMTM8 protein in GC tissues was significantly lower than that of adjacent nontumor tissues and correlated with differentiation, tumor node metastasis stage, and distal metastasis of GC patients (p < 0.05). Moreover, the survival time of GC patients with negative CMTM8 protein expression group was shorter than that of positive CMTM8 protein expression group by Kaplan-Meier survival analysis (p < 0.05). Cox proportional hazards model (COX) regression analysis indicated that CMTM8 protein was an independent protective factor for the overall survival of GC patients. Further Gene Set Enrichment Analysis suggested that CMTM8 may be involved in regulating the calcium signaling pathway, cell adhesion molecules, and cytokine-cytokine receptor interaction in GC. Our study shows that CMTM8 protein is downregulated in GC tissues, and CMTM8 protein expression is related to GC metastasis and the prognosis of GC patients.
Collapse
Affiliation(s)
- Meng Yan
- Department of Surgery, Hebei Medical University, Shijiazhuang, China.,Department of Surgery and Oncology, Hebei General Hospital, Shijiazhuang, China.,Department of General Surgery III, Baoding First Central Hospital, Baoding, China
| | - Xiaonian Zhu
- Department of Epidemiology and Statistics, School of Public Health, Guilin Medical University, Guilin, China
| | - Haizhi Qiao
- Department of Pathology, Baoding First Central Hospital, Baoding, China
| | - Huiqing Zhang
- Department of General Surgery III, Baoding First Central Hospital, Baoding, China
| | - Wenjie Xie
- Department of General Surgery III, Baoding First Central Hospital, Baoding, China
| | - Jianhui Cai
- Department of Surgery, Hebei Medical University, Shijiazhuang, China.,Department of Surgery and Oncology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
9
|
Guo X, Song C, Fang L, Li M, Yue L, Sun Q. FLRT2 functions as Tumor Suppressor gene inactivated by promoter methylation in Colorectal Cancer. J Cancer 2020; 11:7329-7338. [PMID: 33193897 PMCID: PMC7646184 DOI: 10.7150/jca.47558] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/11/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death worldwide. Epigenetic alterations, especially DNA methylation, contribute to the initiation and progression of CRC. To identify novel methylated tumor suppressors in CRC, MethylRAD-Seq screening was performed. As the result, FLRT2 was found to be preferentially methylated. In the present study, we aimed to elucidate the epigenetic regulations and biological functions of FLRT2 in CRC. Significant FLRT2 hypermethylation was initially confirmed in CRC samples and cell lines. Meanwhile, downregulated expression of FLRT2 was observed in CRC, which is probably attributed to promoter methylation of FLRT2. Consistently, the expression of FLRT2 was restored after treatment with DNA demethylating agent 5-AZA. FLRT2 overexpression resulted in impaired cell viability and colony formation. Additionally, FLRT2 overexpression led to a reduction in cell migration and cell invasion. Furthermore, we also observed that FLRT2 induced cell cycle arrest. Mechanistically, these effects were associated with the downregulation of phosphor-AKT, phosphor-ERK, CDK2, Cyclin A, and MMP2, and upregulation of P21. Taken together, these results define a tumor-suppressor role of FLRT2 with epigenetic silencing in the pathogenesis of CRC. Moreover, FLRT2 promoter methylation may be a useful epigenetic biomarker for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Xiaohong Guo
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Chao Song
- Department of Pathology, Zibo Central Hospital, Zibo, Shandong, China
| | - Lei Fang
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Min Li
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Longtao Yue
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Qing Sun
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
10
|
Brocard M, Khasnis S, Wood CD, Shannon-Lowe C, West MJ. Pumilio directs deadenylation-associated translational repression of the cyclin-dependent kinase 1 activator RGC-32. Nucleic Acids Res 2019; 46:3707-3725. [PMID: 29385536 PMCID: PMC5909466 DOI: 10.1093/nar/gky038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 01/22/2018] [Indexed: 12/11/2022] Open
Abstract
Response gene to complement-32 (RGC-32) activates cyclin-dependent kinase 1, regulates the cell cycle and is deregulated in many human tumours. We previously showed that RGC-32 expression is upregulated by the cancer-associated Epstein-Barr virus (EBV) in latently infected B cells through the relief of translational repression. We now show that EBV infection of naïve primary B cells also induces RGC-32 protein translation. In EBV-immortalised cell lines, we found that RGC-32 depletion resulted in cell death, indicating a key role in B cell survival. Studying RGC-32 translational control in EBV-infected cells, we found that the RGC-32 3′untranslated region (3′UTR) mediates translational repression. Repression was dependent on a single Pumilio binding element (PBE) adjacent to the polyadenylation signal. Mutation of this PBE did not affect mRNA cleavage, but resulted in increased polyA tail length. Consistent with Pumilio-dependent recruitment of deadenylases, we found that depletion of Pumilio in EBV-infected cells increased RGC-32 protein expression and polyA tail length. The extent of Pumilio binding to the endogenous RGC-32 mRNA in EBV-infected cell lines also correlated with RGC-32 protein expression. Our data demonstrate the importance of RGC-32 for the survival of EBV-immortalised B cells and identify Pumilio as a key regulator of RGC-32 translation.
Collapse
Affiliation(s)
- Michèle Brocard
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK
| | - Sarika Khasnis
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK
| | - C David Wood
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK
| | - Claire Shannon-Lowe
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Michelle J West
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK
| |
Collapse
|
11
|
Vlaicu SI, Tatomir A, Anselmo F, Boodhoo D, Chira R, Rus V, Rus H. RGC-32 and diseases: the first 20 years. Immunol Res 2019; 67:267-279. [DOI: 10.1007/s12026-019-09080-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
12
|
Vlaicu SI, Tatomir A, Rus V, Rus H. Role of C5b-9 and RGC-32 in Cancer. Front Immunol 2019; 10:1054. [PMID: 31156630 PMCID: PMC6530392 DOI: 10.3389/fimmu.2019.01054] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/24/2019] [Indexed: 01/13/2023] Open
Abstract
The complement system represents an effective arsenal of innate immunity as well as an interface between innate and adaptive immunity. Activation of the complement system culminates with the assembly of the C5b-9 terminal complement complex on cell membranes, inducing target cell lysis. Translation of this sequence of events into a malignant setting has traditionally afforded C5b-9 a strict antitumoral role, in synergy with antibody-dependent tumor cytolysis. However, in recent decades, a plethora of evidence has revised this view, highlighting the tumor-promoting properties of C5b-9. Sublytic C5b-9 induces cell cycle progression by activating signal transduction pathways (e.g., Gi protein/ phosphatidylinositol 3-kinase (PI3K)/Akt kinase and Ras/Raf1/ERK1) and modulating the activation of cancer-related transcription factors, while shielding malignant cells from apoptosis. C5b-9 also induces Response Gene to Complement (RGC)-32, a gene that contributes to cell cycle regulation by activating the Akt and CDC2 kinases. RGC-32 is expressed by tumor cells and plays a dual role in cancer, functioning as either a tumor promoter by endorsing malignancy initiation, progression, invasion, metastasis, and angiogenesis, or as a tumor suppressor. In this review, we present recent data describing the versatile, multifaceted roles of C5b-9 and its effector, RGC-32, in cancer.
Collapse
Affiliation(s)
- Sonia I Vlaicu
- Department of Internal Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Alexandru Tatomir
- Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Violeta Rus
- Division of Rheumatology and Immunology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
13
|
Chen LY, Huang RL, Chan MW, Yan PS, Huang TS, Wu RC, Suryo Rahmanto Y, Su PH, Weng YC, Chou JL, Chao TK, Wang YC, Shih IM, Lai HC. TET1 reprograms the epithelial ovarian cancer epigenome and reveals casein kinase 2α as a therapeutic target. J Pathol 2019; 248:363-376. [PMID: 30883733 DOI: 10.1002/path.5266] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/09/2019] [Accepted: 03/11/2019] [Indexed: 12/22/2022]
Abstract
Ten-eleven translocation methylcytosine dioxygenase-1, TET1, takes part in active DNA demethylation. However, our understanding of DNA demethylation in cancer biology and its clinical significance remain limited. This study showed that TET1 expression correlated with poor survival in advanced-stage epithelial ovarian carcinoma (EOC), and with cell migration, anchorage-independent growth, cancer stemness, and tumorigenicity. In particular, TET1 was highly expressed in serous tubal intraepithelial carcinoma (STIC), a currently accepted type II EOC precursor, and inversely correlated with TP53 mutations. Moreover, TET1 could demethylate the epigenome and activate multiple oncogenic pathways, including an immunomodulation network having casein kinase II subunit alpha (CK2α) as a hub. Patients with TET1high CK2αhigh EOCs had the worst outcomes, and TET1-expressing EOCs were more sensitive to a CK2 inhibitor, both in vitro and in vivo. Our findings uncover the oncogenic and poor prognostic roles of TET1 in EOC and suggest an unexplored role of epigenetic reprogramming in early ovarian carcinogenesis. Moreover, the immunomodulator CK2α represents a promising new therapeutic target, warranting clinical trials of the tolerable CK2 inhibitor, CX4945, for precision medicine against EOC. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lin-Yu Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Rui-Lan Huang
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Michael Wy Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan.,Institute of Molecular Biology, National Chung Cheng University, Chiayi, Taiwan
| | - Pearlly S Yan
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Tien-Shuo Huang
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Ren-Chin Wu
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yohan Suryo Rahmanto
- Department of Pathology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Po-Hsuan Su
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yu-Chun Weng
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Jian-Liang Chou
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Tai-Kuang Chao
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Chi Wang
- Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ie-Ming Shih
- Department of Pathology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA.,Department of Gynecology and Obstetrics, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Hung-Cheng Lai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
14
|
Gabbasov R, Xiao F, Howe CG, Bickel LE, O'Brien SW, Benrubi D, Do TV, Zhou Y, Nicolas E, Cai KQ, Litwin S, Seo S, Golemis EA, Connolly DC. NEDD9 promotes oncogenic signaling, a stem/mesenchymal gene signature, and aggressive ovarian cancer growth in mice. Oncogene 2018; 37:4854-4870. [PMID: 29773902 PMCID: PMC6119087 DOI: 10.1038/s41388-018-0296-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 03/23/2018] [Accepted: 04/13/2018] [Indexed: 12/20/2022]
Abstract
Neural precursor cell expressed, developmentally downregulated 9 (NEDD9) supports oncogenic signaling in a number of solid and hematologic tumors. Little is known about the role of NEDD9 in ovarian carcinoma (OC), but available data suggest elevated mRNA and protein expression in advanced stage high-grade cancers. We used a transgenic MISIIR-TAg mouse OC model combined with genetic ablation of Nedd9 to investigate its action in the development and progression of OC. A Nedd9-/- genotype delayed tumor growth rate, reduced incidence of ascites, and reduced expression and activation of signaling proteins including SRC, STAT3, E-cadherin, and AURKA. Cell lines established from MISIIR-TAg;Nedd9-/- and MISIIR-TAg;Nedd9+/+ mice exhibited altered migration and invasion. Growth of these cells in a syngeneic allograft model indicated that systemic Nedd9 loss in the microenvironment had little impact on tumor allograft growth, but in a Nedd9 wild-type background Nedd9-/- allografts exhibited significantly reduced growth, dissemination, and oncogenic signaling compared to Nedd9+/+ allografts. Gene expression analysis revealed that Nedd9+/+ tumors exhibited more mesenchymal "stem-like" transcriptional program, including increased expression of Aldh1a1 and Aldh1a2. Conversely, loss of Nedd9 resulted in increased expression of differentiation genes, including fallopian tube markers Foxj1, Ovgp1, and Pax8. Collectively, these data suggest that tumor cell-intrinsic Nedd9 expression promotes OC development and progression by broad induction of oncogenic protein signaling and stem/mesenchymal gene expression.
Collapse
Affiliation(s)
- Rashid Gabbasov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan, Russia
| | - Fang Xiao
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Caitlin G Howe
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Laura E Bickel
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Shane W O'Brien
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniel Benrubi
- Division of Gynecologic Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Thuy-Vy Do
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yan Zhou
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Samuel Litwin
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Sachiko Seo
- Department of Hematology & Oncology, National Cancer Research Center East, Kashiwa, Japan
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Denise C Connolly
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
15
|
MiR-514 attenuates proliferation and increases chemoresistance by targeting ATP binding cassette subfamily in ovarian cancer. Mol Genet Genomics 2018; 293:1159-1167. [PMID: 29752546 DOI: 10.1007/s00438-018-1447-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Cisplatin is one of the most popular chemotherapeutic drugs in treating ovarian cancer. Resistance to cisplatin is a common clinical challenge that needs to be solved to increase its anti-tumor effects. The relation of miR-514 expression with prognosis in ovarian cancer patients was analyzed based on GSE73584 datasets. The regulation of miR-514 on proliferation and cisplatin chemosensitivity of ovarian cells was examined by MTT assay, colony-formation assay and soft-agar colony-formation assay. Dual luciferase assay was performed to detect the direct interaction of miR-514 with its downstream targets. Immunobloting and qRT-PCR were performed for target gene expression analysis. Low expression of miR-514 was related to poor prognosis in ovarian cancer patients. MiR-514 repressed proliferation and decreased cisplatin chemosensitivity in ovarian cancer cells by targeting ATP binding cassette subfamily. MiR-514 is of clinically significance in ovarian cancer by attenuating proliferation of ovarian cancer cells and decreasing chemoresistance of cisplatin by targeting ATP binding cassette subfamily.
Collapse
|
16
|
Noh K, Mangala LS, Han HD, Zhang N, Pradeep S, Wu SY, Ma S, Mora E, Rupaimoole R, Jiang D, Wen Y, Shahzad MMK, Lyons Y, Cho M, Hu W, Nagaraja AS, Haemmerle M, Mak CSL, Chen X, Gharpure KM, Deng H, Xiong W, Kingsley CV, Liu J, Jennings N, Birrer MJ, Bouchard RR, Lopez-Berestein G, Coleman RL, An Z, Sood AK. Differential Effects of EGFL6 on Tumor versus Wound Angiogenesis. Cell Rep 2017; 21:2785-2795. [PMID: 29212026 PMCID: PMC5749980 DOI: 10.1016/j.celrep.2017.11.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 09/18/2017] [Accepted: 11/02/2017] [Indexed: 11/25/2022] Open
Abstract
Angiogenesis inhibitors are important for cancer therapy, but clinically approved anti-angiogenic agents have shown only modest efficacy and can compromise wound healing. This necessitates the development of novel anti-angiogenesis therapies. Here, we show significantly increased EGFL6 expression in tumor versus wound or normal endothelial cells. Using a series of in vitro and in vivo studies with orthotopic and genetically engineered mouse models, we demonstrate the mechanisms by which EGFL6 stimulates tumor angiogenesis. In contrast to its antagonistic effects on tumor angiogenesis, EGFL6 blockage did not affect normal wound healing. These findings have significant implications for development of anti-angiogenesis therapies.
Collapse
Affiliation(s)
- Kyunghee Noh
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Gene Therapy Research Unit, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hee-Dong Han
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sherry Y Wu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaolin Ma
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Edna Mora
- Department of Surgery, University of Puerto Rico, San Juan 00936, Puerto Rico; University of Puerto Rico Comprehensive Cancer Center, San Juan 00936, Puerto Rico; Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77584, USA
| | - Rajesha Rupaimoole
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dahai Jiang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yunfei Wen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mian M K Shahzad
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasmin Lyons
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - MinSoon Cho
- Department of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Archana S Nagaraja
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Monika Haemmerle
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Celia S L Mak
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiuhui Chen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kshipra M Gharpure
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Charles V Kingsley
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas Jennings
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael J Birrer
- University of Alabama Comprehensive Cancer Center, Birmingham, AL 35294, USA
| | - Richard R Bouchard
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
17
|
Ou-Yang L, Zhang XF, Wu M, Li XL. Node-based learning of differential networks from multi-platform gene expression data. Methods 2017; 129:41-49. [DOI: 10.1016/j.ymeth.2017.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 04/11/2017] [Accepted: 05/18/2017] [Indexed: 01/07/2023] Open
|
18
|
Wang XY, Li SN, Zhu HF, Hu ZY, Zhong Y, Gu CS, Chen SY, Liu TF, Li ZG. RGC32 induces epithelial-mesenchymal transition by activating the Smad/Sip1 signaling pathway in CRC. Sci Rep 2017; 7:46078. [PMID: 28470188 PMCID: PMC5415763 DOI: 10.1038/srep46078] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/10/2017] [Indexed: 01/01/2023] Open
Abstract
Response gene to complement 32 (RGC32) is a transcription factor that regulates the expression of multiple genes involved in cell growth, viability and tissue-specific differentiation. However, the role of RGC32 in tumorigenesis and tumor progression in colorectal cancer (CRC) has not been fully elucidated. Here, we showed that the expression of RGC32 was significantly up-regulated in human CRC tissues versus adjacent normal tissues. RGC32 expression was significantly correlated with invasive and aggressive characteristics of tumor cells, as well as poor survival of CRC patients. We also demonstrated that RGC32 overexpression promoted proliferation, migration and tumorigenic growth of human CRC cells in vitro and in vivo. Functionally, RGC32 facilitated epithelial-mesenchymal transition (EMT) in CRC via the Smad/Sip1 signaling pathway, as shown by decreasing E-cadherin expression and increasing vimentin expression. In conclusion, our findings suggested that overexpression of RGC32 facilitates EMT of CRC cells by activating Smad/Sip1 signaling.
Collapse
Affiliation(s)
- Xiao-Yan Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sheng-Nan Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hui-Fang Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhi-Yan Hu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Zhong
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chuan-Sha Gu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shi-You Chen
- Department of Physiology &Pharmacology, University of Georgia, Athens, GA, United States
| | - Teng-Fei Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zu-Guo Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
19
|
Abou-ElNaga A, Mutawa G, El-Sherbiny IM, Abd-ElGhaffar H, Allam AA, Ajarem J, Mousa SA. Novel Nano-Therapeutic Approach Actively Targets Human Ovarian Cancer Stem Cells after Xenograft into Nude Mice. Int J Mol Sci 2017; 18:ijms18040813. [PMID: 28417924 PMCID: PMC5412397 DOI: 10.3390/ijms18040813] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/16/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023] Open
Abstract
The power of tumorigenesis, chemo-resistance and metastasis in malignant ovarian tumors resides in a tiny population of cancer cells known as ovarian cancer stem cells (OCSCs). Developing nano-therapeutic targeting of OCSCs is considered a great challenge. The potential use of poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) was investigated as a drug delivery system for paclitaxel (PTX) against OCSCs in vitro and in vivo. PTX-loaded PLGA NPs were prepared by an emulsion solvent evaporation method, supported by incorporation of folic acid (FA) as the ligand. NPs were characterized for size, surface morphology, drug loading, and encapsulation efficiency. In vitro cytotoxicity of PTX-loaded FA/PLGA NPs was tested against OCSCs with MTT assay. In vivo anti-tumoral efficiency and active targeting potential of prepared NPs against tumors in nude mice were investigated. In vitro results revealed that IC50 of PTX was significantly reduced after loading on PLGA NPs. On the other hand, in vivo results showed that PLGA NPs enhanced the tumor suppression efficiency of PTX. Investigation with real time quantitative PCR analysis revealed the limiting expression of chemo-resistant genes (ABCG2 and MDR1) after applying PLGA NPs as a drug delivery system for PTX. Histopathological examination of tumors showed the effective biological influence of PTX-loaded FA/PLGA NPs through the appearance of reactive lymphoid follicles. Targeting potential of PTX was activated by FA/PLGA NPs through significant preservation of body weight (p < 0.0001) and minimizing the systemic toxicity in healthy tissues. Immunohistochemical investigation revealed a high expression of apoptotic markers in tumor tissue, supporting the targeting effect of FA/PLGA NPs. A drug delivery system based on FA/PLGA NPs can enhance PTX’s in vitro cytotoxicity and in vivo targeting potential against OCSCs.
Collapse
Affiliation(s)
- Amoura Abou-ElNaga
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura 35516, Egypt.
| | - Ghada Mutawa
- Zoology Department, Faculty of Sciences, Mansoura University, Mansoura 35516, Egypt.
| | - Ibrahim M El-Sherbiny
- Center for Materials Science, Zewail City of Science and Technology, Cairo 12588, Egypt.
| | - Hassan Abd-ElGhaffar
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Ahmed A Allam
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
- Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt.
| | - Jamaan Ajarem
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| |
Collapse
|
20
|
Zhang W, Qi H, Mo X, Sun Q, Li T, Song Q, Xu K, Hu H, Ma D, Wang Y. CMTM8 is Frequently Downregulated in Multiple Solid Tumors. Appl Immunohistochem Mol Morphol 2017; 25:122-128. [DOI: 10.1097/pai.0000000000000274] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
ZHANG SISEN, WU LIHUA. Roles of neural precursor cell expressed, developmentally downregulated 9 in tumor-associated cellular processes (Review). Mol Med Rep 2015; 12:6415-21. [DOI: 10.3892/mmr.2015.4240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 06/15/2015] [Indexed: 11/05/2022] Open
|
22
|
Zollinger A, Davison AC, Goldstein DR. Meta-analysis of incomplete microarray studies. Biostatistics 2015; 16:686-700. [PMID: 25987649 DOI: 10.1093/biostatistics/kxv014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/12/2015] [Indexed: 12/18/2022] Open
Abstract
Meta-analysis of microarray studies to produce an overall gene list is relatively straightforward when complete data are available. When some studies lack information-providing only a ranked list of genes, for example-it is common to reduce all studies to ranked lists prior to combining them. Since this entails a loss of information, we consider a hierarchical Bayes approach to meta-analysis using different types of information from different studies: the full data matrix, summary statistics, or ranks. The model uses an informative prior for the parameter of interest to aid the detection of differentially expressed genes. Simulations show that the new approach can give substantial power gains compared with classical meta-analysis and list aggregation methods. A meta-analysis of 11 published studies with different data types identifies genes known to be involved in ovarian cancer and shows significant enrichment.
Collapse
Affiliation(s)
- Alix Zollinger
- Ecole Polytechnique Fédérale de Lausanne, EPFL-FSB-MATHAA-STAT, Station 8, 1015 Lausanne, Switzerland
| | - Anthony C Davison
- Ecole Polytechnique Fédérale de Lausanne, EPFL-FSB-MATHAA-STAT, Station 8, 1015 Lausanne, Switzerland
| | - Darlene R Goldstein
- Ecole Polytechnique Fédérale de Lausanne, EPFL-FSB-MATHAA-STAT, Station 8, 1015 Lausanne, Switzerland
| |
Collapse
|
23
|
Kwon AY, Kim GI, Jeong JY, Song JY, Kwack KB, Lee C, Kang HY, Kim TH, Heo JH, An HJ. VAV3 Overexpressed in Cancer Stem Cells Is a Poor Prognostic Indicator in Ovarian Cancer Patients. Stem Cells Dev 2015; 24:1521-35. [PMID: 25715123 DOI: 10.1089/scd.2014.0588] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ovarian carcinoma is a highly lethal malignancy due to frequent relapse and drug resistance. Cancer stem cells (CSCs) are thought to contribute significantly to disease relapse and drug resistance. In this study, a subpopulation of CSCs of ovarian carcinoma was isolated and the genes differentially expressed in these cells were identified to characterize CSCs and to find candidate biomarkers. Ovarian carcinoma cells from patients were primarily cultured, and spheroid-forming cells (SFCs) were isolated. The characteristic genes of SFCs were identified through cDNA microarray and validation by quantitative real-time polymerase chain reaction and immunohistochemistry, and the association of their expression with clinicopathologic parameters was analyzed. GSC (4.26-fold), VAV3 (7.05-fold), FOXA2 (12.06-fold), LEF1 (17.26-fold), COMP (21.33-fold), GRIN2A (9.36-fold), CD86 (23.14-fold), PYY (4.18-fold), NKX3-2 (10.35-fold), and PDK4 (74.26-fold) were significantly upregulated in SFCs compared with parental cancer cells. With validation for human ovarian carcinomas, LEF1, PYY, NKX3-2, and WNT3A were significantly upregulated in chemoresistant cancers compared with chemosensitive cancers. Overexpression of LEF1, VAV3, and NKX3-2 was significantly associated with distant metastasis by immunohistochemistry. VAV3 overexpression was an independent poor survival indicator (hazard ratio=15.27, P<0.05) by multivariate Cox analysis. The further functional assay revealed that VAV3 knockdown regulated CSC activation and ovarian cancer cell proliferation and sensitized paclitaxel (PTX)-resistant cancer cells to PTX treatment. Taken together, we identified by high-throughput analysis of CSCs that VAV3 overexpression is a novel biomarker for poor prognosis and survival in ovarian carcinoma.
Collapse
Affiliation(s)
- Ah-Young Kwon
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Gwang-Il Kim
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Ju-Yeon Jeong
- 2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Ji-Ye Song
- 2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Kyu-Beom Kwack
- 3 Department of Biomedical Science, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Chan Lee
- 4 Department of Gynecologic Oncology, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Hae-Youn Kang
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Tae-Heon Kim
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Jin-Hyung Heo
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Hee Jung An
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| |
Collapse
|
24
|
Expression of RGC32 in human normal and preeclamptic placentas and its role in trophoblast cell invasion and migration. Placenta 2015; 36:350-6. [DOI: 10.1016/j.placenta.2014.12.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/18/2014] [Accepted: 12/15/2014] [Indexed: 11/18/2022]
|
25
|
Abstract
Ovarian cancer is the most lethal malignancy of the female genital tract, mainly due to the failure of early diagnosis and the limitations posed by the conventional chemotherapies. Current research has focused in the study of cascades of various cellular molecular reactions, known as signaling pathways. In this review article, authors try to describe the current knowledge regarding the signaling pathways that influence multiple cellular processes in serous ovarian cancer and especially the pathogenesis. Thorough understanding of the precise role of these pathways can lead to the development of new and more effective targeted therapies as well as novel biomarkers in ovarian cancer.
Collapse
|
26
|
Stone RL, Baggerly KA, Armaiz-Pena GN, Kang Y, Sanguino AM, Thanapprapasr D, Dalton HJ, Bottsford-Miller J, Zand B, Akbani R, Diao L, Nick AM, DeGeest K, Lopez-Berestein G, Coleman RL, Lutgendorf S, Sood AK. Focal adhesion kinase: an alternative focus for anti-angiogenesis therapy in ovarian cancer. Cancer Biol Ther 2014; 15:919-29. [PMID: 24755674 DOI: 10.4161/cbt.28882] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
This investigation describes the clinical significance of phosphorylated focal adhesion kinase (FAK) at the major activating tyrosine site (Y397) in epithelial ovarian cancer (EOC) cells and tumor-associated endothelial cells. FAK gene amplification as a mechanism for FAK overexpression and the effects of FAK tyrosine kinase inhibitor VS-6062 on tumor growth, metastasis, and angiogenesis were examined. FAK and phospho-FAK(Y397) were quantified in tumor (FAK-T; pFAK-T) and tumor-associated endothelial (FAK-endo; pFAK-endo) cell compartments of EOCs using immunostaining and qRT-PCR. Associations between expression levels and clinical variables were evaluated. Data from The Cancer Genome Atlas were used to correlate FAK gene copy number and expression levels in EOC specimens. The in vitro and in vivo effects of VS-6062 were assayed in preclinical models. FAK-T and pFAK-T overexpression was significantly associated with advanced stage disease and increased microvessel density (MVD). High MVD was observed in tumors with elevated endothelial cell FAK (59%) and pFAK (44%). Survival was adversely affected by FAK-T overexpression (3.03 vs 2.06 y, P = 0.004), pFAK-T (2.83 vs 1.78 y, P<0.001), and pFAK-endo (2.33 vs 2.17 y, P = 0.005). FAK gene copy number was increased in 34% of tumors and correlated with expression levels (P<0.001). VS-6062 significantly blocked EOC and endothelial cell migration as well as endothelial cell tube formation in vitro. VS-6062 reduced mean tumor weight by 56% (P = 0.005), tumor MVD by 40% (P = 0.0001), and extraovarian metastasis (P<0.01) in orthotopic EOC mouse models. FAK may be a unique therapeutic target in EOC given the dual anti-angiogenic and anti-metastatic potential of FAK inhibitors.
Collapse
Affiliation(s)
- Rebecca L Stone
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Keith A Baggerly
- Department of Bioinformatics and Computational Biology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Guillermo N Armaiz-Pena
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Yu Kang
- Department of Obstetrics and Gynecology; Hospital of Fudan University; Shanghai, PR China
| | - Angela M Sanguino
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Duangmani Thanapprapasr
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Heather J Dalton
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Justin Bottsford-Miller
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Behrouz Zand
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Alpa M Nick
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Koen DeGeest
- Department of Obstetrics and Gynecology; The University of Iowa; Iowa City, IA USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Robert L Coleman
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Susan Lutgendorf
- Department of Psychology; The University of Iowa; Iowa City, IA USA
| | - Anil K Sood
- Department of Gynecologic Oncology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA; Department of Cancer Biology; The University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| |
Collapse
|
27
|
Lisowska KM, Olbryt M, Dudaladava V, Pamuła-Piłat J, Kujawa K, Grzybowska E, Jarząb M, Student S, Rzepecka IK, Jarząb B, Kupryjańczyk J. Gene expression analysis in ovarian cancer - faults and hints from DNA microarray study. Front Oncol 2014; 4:6. [PMID: 24478986 PMCID: PMC3904181 DOI: 10.3389/fonc.2014.00006] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 01/09/2014] [Indexed: 01/22/2023] Open
Abstract
The introduction of microarray techniques to cancer research brought great expectations for finding biomarkers that would improve patients’ treatment; however, the results of such studies are poorly reproducible and critical analyses of these methods are rare. In this study, we examined global gene expression in 97 ovarian cancer samples. Also, validation of results by quantitative RT-PCR was performed on 30 additional ovarian cancer samples. We carried out a number of systematic analyses in relation to several defined clinicopathological features. The main goal of our study was to delineate the molecular background of ovarian cancer chemoresistance and find biomarkers suitable for prediction of patients’ prognosis. We found that histological tumor type was the major source of variability in genes expression, except for serous and undifferentiated tumors that showed nearly identical profiles. Analysis of clinical endpoints [tumor response to chemotherapy, overall survival, disease-free survival (DFS)] brought results that were not confirmed by validation either on the same group or on the independent group of patients. CLASP1 was the only gene that was found to be important for DFS in the independent group, whereas in the preceding experiments it showed associations with other clinical endpoints and with BRCA1 gene mutation; thus, it may be worthy of further testing. Our results confirm that histological tumor type may be a strong confounding factor and we conclude that gene expression studies of ovarian carcinomas should be performed on histologically homogeneous groups. Among the reasons of poor reproducibility of statistical results may be the fact that despite relatively large patients’ group, in some analyses one has to compare small and unequal classes of samples. In addition, arbitrarily performed division of samples into classes compared may not always reflect their true biological diversity. And finally, we think that clinical endpoints of the tumor probably depend on subtle changes in many and, possibly, alternative molecular pathways, and such changes may be difficult to demonstrate.
Collapse
Affiliation(s)
- Katarzyna Marta Lisowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Gliwice , Poland
| | - Magdalena Olbryt
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Gliwice , Poland
| | - Volha Dudaladava
- Department of Medical Biology and Genetics, Grodno State Medical University , Grodno , Belarus
| | - Jolanta Pamuła-Piłat
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Gliwice , Poland
| | - Katarzyna Kujawa
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Gliwice , Poland
| | - Ewa Grzybowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Gliwice , Poland
| | - Michał Jarząb
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Gliwice , Poland
| | - Sebastian Student
- Faculty of Automated Control, Electronics and Computer Science, Silesian University of Technology , Gliwice , Poland
| | - Iwona Krystyna Rzepecka
- Department of Pathology, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Warsaw , Poland
| | - Barbara Jarząb
- Department of Nuclear Medicine and Oncological Endocrinology, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Gliwice , Poland
| | - Jolanta Kupryjańczyk
- Department of Pathology, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology , Warsaw , Poland
| |
Collapse
|
28
|
Janczar S, Graham JS, Paige AJW, Gabra H. Targeting locoregional peritoneal dissemination in ovarian cancer. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17474108.4.2.133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
29
|
Wang H, Mu X, Zhou S, Zhang J, Dai J, Tang L, Xiao L, Duan Z, Jia L, Chen S. NEDD9 overexpression is associated with the progression of and an unfavorable prognosis in epithelial ovarian cancer. Hum Pathol 2013; 45:401-8. [PMID: 24439227 DOI: 10.1016/j.humpath.2013.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 11/16/2022]
Abstract
Neural precursor cell-expressed, developmentally down-regulated 9 (NEDD9), a scaffolding protein, has been identified as a prometastatic and poor prognostic gene in multiple malignant tumors. However, the potential role of the NEDD9 protein in epithelial ovarian cancer (EOC) remains unclear. In the present study, we investigated the expression of NEDD9 and the correlation between NEDD9 expression and prognosis in EOC. NEDD9 expression was detected in 129 archived EOC specimens by immunohistochemical staining and in 28 freshly frozen EOC specimens by Western blotting. The expression of NEDD9 was evaluated in ovarian cancer cell lines by Western blotting and immunofluorescence. The association between the expression of NEDD9 and prognosis was determined by survival analysis. Results suggested that NEDD9 was overexpressed in EOC specimens compared with noninvasive epithelial ovarian tumors and normal ovarian specimens. A high level of NEDD9 expression significantly correlated with advanced-stage tumors (International Federation of Gynecology and Obstetrics classes III-IV, P < .001), high-grade carcinoma (grades 2-3, P < .001), and suboptimal primary cytoreductive surgery (residual disease <1cm, P = .021). The expression level of NEDD9 varied in ovarian cancer cell lines. Multivariate analysis indicated that NEDD9 overexpression (P = .033), advanced stage (P < .001), and high-grade carcinoma (P = .01) were independent predictors of poor survival. In conclusion, NEDD9 is overexpressed and associated with an unfavorable prognosis in EOC. NEDD9 overexpression is an independent factor of poor prognosis and may serve as a potential biomarker in EOC.
Collapse
Affiliation(s)
- Haixia Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Xiaoling Mu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Suiyang Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Jing Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Jiemin Dai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Liangdan Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Lin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Zhaoning Duan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Li Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Luzhou Medical College, Luzhou 626000, People's Republic of China.
| | - Shu Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
30
|
RASSF1A and the Taxol Response in Ovarian Cancer. Mol Biol Int 2012; 2012:263267. [PMID: 22548172 PMCID: PMC3324163 DOI: 10.1155/2012/263267] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 01/31/2012] [Accepted: 02/01/2012] [Indexed: 12/04/2022] Open
Abstract
The RASSF1A tumor suppressor gene is frequently inactivated by promoter methylation in human tumors. The RASSF1A protein forms an endogenous complex with tubulin and promotes the stabilization of microtubules. Loss of RASSF1A expression sensitizes cells to microtubule destabilizing stimuli. We have observed a strong correlation between the loss of RASSF1A expression and the development of Taxol resistance in primary ovarian cancer samples. Thus, we sought to determine if RASSF1A levels could dictate the response to Taxol and whether an epigenetic therapy approach might be able to reverse the Taxol resistant phenotype of RASSF1A negative ovarian tumor cells. We found that knocking down RASSF1A expression in an ovarian cancer cell line inhibited Taxol-mediated apoptosis and promoted cell survival during Taxol treatment. Moreover, using a combination of small molecule inhibitors of DNA Methyl Transferase enzymes, we were able restore RASSF1A expression and Taxol sensitivity. This identifies a role for RASSF1A in modulating the tumor response to Taxol and provides proof of principal for the use of epigenetic therapy to overcome Taxol resistance.
Collapse
|
31
|
Miles GD, Seiler M, Rodriguez L, Rajagopal G, Bhanot G. Identifying microRNA/mRNA dysregulations in ovarian cancer. BMC Res Notes 2012; 5:164. [PMID: 22452920 PMCID: PMC3342161 DOI: 10.1186/1756-0500-5-164] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 03/27/2012] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND MicroRNAs are a class of noncoding RNA molecules that co-regulate the expression of multiple genes via mRNA transcript degradation or translation inhibition. Since they often target entire pathways, they may be better drug targets than genes or proteins. MicroRNAs are known to be dysregulated in many tumours and associated with aggressive or poor prognosis phenotypes. Since they regulate mRNA in a tissue specific manner, their functional mRNA targets are poorly understood. In previous work, we developed a method to identify direct mRNA targets of microRNA using patient matched microRNA/mRNA expression data using an anti-correlation signature. This method, applied to clear cell Renal Cell Carcinoma (ccRCC), revealed many new regulatory pathways compromised in ccRCC. In the present paper, we apply this method to identify dysregulated microRNA/mRNA mechanisms in ovarian cancer using data from The Cancer Genome Atlas (TCGA). METHODS TCGA Microarray data was normalized and samples whose class labels (tumour or normal) were ambiguous with respect to consensus ensemble K-Means clustering were removed. Significantly anti-correlated and correlated genes/microRNA differentially expressed between tumour and normal samples were identified. TargetScan was used to identify gene targets of microRNA. RESULTS We identified novel microRNA/mRNA mechanisms in ovarian cancer. For example, the expression level of RAD51AP1 was found to be strongly anti-correlated with the expression of hsa-miR-140-3p, which was significantly down-regulated in the tumour samples. The anti-correlation signature was present separately in the tumour and normal samples, suggesting a direct causal dysregulation of RAD51AP1 by hsa-miR-140-3p in the ovary. Other pairs of potentially biological relevance include: hsa-miR-145/E2F3, hsa-miR-139-5p/TOP2A, and hsa-miR-133a/GCLC. We also identified sets of positively correlated microRNA/mRNA pairs that are most likely result from indirect regulatory mechanisms. CONCLUSIONS Our findings identify novel microRNA/mRNA relationships that can be verified experimentally. We identify both generic microRNA/mRNA regulation mechanisms in the ovary as well as specific microRNA/mRNA controls which are turned on or off in ovarian tumours. Our results suggest that the disease process uses specific mechanisms which may be significant for their utility as early detection biomarkers or in the development of microRNA therapies in treating ovarian cancers. The positively correlated microRNA/mRNA pairs suggest the existence of novel regulatory mechanisms that proceed via intermediate states (indirect regulation) in ovarian tumorigenesis.
Collapse
|
32
|
Kolbe DL, DeLoia JA, Porter-Gill P, Strange M, Petrykowska HM, Guirguis A, Krivak TC, Brody LC, Elnitski L. Differential analysis of ovarian and endometrial cancers identifies a methylator phenotype. PLoS One 2012; 7:e32941. [PMID: 22403726 PMCID: PMC3293923 DOI: 10.1371/journal.pone.0032941] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 02/02/2012] [Indexed: 11/28/2022] Open
Abstract
Despite improved outcomes in the past 30 years, less than half of all women diagnosed with epithelial ovarian cancer live five years beyond their diagnosis. Although typically treated as a single disease, epithelial ovarian cancer includes several distinct histological subtypes, such as papillary serous and endometrioid carcinomas. To address whether the morphological differences seen in these carcinomas represent distinct characteristics at the molecular level we analyzed DNA methylation patterns in 11 papillary serous tumors, 9 endometrioid ovarian tumors, 4 normal fallopian tube samples and 6 normal endometrial tissues, plus 8 normal fallopian tube and 4 serous samples from TCGA. For comparison within the endometrioid subtype we added 6 primary uterine endometrioid tumors and 5 endometrioid metastases from uterus to ovary. Data was obtained from 27,578 CpG dinucleotides occurring in or near promoter regions of 14,495 genes. We identified 36 locations with significant increases or decreases in methylation in comparisons of serous tumors and normal fallopian tube samples. Moreover, unsupervised clustering techniques applied to all samples showed three major profiles comprising mostly normal samples, serous tumors, and endometrioid tumors including ovarian, uterine and metastatic origins. The clustering analysis identified 60 differentially methylated sites between the serous group and the normal group. An unrelated set of 25 serous tumors validated the reproducibility of the methylation patterns. In contrast, >1,000 genes were differentially methylated between endometrioid tumors and normal samples. This finding is consistent with a generalized regulatory disruption caused by a methylator phenotype. Through DNA methylation analyses we have identified genes with known roles in ovarian carcinoma etiology, whereas pathway analyses provided biological insight to the role of novel genes. Our finding of differences between serous and endometrioid ovarian tumors indicates that intervention strategies could be developed to specifically address subtypes of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Diana L. Kolbe
- DIR/GTB Genomic Functional Analysis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Julie A. DeLoia
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
- School of Public Health and Health Services, George Washington University, Washington DC, United States of America
| | - Patricia Porter-Gill
- DIR/GTB Molecular Pathogenesis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mary Strange
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
| | - Hanna M. Petrykowska
- DIR/GTB Genomic Functional Analysis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alfred Guirguis
- Department of Obstetrics and Gynecology, Rush University, Chicago, Illinois, United States of America
| | - Thomas C. Krivak
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, United States of America
| | - Lawrence C. Brody
- DIR/GTB Molecular Pathogenesis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Laura Elnitski
- DIR/GTB Genomic Functional Analysis Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
33
|
Ahn J, Sanz-Moreno V, Marshall CJ. The metastasis gene NEDD9 product acts through integrin β3 and Src to promote mesenchymal motility and inhibit amoeboid motility. J Cell Sci 2012; 125:1814-26. [PMID: 22328516 DOI: 10.1242/jcs.101444] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Neural precursor expressed, developmentally down-regulated 9 (NEDD9), a member of the Cas family of signal transduction molecules, is amplified at the genetic level in melanoma, and elevated expression levels have been shown to correlate with melanoma progression and metastasis. NEDD9 interacts with the guanine nucleotide exchange factor DOCK3 to promote Rac activation and the elongated, mesenchymal-type of tumour cell invasion, but the molecular mechanisms through which NEDD9 promotes melanoma metastasis are not fully understood. We show that signalling through increased NEDD9 levels requires integrin β3 signalling, which leads to elevated phosphorylation of integrin β3. This results in increased Src and FAK but decreased ROCK signalling to drive elongated, mesenchymal-type invasion in environments that contain vitronectin. NEDD9 overexpression does not affect ROCK signalling through activation of RhoA but decreases ROCKII signalling through Src-dependent phosphorylation of a negative regulatory site Tyr722. In NEDD9-overexpressing melanoma cells, inhibition of Src with dasatinib results in a switch from Rac-driven elongated, mesenchymal-type invasion to ROCK-dependent rounded, amoeboid invasion. These findings brings into question whether dasatinib would work as a therapeutic agent to block melanoma invasion and metastasis. On the basis of the in vitro data presented here, a combination treatment of dasatinib and a ROCK inhibitor might be a better alternative in order to inhibit both elongated, mesenchymal-type and rounded, amoeboid motility.
Collapse
Affiliation(s)
- Jessica Ahn
- Division of Cancer Biology, Institute of Cancer Research, Cancer Research UK Centre Tumour Cell Signalling Unit, London, UK
| | | | | |
Collapse
|
34
|
Taylor-Harding B, Agadjanian H, Nassanian H, Kwon S, Guo X, Miller C, Karlan BY, Orsulic S, Walsh CS. Indole-3-carbinol synergistically sensitises ovarian cancer cells to bortezomib treatment. Br J Cancer 2011; 106:333-43. [PMID: 22166800 PMCID: PMC3261668 DOI: 10.1038/bjc.2011.546] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Bortezomib is a proteasome inhibitor with minimal clinical activity as a monotherapy in solid tumours, but its combination with other targeted therapies is being actively investigated as a way to increase its anticarcinogenic properties. Here, we evaluate the therapeutic potential of co-treatment with bortezomib and indole-3-carbinol (I3C), a natural compound found in cruciferous vegetables, in human ovarian cancer. METHODS We examined the effects of I3C, bortezomib and cisplatin in several human ovarian cancer cell lines. Synergy was determined using proliferation assays and isobologram analysis. Cell cycle and apoptotic effects were assessed by flow cytometry. The mechanism of I3C and bortezomib action was determined by RNA microarray studies, quantitative RT-PCR and western blotting. Antitumour activity of I3C and bortezomib was evaluated using an OVCAR5 xenograft mouse model. RESULTS I3C sensitised ovarian cancer cell lines to bortezomib treatment through potent synergistic mechanisms. Combination treatment with bortezomib and I3C led to profound cell cycle arrest and apoptosis as well as disruptions to multiple pathways, including those regulating endoplasmic reticulum stress, cytoskeleton, chemoresistance and carcinogen metabolism. Moreover, I3C and bortezomib co-treatment sensitised ovarian cancer cells to the standard chemotherapeutic agents, cisplatin and carboplatin. Importantly, in vivo studies demonstrated that co-treatment with I3C and bortezomib significantly inhibited tumour growth and reduced tumour weight compared with either drug alone. CONCLUSION Together, these data provide a novel rationale for the clinical application of I3C and bortezomib in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- B Taylor-Harding
- Women's Cancer Program and Division of Gynecologic Oncology, Burns and Allen Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Upregulation of the cell-cycle regulator RGC-32 in Epstein-Barr virus-immortalized cells. PLoS One 2011; 6:e28638. [PMID: 22163048 PMCID: PMC3232240 DOI: 10.1371/journal.pone.0028638] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 11/11/2011] [Indexed: 12/19/2022] Open
Abstract
Epstein-Barr virus (EBV) is implicated in the pathogenesis of multiple human tumours of lymphoid and epithelial origin. The virus infects and immortalizes B cells establishing a persistent latent infection characterized by varying patterns of EBV latent gene expression (latency 0, I, II and III). The CDK1 activator, Response Gene to Complement-32 (RGC-32, C13ORF15), is overexpressed in colon, breast and ovarian cancer tissues and we have detected selective high-level RGC-32 protein expression in EBV-immortalized latency III cells. Significantly, we show that overexpression of RGC-32 in B cells is sufficient to disrupt G2 cell-cycle arrest consistent with activation of CDK1, implicating RGC-32 in the EBV transformation process. Surprisingly, RGC-32 mRNA is expressed at high levels in latency I Burkitt's lymphoma (BL) cells and in some EBV-negative BL cell-lines, although RGC-32 protein expression is not detectable. We show that RGC-32 mRNA expression is elevated in latency I cells due to transcriptional activation by high levels of the differentially expressed RUNX1c transcription factor. We found that proteosomal degradation or blocked cytoplasmic export of the RGC-32 message were not responsible for the lack of RGC-32 protein expression in latency I cells. Significantly, analysis of the ribosomal association of the RGC-32 mRNA in latency I and latency III cells revealed that RGC-32 transcripts were associated with multiple ribosomes in both cell-types implicating post-initiation translational repression mechanisms in the block to RGC-32 protein production in latency I cells. In summary, our results are the first to demonstrate RGC-32 protein upregulation in cells transformed by a human tumour virus and to identify post-initiation translational mechanisms as an expression control point for this key cell-cycle regulator.
Collapse
|
36
|
Fekete T, Rásó E, Pete I, Tegze B, Liko I, Munkácsy G, Sipos N, Rigó J, Györffy B. Meta-analysis of gene expression profiles associated with histological classification and survival in 829 ovarian cancer samples. Int J Cancer 2011; 131:95-105. [PMID: 21858809 DOI: 10.1002/ijc.26364] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 06/27/2011] [Indexed: 01/16/2023]
Abstract
Transcriptomic analysis of global gene expression in ovarian carcinoma can identify dysregulated genes capable to serve as molecular markers for histology subtypes and survival. The aim of our study was to validate previous candidate signatures in an independent setting and to identify single genes capable to serve as biomarkers for ovarian cancer progression. As several datasets are available in the GEO today, we were able to perform a true meta-analysis. First, 829 samples (11 datasets) were downloaded, and the predictive power of 16 previously published gene sets was assessed. Of these, eight were capable to discriminate histology subtypes, and none was capable to predict survival. To overcome the differences in previous studies, we used the 829 samples to identify new predictors. Then, we collected 64 ovarian cancer samples (median relapse-free survival 24.5 months) and performed TaqMan Real Time Polimerase Chain Reaction (RT-PCR) analysis for the best 40 genes associated with histology subtypes and survival. Over 90% of subtype-associated genes were confirmed. Overall survival was effectively predicted by hormone receptors (PGR and ESR2) and by TSPAN8. Relapse-free survival was predicted by MAPT and SNCG. In summary, we successfully validated several gene sets in a meta-analysis in large datasets of ovarian samples. Additionally, several individual genes identified were validated in a clinical cohort.
Collapse
Affiliation(s)
- Tibor Fekete
- Semmelweis University, 1st Department of Gynecology, Budapest.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hagemann AR, Hagemann IS, Cadungog M, Hwang WT, Patel P, Lal P, Hammond R, Gimotty PA, Chu CS, Rubin SC, Birrer MJ, Powell DJ, Feldman MD, Coukos G. Tissue-based immune monitoring II: multiple tumor sites reveal immunologic homogeneity in serous ovarian carcinoma. Cancer Biol Ther 2011; 12:367-77. [PMID: 21785280 DOI: 10.4161/cbt.12.4.16908] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The presence of tumor-infiltrating lymphocytes (TILs) in epithelial ovarian cancer indicates a host antitumor response and is associated with improved survival. We wished to determine the extent to which TIL density differs from site to site within a given patient. We initially studied multiple paired metastases from serous ovarian carcinoma obtained at the time of primary debulking. The expression of genes in specific immune-related pathways was profiled on a pilot set of five patients. We then used immunohistochemistry and quantitative PCR to estimate the density of CD3+, CD8+, and FoxP3+ TILs in these same tumors. To extend the findings to a larger cohort, we semiquantitatively measured intraepithelial and stromal TILs in a tissue microarray (TMA) containing both primary tumors and metastases from 50 patients. In the pilot group, genes related to antimicrobial signaling and TGF-beta signaling showed between-site heterogeneity, whereas cytokines and antigen presentation transcripts were more homogeneous in any given patient. IHC and qPCR for T cell markers were concordant. In the TMA cohort, 2-way ANOVA showed that TIL heterogeneity between sites was present in some but not all patients. The stroma of extra-ovarian metastases showed significantly greater TIL infiltration than ovarian sites. A simulation showed that at clinically meaningful levels of precision, up to 3% of patients will be misclassified for intraepithelial TILs by a single biopsy. In conclusion, between-site heterogeneity exists in some patients with metastatic serous ovarian cancer. The predictive value of biopsies should be considered in clinical trial design.
Collapse
Affiliation(s)
- Andrea R Hagemann
- Ovarian Cancer Research Center, Division of Gynecologic Oncology, University of Pennsylvania, Philadelphia, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hagemann AR, Cadungog M, Hagemann IS, Hammond R, Adams SF, Chu CS, Rubin SC, Zhang L, Addya K, Birrer MJ, Gimotty PA, Coukos G. Tissue-based immune monitoring I: tumor core needle biopsies allow in-depth interrogation of the tumor microenvironment. Cancer Biol Ther 2011; 12:357-66. [PMID: 21785264 DOI: 10.4161/cbt.12.4.16951] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We sought to assess the feasibility and reproducibility of performing tissue-based immune characterization of the tumor microenvironment using CT-compatible needle biopsy material. Three independent biopsies were obtained intraoperatively from one metastatic epithelial ovarian cancer lesion of 7 consecutive patients undergoing surgical cytoreduction using a 16-gauge core biopsy needle. Core specimens were snap-frozen and subjected to immunohistochemistry (IHC) against human CD3, CD4, CD8, and FoxP3. A portion of the cores was used to isolate RNA for 1) real-time quantitative (q)PCR for CD3, CD4, CD8, FoxP3, IL-10 and TGF-beta, 2) multiplexed PCR-based T cell receptor (TCR) CDR3 Vβ region spectratyping, and 3) gene expression profiling. Pearson's correlations were examined for immunohistochemistry and PCR gene expression, as well as for gene expression array data obtained from different tumor biopsies. Needle biopsy yielded sufficient tissue for all assays in all patients. IHC was highly reproducible and informative. Significant correlations were seen between the frequency of CD3+, CD8+ and FoxP3+ T cells by IHC with CD3ε, CD8A, and FoxP3 gene expression, respectively, by qPCR (r=0.61, 0.86, and 0.89; all p< 0.05). CDR3 spectratyping was feasible and highly reproducible in each tumor, and indicated a restricted repertoire for specific TCR Vβ chains in tumor-infiltrating T cells. Microarray gene expression revealed strong correlation between different biopsies collected from the same tumor. Our results demonstrate a feasible and reproducible method of immune monitoring using CT-compatible needle biopsies from tumor tissue, thereby paving the way for sophisticated translational studies during tumor biological therapy.
Collapse
Affiliation(s)
- Andrea R Hagemann
- Ovarian Cancer Research Center, Division of Gynecologic Oncology, University of Pennsylvania, Philadelphia, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Stany MP, Vathipadiekal V, Ozbun L, Stone RL, Mok SC, Xue H, Kagami T, Wang Y, McAlpine JN, Bowtell D, Gout PW, Miller DM, Gilks CB, Huntsman DG, Ellard SL, Wang YZ, Vivas-Mejia P, Lopez-Berestein G, Sood AK, Birrer MJ. Identification of novel therapeutic targets in microdissected clear cell ovarian cancers. PLoS One 2011; 6:e21121. [PMID: 21754983 PMCID: PMC3130734 DOI: 10.1371/journal.pone.0021121] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 05/19/2011] [Indexed: 01/01/2023] Open
Abstract
Clear cell ovarian cancer is an epithelial ovarian cancer histotype that is less responsive to chemotherapy and carries poorer prognosis than serous and endometrioid histotypes. Despite this, patients with these tumors are treated in a similar fashion as all other ovarian cancers. Previous genomic analysis has suggested that clear cell cancers represent a unique tumor subtype. Here we generated the first whole genomic expression profiling using epithelial component of clear cell ovarian cancers and normal ovarian surface specimens isolated by laser capture microdissection. All the arrays were analyzed using BRB ArrayTools and PathwayStudio software to identify the signaling pathways. Identified pathways validated using serous, clear cell cancer cell lines and RNAi technology. In vivo validations carried out using an orthotopic mouse model and liposomal encapsulated siRNA. Patient-derived clear cell and serous ovarian tumors were grafted under the renal capsule of NOD-SCID mice to evaluate the therapeutic potential of the identified pathway. We identified major activated pathways in clear cells involving in hypoxic cell growth, angiogenesis, and glucose metabolism not seen in other histotypes. Knockdown of key genes in these pathways sensitized clear cell ovarian cancer cell lines to hypoxia/glucose deprivation. In vivo experiments using patient derived tumors demonstrate that clear cell tumors are exquisitely sensitive to antiangiogenesis therapy (i.e. sunitinib) compared with serous tumors. We generated a histotype specific, gene signature associated with clear cell ovarian cancer which identifies important activated pathways critical for their clinicopathologic characteristics. These results provide a rational basis for a radically different treatment for ovarian clear cell patients.
Collapse
Affiliation(s)
- Michael P. Stany
- Walter Reed Army Medical Center, Washington D.C., United States of America
| | - Vinod Vathipadiekal
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Laurent Ozbun
- Cell and Cancer Biology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rebecca L. Stone
- Department of Gynecologic Oncology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Samuel C. Mok
- Brigham and Women's Hospital, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Hui Xue
- Living Tumor Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Takashi Kagami
- Living Tumor Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Yuwei Wang
- Living Tumor Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Jessica N. McAlpine
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Peter W. Gout
- Living Tumor Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Dianne M. Miller
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of British Columbia, Vancouver, British Columbia, Canada
| | - C. Blake Gilks
- Department of Pathology, Genetic Pathology Evaluation Centre, Vancouver General Hospital, Centre for Translation and Applied Genomics, British Columbia Cancer Agency and University of British Columbia, Vancouver, British Columbia, Canada
| | - David G. Huntsman
- Department of Pathology, Genetic Pathology Evaluation Centre, Vancouver General Hospital, Centre for Translation and Applied Genomics, British Columbia Cancer Agency and University of British Columbia, Vancouver, British Columbia, Canada
| | - Susan L. Ellard
- Department of Medical Oncology, British Columbia Cancer Agency - Southern Interior, Kelowna, British Columbia, Canada
| | - Yu-Zhuo Wang
- Living Tumor Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pablo Vivas-Mejia
- Department of Experimental Therapeutics, M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Cancer Biology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, M. D. Anderson Cancer Center, Texas, United States of America
| | - Anil K. Sood
- Department of Gynecologic Oncology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Cancer Biology, M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Center for RNA Interference and Non-Coding RNA, M. D. Anderson Cancer Center, Texas, United States of America
| | - Michael J. Birrer
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
40
|
Macitentan (ACT-064992), a tissue-targeting endothelin receptor antagonist, enhances therapeutic efficacy of paclitaxel by modulating survival pathways in orthotopic models of metastatic human ovarian cancer. Neoplasia 2011; 13:167-79. [PMID: 21403842 DOI: 10.1593/neo.10806] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 10/18/2010] [Accepted: 10/22/2010] [Indexed: 11/18/2022] Open
Abstract
Potential treatments for ovarian cancers that have become resistant to standard chemotherapies include modulators of tumor cell survival, such as endothelin receptor (ETR) antagonist. We investigated the therapeutic efficacy of the dual ETR antagonist, macitentan, on human ovarian cancer cells, SKOV3ip1 and IGROV1, growing orthotopically in nude mice. Mice with established disease were treated with vehicle (control), paclitaxel (weekly, intraperitoneal injections), macitentan (daily oral administrations), or a combination of paclitaxel and macitentan. Treatment with paclitaxel decreased tumor weight and volume of ascites. Combination therapy with macitentan and paclitaxel reduced tumor incidence and further reduced tumor weight and volume of ascites when compared with paclitaxel alone. Macitentan alone occasionally reduced tumor weight but alone had no effect on tumor incidence or ascites. Immunohistochemical analyses revealed that treatment with macitentan and macitentan plus paclitaxel inhibited the phosphorylation of ETRs and suppressed the survival pathways of tumor cells by decreasing the levels of pVEGFR2, pAkt, and pMAPK. The dose of macitentan necessary for inhibition of phosphorylation correlated with the dose required to increase antitumor efficacy of paclitaxel. Treatment with macitentan enhanced the cytotoxicity mediated by paclitaxel as measured by the degree of apoptosis in tumor cells and tumor-associated endothelial cells. Collectively, these results show that administration of macitentan in combination with paclitaxel prevents the progression of ovarian cancer in the peritoneal cavity of nude mice in part by inhibiting survival pathways of both tumor cells and tumor-associated endothelial cells.
Collapse
|
41
|
Emmanuel C, Gava N, Kennedy C, Balleine RL, Sharma R, Wain G, Brand A, Hogg R, Etemadmoghadam D, George J, Birrer MJ, Clarke CL, Chenevix-Trench G, Bowtell DDL, Harnett PR, deFazio A. Comparison of expression profiles in ovarian epithelium in vivo and ovarian cancer identifies novel candidate genes involved in disease pathogenesis. PLoS One 2011; 6:e17617. [PMID: 21423607 PMCID: PMC3057977 DOI: 10.1371/journal.pone.0017617] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 02/02/2011] [Indexed: 12/30/2022] Open
Abstract
Molecular events leading to epithelial ovarian cancer are poorly understood but
ovulatory hormones and a high number of life-time ovulations with concomitant
proliferation, apoptosis, and inflammation, increases risk. We identified genes
that are regulated during the estrous cycle in murine ovarian surface epithelium
and analysed these profiles to identify genes dysregulated in human ovarian
cancer, using publically available datasets. We identified 338 genes that are
regulated in murine ovarian surface epithelium during the estrous cycle and
dysregulated in ovarian cancer. Six of seven candidates selected for
immunohistochemical validation were expressed in serous ovarian cancer,
inclusion cysts, ovarian surface epithelium and in fallopian tube epithelium.
Most were overexpressed in ovarian cancer compared with ovarian surface
epithelium and/or inclusion cysts (EpCAM, EZH2, BIRC5) although BIRC5 and EZH2
were expressed as highly in fallopian tube epithelium as in ovarian cancer. We
prioritised the 338 genes for those likely to be important for ovarian cancer
development by in silico analyses of copy number aberration and
mutation using publically available datasets and identified genes with
established roles in ovarian cancer as well as novel genes for which we have
evidence for involvement in ovarian cancer. Chromosome segregation emerged as an
important process in which genes from our list of 338 were over-represented
including two (BUB1, NCAPD2) for which there
is evidence of amplification and mutation. NUAK2, upregulated in ovarian surface
epithelium in proestrus and predicted to have a driver mutation in ovarian
cancer, was examined in a larger cohort of serous ovarian cancer where patients
with lower NUAK2 expression had shorter overall survival. In conclusion,
defining genes that are activated in normal epithelium in the course of
ovulation that are also dysregulated in cancer has identified a number of
pathways and novel candidate genes that may contribute to the development of
ovarian cancer.
Collapse
Affiliation(s)
- Catherine Emmanuel
- Department of Gynaecological Oncology, Westmead Hospital, Westmead, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Li Y, Bavarva JH, Wang Z, Guo J, Qian C, Thibodeau SN, Golemis EA, Liu W. HEF1, a novel target of Wnt signaling, promotes colonic cell migration and cancer progression. Oncogene 2011; 30:2633-43. [PMID: 21317929 DOI: 10.1038/onc.2010.632] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Misregulation of the canonical Wnt/β-catenin pathway and aberrant activation of Wnt signaling target genes are common in colorectal cancer (CRC) and contribute to cancer progression. Altered expression of human enhancer of filamentation 1 (HEF1; also known as NEDD9 or Cas-L) has been implicated in progression of melanoma, breast, and CRC. However, the regulation of HEF1 and the role of HEF1 in CRC tumorigenesis are not fully understood. We here identify HEF1 as a novel Wnt signaling target. The expression of HEF1 was upregulated by Wnt-3a, β-catenin, and Dvl2 in a dose-dependent manner, and was suppressed following β-catenin downregulation by shRNA. In addition, elevated HEF1 mRNA and protein levels were observed in CRC cell lines and primary tumor tissues, as well as in the colon and adenoma polyps of Apc(Min/+) mice. Moreover, HEF1 levels in human colorectal tumor tissues increased with the tumor grade. Chromatin immunoprecipitation (ChIP) assays and promoter analyses revealed three functional T-cell factor (TCF)-binding sites in the promoter of HEF1 responsible for HEF1 induction by Wnt signaling. Ectopic expression of HEF1 increased cell proliferation and colony formation, while downregulation of HEF1 in SW480 cells by shRNA had the opposite effects and inhibited the xenograft tumor growth. Furthermore, overexpression of HEF1 in SW480 cells promoted cell migration and invasion. Together, our results determined a novel role of HEF1 as a mediator of the canonical Wnt/β-catenin signaling pathway for cell proliferation, migration, and tumorigenesis, as well as an important player in colorectal tumorigenesis and progression. HEF1 may represent an attractive candidate for drug targeting in CRC.
Collapse
Affiliation(s)
- Y Li
- Department of Genetics, Louisiana State University Health Sciences Center/Stanley S Scott Cancer Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Lu C, Han HD, Mangala LS, Ali-Fehmi R, Newton CS, Ozbun L, Armaiz-Pena GN, Hu W, Stone RL, Munkarah A, Ravoori MK, Shahzad MMK, Lee JW, Mora E, Langley RR, Carroll AR, Matsuo K, Spannuth WA, Schmandt R, Jennings NB, Goodman BW, Jaffe RB, Nick AM, Kim HS, Guven EO, Chen YH, Li LY, Hsu MC, Coleman RL, Calin GA, Denkbas EB, Lim JY, Lee JS, Kundra V, Birrer MJ, Hung MC, Lopez-Berestein G, Sood AK. Regulation of tumor angiogenesis by EZH2. Cancer Cell 2010; 18:185-97. [PMID: 20708159 PMCID: PMC2923653 DOI: 10.1016/j.ccr.2010.06.016] [Citation(s) in RCA: 306] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 02/15/2010] [Accepted: 06/24/2010] [Indexed: 02/03/2023]
Abstract
Although VEGF-targeted therapies are showing promise, new angiogenesis targets are needed to make additional gains. Here, we show that increased Zeste homolog 2 (EZH2) expression in either tumor cells or in tumor vasculature is predictive of poor clinical outcome. The increase in endothelial EZH2 is a direct result of VEGF stimulation by a paracrine circuit that promotes angiogenesis by methylating and silencing vasohibin1 (vash1). Ezh2 silencing in the tumor-associated endothelial cells inhibited angiogenesis mediated by reactivation of VASH1, and reduced ovarian cancer growth, which is further enhanced in combination with ezh2 silencing in tumor cells. Collectively, these data support the potential for targeting ezh2 as an important therapeutic approach.
Collapse
Affiliation(s)
- Chunhua Lu
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Hee Dong Han
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Lingegowda S. Mangala
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Rouba Ali-Fehmi
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201
| | - Christopher S. Newton
- Department of Cell and Cancer Biology, National Cancer Institute, Bethesda, MD 20892
| | - Laurent Ozbun
- Department of Cell and Cancer Biology, National Cancer Institute, Bethesda, MD 20892
| | - Guillermo N. Armaiz-Pena
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Wei Hu
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Rebecca L. Stone
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Adnan Munkarah
- Women’s Health Services, Henry Ford Health System, Detroit, MI 48202
| | - Murali K. Ravoori
- Department of Experimental Diagnostic Imaging, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 368, Houston, TX 77030
| | - Mian M. K. Shahzad
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
- Baylor College of Medicine, Department of Obstetrics and Gynecology, Houston, TX 77030
| | - Jeong-Won Lee
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea 135-710
| | - Edna Mora
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
- Department of Surgery, University of Puerto Rico, San Juan, PR 00935
| | - Robert R. Langley
- Department of Cancer Biology, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030
| | - Amy R. Carroll
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Koji Matsuo
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Whitney A. Spannuth
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Rosemarie Schmandt
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Nicholas B. Jennings
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Blake W. Goodman
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Robert B. Jaffe
- Center for Reproductive Sciences, 505 Parnassus, University of California, San Francisco, CA 94143
| | - Alpa M. Nick
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
| | - Hye Sun Kim
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
- Department of Pathology, Cheil General Hospital and Women’s Healthcare Center, Kwandong University College of Medicine, Seoul, Korea 100-380
| | - Eylem Ozturk Guven
- Hacettepe University, Nanotechnology and Nanomedicine Division, Ankara, Turkey 06532
| | - Ya-Huey Chen
- Center for Molecular Medicine, China Medical University and Hospital, Taichung, Taiwan 404
| | - Long-Yuan Li
- Graduate Institute of Cancer Biology, China Medical University and Hospital, Taichung, Taiwan 404
| | - Ming-Chuan Hsu
- Department of Cellular and Molecular Oncology, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
| | - Robert L. Coleman
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
- Center for RNAi and Non-Coding RNA, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
| | - George A. Calin
- Center for RNAi and Non-Coding RNA, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
- Department of Experimental Therapeutics, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
| | - Emir B. Denkbas
- Hacettepe University, Nanotechnology and Nanomedicine Division, Ankara, Turkey 06532
| | - Jae Yun Lim
- Department of Systems Biology, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
| | - Ju-Seog Lee
- Department of Systems Biology, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
| | - Vikas Kundra
- Department of Experimental Diagnostic Imaging, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 368, Houston, TX 77030
| | - Michael J. Birrer
- Department of Medicine, Harvard Medical School, Massachusetts General Hospital Cancer Center, Boston, MA 02114
| | - Mien-Chie Hung
- Center for Molecular Medicine, China Medical University and Hospital, Taichung, Taiwan 404
- Department of Cellular and Molecular Oncology, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
| | - Gabriel Lopez-Berestein
- Department of Cancer Biology, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030
- Center for RNAi and Non-Coding RNA, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
- Department of Experimental Therapeutics, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
| | - Anil K. Sood
- Department of Gynecologic Oncology, U.T. M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030
- Department of Cancer Biology, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030
- Center for RNAi and Non-Coding RNA, U.T. M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Unit 950, Houston, TX 77030
- Correspondence and Reprint Requests: Anil K. Sood, Professor, Departments of Gynecologic Oncology and Cancer Biology, The University of Texas, M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030 Phone: 713-745-5266, Fax: 713-792-7586,
| |
Collapse
|
44
|
Han HD, Mangala LS, Lee JW, Shahzad MMK, Kim HS, Shen D, Nam EJ, Mora EM, Stone RL, Lu C, Lee SJ, Roh JW, Nick AM, Lopez-Berestein G, Sood AK. Targeted gene silencing using RGD-labeled chitosan nanoparticles. Clin Cancer Res 2010; 16:3910-22. [PMID: 20538762 PMCID: PMC2912984 DOI: 10.1158/1078-0432.ccr-10-0005] [Citation(s) in RCA: 212] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE This study aimed to develop an Arg-Gly-Asp (RGD) peptide-labeled chitosan nanoparticle (RGD-CH-NP) as a novel tumor targeted delivery system for short interfering RNA (siRNA). EXPERIMENTAL DESIGN RGD peptide conjugated with chitosan by thiolation reaction was confirmed by proton-NMR (H-NMR). Binding of RGD-CH-NP with alphanubeta3 integrin was examined by flow cytometry and fluorescence microscopy. Antitumor efficacy was examined in orthotopic mouse models of ovarian carcinoma. RESULTS We show that RGD-CH-NP loaded with siRNA significantly increased selective intratumoral delivery in orthotopic animal models of ovarian cancer. In addition, we show targeted silencing of multiple growth-promoting genes (POSTN, FAK, and PLXDC1) along with therapeutic efficacy in the SKOV3ip1, HeyA8, and A2780 models using siRNA incorporated into RGD-CH-NP (siRNA/RGD-CH-NP). Furthermore, we show in vivo tumor vascular targeting using RGD-CH-NP by delivering PLXDC1-targeted siRNA into the alphanubeta3 integrin-positive tumor endothelial cells in the A2780 tumor-bearing mice. This approach resulted in significant inhibition of tumor growth compared with controls. CONCLUSIONS This study shows that RGD-CH-NP is a novel and highly selective delivery system for siRNA with the potential for broad applications in human disease.
Collapse
Affiliation(s)
- Hee Dong Han
- Department of Gynecologic Oncology, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Johnatty SE, Beesley J, Chen X, Macgregor S, Duffy DL, Spurdle AB, deFazio A, Gava N, Webb PM, Rossing MA, Doherty JA, Goodman MT, Lurie G, Thompson PJ, Wilkens LR, Ness RB, Moysich KB, Chang-Claude J, Wang-Gohrke S, Cramer DW, Terry KL, Hankinson SE, Tworoger SS, Garcia-Closas M, Yang H, Lissowska J, Chanock SJ, Pharoah PD, Song H, Whitemore AS, Pearce CL, Stram DO, Wu AH, Pike MC, Gayther SA, Ramus SJ, Menon U, Gentry-Maharaj A, Anton-Culver H, Ziogas A, Hogdall E, Kjaer SK, Hogdall C, Berchuck A, Schildkraut JM, Iversen ES, Moorman PG, Phelan CM, Sellers TA, Cunningham JM, Vierkant RA, Rider DN, Goode EL, Haviv I, Chenevix-Trench G. Evaluation of candidate stromal epithelial cross-talk genes identifies association between risk of serous ovarian cancer and TERT, a cancer susceptibility "hot-spot". PLoS Genet 2010; 6:e1001016. [PMID: 20628624 PMCID: PMC2900295 DOI: 10.1371/journal.pgen.1001016] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 06/03/2010] [Indexed: 11/19/2022] Open
Abstract
We hypothesized that variants in genes expressed as a consequence of interactions between ovarian cancer cells and the host micro-environment could contribute to cancer susceptibility. We therefore used a two-stage approach to evaluate common single nucleotide polymorphisms (SNPs) in 173 genes involved in stromal epithelial interactions in the Ovarian Cancer Association Consortium (OCAC). In the discovery stage, cases with epithelial ovarian cancer (n=675) and controls (n=1,162) were genotyped at 1,536 SNPs using an Illumina GoldenGate assay. Based on Positive Predictive Value estimates, three SNPs-PODXL rs1013368, ITGA6 rs13027811, and MMP3 rs522616-were selected for replication using TaqMan genotyping in up to 3,059 serous invasive cases and 8,905 controls from 16 OCAC case-control studies. An additional 18 SNPs with Pper-allele<0.05 in the discovery stage were selected for replication in a subset of five OCAC studies (n=1,233 serous invasive cases; n=3,364 controls). The discovery stage associations in PODXL, ITGA6, and MMP3 were attenuated in the larger replication set (adj. Pper-allele>or=0.5). However genotypes at TERT rs7726159 were associated with ovarian cancer risk in the smaller, five-study replication study (Pper-allele=0.03). Combined analysis of the discovery and replication sets for this TERT SNP showed an increased risk of serous ovarian cancer among non-Hispanic whites [adj. ORper-allele 1.14 (1.04-1.24) p=0.003]. Our study adds to the growing evidence that, like the 8q24 locus, the telomerase reverse transcriptase locus at 5p15.33, is a general cancer susceptibility locus.
Collapse
|
46
|
Rosanò L, Spinella F, Bagnato A. The importance of endothelin axis in initiation, progression, and therapy of ovarian cancer. Am J Physiol Regul Integr Comp Physiol 2010; 299:R395-404. [PMID: 20538897 DOI: 10.1152/ajpregu.00304.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The endothelin-1 (ET-1)/ET A receptor (ET(A)R) axis is involved in the pathobiology of different tumors, including ovarian carcinoma. Acting selectively on ET(A)R, ET-1 regulates, through multiple signaling pathways, mitogenesis, cell survival, angiogenesis, lymphangiogenesis, invasion, and metastatic dissemination. Moreover, ET-1/ET(A)R axis appears to be critical in epithelial-to-mesenchymal transition (EMT), providing a mechanism of escape to a new, less adverse niche, in which resistance to apoptosis ensures cell survival in conditions of stress in the primary tumor, and acquisition of "stemness" ensures generation of the critical mass required for tumor progression. Emerging experimental and preclinical data demonstrate that interfering with ET(A)R pathways provides an opportunity for the development of new mechanism-based antitumor strategies by using ET(A)R antagonists alone and in combination with cytotoxic drugs or molecular inhibitors. A specific ET(A)R antagonist in combination with standard chemotherapy is currently evaluated in clinical and translational studies to provide us with new options to treat ovarian cancer and to predict response to therapy. Deeper understanding of molecular mechanism activated by ET(A)R in ovarian cancer will be of paramount importance in the study of ET(A)R-targeted therapy that, regulating EMT and other tumor-associated processes, represents an attractive but challenging approach to improve clinical management of ovarian cancer.
Collapse
Affiliation(s)
- Laura Rosanò
- Molecular Pathology Laboratory, Regina Elena National Cancer Institute, Via delle Messi D'Oro 156, 00158 Rome, Italy
| | | | | |
Collapse
|
47
|
Jochumsen KM, Tan Q, Høgdall EV, Høgdall C, Kjaer SK, Blaakaer J, Kruse TA, Mogensen O. Gene expression profiles as prognostic markers in women with ovarian cancer. Int J Gynecol Cancer 2009; 19:1205-13. [PMID: 19823056 DOI: 10.1111/igc.0b013e3181a3cf55] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The purpose was to find a gene expression profile that could distinguish short-term from long-term survivors in our collection of serous epithelial ovarian carcinomas. Furthermore, it should be able to stratify in an external validation set. Such a classifier profile will take us a step forward toward investigations for more individualized therapies and the use of gene expression profiles in the clinical practice. RNA from tumor tissue from 43 Danish patients with serous epithelial ovarian carcinoma (11 International Federation of Gynecology and Obstetrics [FIGO] stage I/II, 32 FIGO stage III/IV) was analyzed using Affymetrix U133 plus 2.0 microarrays. A multistep statistical procedure was applied to the data to find the gene set that optimally split the patients into short-term and long-term survivors in a Kaplan-Meier plot. A 14-gene prognostic profile with the ability to distinguish short-term survivors (median overall survival of 32 months) from long-term survivors (median overall survival not yet reached after a median follow-up of 76 months) with a P value of 3.4 x 10 was found. The prognostic gene set was also able to distinguish short-term from long-term survival in patients with advanced disease. Furthermore, its ability to classify in an external validation set was demonstrated. The identified 14-gene prognostic profile was able to predict survival (short- vs long-term survival) with a strength that is better than any other prognostic factor in epithelial ovarian cancer including FIGO stage. This stratification method may form the basis of determinations for new therapeutic approaches, as patients with poor prognosis could obtain the biggest advantage from new treatment modalities.
Collapse
Affiliation(s)
- Kirsten M Jochumsen
- Department of Obstetrics and Gynecology, Odense University Hospital, Odense C, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Role of microRNAs in ovarian cancer pathogenesis and potential clinical implications. Int J Biochem Cell Biol 2009; 42:1262-72. [PMID: 20035894 DOI: 10.1016/j.biocel.2009.12.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 12/14/2009] [Accepted: 12/16/2009] [Indexed: 11/24/2022]
Abstract
Despite important improvements over the past two decades, the overall cure rate of epithelial ovarian cancer (EOC) remains only approximately 30%. Although much has been learned about the proteins and pathways involved in early events of malignant transformation and drug resistance, a major challenge still remaining is the identification of markers for early diagnosis and prediction of response to chemotherapy. Recently, it has become clear that alterations in the expression of microRNAs (miRNAs) contribute to the pathogenesis and progression of several human malignancies. In this review we discuss current data concerning the accumulating evidence of the role of miRNAs in EOC pathogenesis and tumor characterization; their dysregulated expression in EOC; and their still undefined role in diagnosis, prognosis and prediction of response to therapy. The most frequently deregulated miRNAs are members of the let-7 and miR-200 families, the latter involved in epithelial-to-mesenchymal transition (EMT). EMT is part of normal ovarian surface epithelium physiology, being the key regulator of the post-ovulatory repair process, and failure to undergo EMT may be one of the events leading to transformation. A general down-modulation of miRNA expression is observed in EOC compared to normal tissue. However, a clear consensus on the miRNA signatures associated with prognosis or prediction of response to therapy has not yet been reached.
Collapse
|
49
|
Toxicogenomic analysis of N-nitrosomorpholine induced changes in rat liver: Comparison of genomic and proteomic responses and anchoring to histopathological parameters. Toxicol Appl Pharmacol 2009; 241:230-45. [DOI: 10.1016/j.taap.2009.08.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 08/18/2009] [Indexed: 01/26/2023]
|
50
|
Vlaicu SI, Tegla CA, Cudrici CD, Fosbrink M, Nguyen V, Azimzadeh P, Rus V, Chen H, Mircea PA, Shamsuddin A, Rus H. Epigenetic modifications induced by RGC-32 in colon cancer. Exp Mol Pathol 2009; 88:67-76. [PMID: 19883641 DOI: 10.1016/j.yexmp.2009.10.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 10/23/2009] [Accepted: 10/26/2009] [Indexed: 10/20/2022]
Abstract
First described as a cell cycle activator, RGC-32 is both an activator and a substrate for CDC2. Deregulation of RGC-32 expression has been detected in a wide variety of human cancers. We have now shown that RGC-32 is expressed in precancerous states, and its expression is significantly higher in adenomas than in normal colon tissue. The expression of RGC-32 was higher in advanced stages of colon cancer than in precancerous states or the initial stages of colon cancer. In order to identify the genes that are regulated by RGC-32, we used gene array analysis to investigate the effect of RGC-32 knockdown on gene expression in the SW480 colon cancer cell line. Of the 230 genes that were differentially regulated after RGC-32 knockdown, a group of genes involved in chromatin assembly were the most significantly regulated in these cells: RGC-32 knockdown induced an increase in acetylation of histones H2B lysine 5 (H2BK5), H2BK15, H3K9, H3K18, and H4K8. RGC-32 silencing was also associated with decreased expression of SIRT1 and decreased trimethylation of histone H3K27 (H3K27me3). In addition, RGC-32 knockdown caused a significantly higher percentage of SW480 cells to enter S phase and subsequently G2/M. These data suggest that RGC-32 may contribute to the development of colon cancer by regulating chromatin assembly.
Collapse
Affiliation(s)
- Sonia I Vlaicu
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|