63 results on '"Anil K, Sood"'
Search Results
2. Endothelial p130cas confers resistance to anti-angiogenesis therapy
- Author
-
Yunfei Wen, Anca Chelariu-Raicu, Sujanitha Umamaheswaran, Alpa M. Nick, Elaine Stur, Pahul Hanjra, Dahai Jiang, Nicholas B. Jennings, Xiuhui Chen, Sara Corvigno, Deanna Glassman, Gabriel Lopez-Berestein, Jinsong Liu, Mien-Chie Hung, and Anil K. Sood
- Subjects
Biology (General) ,QH301-705.5 - Published
- 2023
- Full Text
- View/download PDF
3. RNA-binding protein FXR1 drives cMYC translation by recruiting eIF4F complex to the translation start site
- Author
-
Jasmine George, Yongsheng Li, Ishaque P. Kadamberi, Deepak Parashar, Shirng-Wern Tsaih, Prachi Gupta, Anjali Geethadevi, Changliang Chen, Chandrima Ghosh, Yunguang Sun, Sonam Mittal, Ramani Ramchandran, Hallgeir Rui, Gabriel Lopez-Berestein, Cristian Rodriguez-Aguayo, Gustavo Leone, Janet S. Rader, Anil K. Sood, Madhusudan Dey, Sunila Pradeep, and Pradeep Chaluvally-Raghavan
- Subjects
Biology (General) ,QH301-705.5 - Published
- 2023
- Full Text
- View/download PDF
4. Exploiting metabolic vulnerabilities after anti-VEGF antibody therapy in ovarian cancer
- Author
-
Deanna Glassman, Mark S. Kim, Meredith Spradlin, Sunil Badal, Mana Taki, Pratip Bhattacharya, Prasanta Dutta, Charles V. Kingsley, Katherine I. Foster, Olamide Animasahun, Jin Heon Jeon, Abhinav Achreja, Anusha Jayaraman, Praveen Kumar, Minal Nenwani, Fulei Wuchu, Emine Bayraktar, Yutuan Wu, Elaine Stur, Lingegowda Mangala, Sanghoon Lee, Timothy A. Yap, Shannon N. Westin, Livia S. Eberlin, Deepak Nagrath, and Anil K. Sood
- Subjects
Oncology ,Cellular physiology ,Cancer ,Science - Abstract
Summary: Despite modest clinical improvement with anti-vascular endothelial growth factor antibody (AVA) therapy in ovarian cancer, adaptive resistance is ubiquitous and additional options are limited. A dependence on glutamine metabolism, via the enzyme glutaminase (GLS), is a known mechanism of adaptive resistance and we aimed to investigate the utility of a GLS inhibitor (GLSi). Our in vitro findings demonstrated increased glutamine abundance and a significant cytotoxic effect in AVA-resistant tumors when GLSi was administered in combination with bevacizumab. In vivo, GLSi led to a reduction in tumor growth as monotherapy and when combined with AVA. Furthermore, GLSi initiated after the emergence of resistance to AVA therapy resulted in a decreased metabolic conversion of pyruvate to lactate as assessed by hyperpolarized magnetic resonance spectroscopy and demonstrated robust antitumor effects with a survival advantage. Given the increasing population of patients receiving AVA therapy, these findings justify further development of GLSi in AVA resistance.
- Published
- 2023
- Full Text
- View/download PDF
5. Corrigendum to 'PTGER3 induces ovary tumorigenesis and confers resistance to cisplatin therapy through up-regulation Ras-MAPK/Erk-ETS1-ELK1/CFTR1 axis' [EBioMedicine 40 (2019) 290-304]
- Author
-
Cristian Rodriguez-Aguayo, Emine Bayraktar, Cristina Ivan, Burcu Aslan, Junhua Mai, Guangan He, Lingegowda S. Mangala, Dahai Jiang, Archana S. Nagaraja, Bulent Ozpolat, Arturo Chavez-Reyes, Mauro Ferrari, Rahul Mitra, Zahid H. Siddik, Haifa Shen, Xianbin Yang, Anil K. Sood, and Gabriel Lopez-Berestein
- Subjects
Medicine ,Medicine (General) ,R5-920 - Published
- 2022
- Full Text
- View/download PDF
6. Phase 2 of extracellular RNA communication consortium charts next-generation approaches for extracellular RNA research
- Author
-
Bogdan Mateescu, Jennifer C. Jones, Roger P. Alexander, Eric Alsop, Ji Yeong An, Mohammad Asghari, Alex Boomgarden, Laura Bouchareychas, Alfonso Cayota, Hsueh-Chia Chang, Al Charest, Daniel T. Chiu, Robert J. Coffey, Saumya Das, Peter De Hoff, Andrew deMello, Crislyn D’Souza-Schorey, David Elashoff, Kiarash R. Eliato, Jeffrey L. Franklin, David J. Galas, Mark B. Gerstein, Ionita H. Ghiran, David B. Go, Stephen Gould, Tristan R. Grogan, James N. Higginbotham, Florian Hladik, Tony Jun Huang, Xiaoye Huo, Elizabeth Hutchins, Dennis K. Jeppesen, Tijana Jovanovic-Talisman, Betty Y.S. Kim, Sung Kim, Kyoung-Mee Kim, Yong Kim, Robert R. Kitchen, Vaughan Knouse, Emily L. LaPlante, Carlito B. Lebrilla, L. James Lee, Kathleen M. Lennon, Guoping Li, Feng Li, Tieyi Li, Tao Liu, Zirui Liu, Adam L. Maddox, Kyle McCarthy, Bessie Meechoovet, Nalin Maniya, Yingchao Meng, Aleksandar Milosavljevic, Byoung-Hoon Min, Amber Morey, Martin Ng, John Nolan, Getulio P. De Oliveira Junior, Michael E. Paulaitis, Tuan Anh Phu, Robert L. Raffai, Eduardo Reátegui, Matthew E. Roth, David A. Routenberg, Joel Rozowsky, Joseph Rufo, Satyajyoti Senapati, Sigal Shachar, Himani Sharma, Anil K. Sood, Stavros Stavrakis, Alessandra Stürchler, Muneesh Tewari, Juan P. Tosar, Alexander K. Tucker-Schwartz, Andrey Turchinovich, Nedyalka Valkov, Kendall Van Keuren-Jensen, Kasey C. Vickers, Lucia Vojtech, Wyatt N. Vreeland, Ceming Wang, Kai Wang, ZeYu Wang, Joshua A. Welsh, Kenneth W. Witwer, David T.W. Wong, Jianping Xia, Ya-Hong Xie, Kaichun Yang, Mikołaj P. Zaborowski, Chenguang Zhang, Qin Zhang, Angela M. Zivkovic, and Louise C. Laurent
- Subjects
Biological sciences ,Biochemistry ,Molecular biology ,Cell biology ,Science - Abstract
Summary: The extracellular RNA communication consortium (ERCC) is an NIH-funded program aiming to promote the development of new technologies, resources, and knowledge about exRNAs and their carriers. After Phase 1 (2013–2018), Phase 2 of the program (ERCC2, 2019–2023) aims to fill critical gaps in knowledge and technology to enable rigorous and reproducible methods for separation and characterization of both bulk populations of exRNA carriers and single EVs. ERCC2 investigators are also developing new bioinformatic pipelines to promote data integration through the exRNA atlas database. ERCC2 has established several Working Groups (Resource Sharing, Reagent Development, Data Analysis and Coordination, Technology Development, nomenclature, and Scientific Outreach) to promote collaboration between ERCC2 members and the broader scientific community. We expect that ERCC2’s current and future achievements will significantly improve our understanding of exRNA biology and the development of accurate and efficient exRNA-based diagnostic, prognostic, and theranostic biomarker assays.
- Published
- 2022
- Full Text
- View/download PDF
7. Spatially resolved transcriptomics of high-grade serous ovarian carcinoma
- Author
-
Elaine Stur, Sara Corvigno, Mingchu Xu, Ken Chen, Yukun Tan, Sanghoon Lee, Jinsong Liu, Emily Ricco, Daniel Kraushaar, Patricia Castro, Jianhua Zhang, and Anil K. Sood
- Subjects
Oncology ,Pathology ,Omics ,Science - Abstract
Summary: Bulk and single-cell RNA sequencing do not provide full characterization of tissue spatial diversity in cancer samples, and currently available in situ techniques (multiplex immunohistochemistry and imaging mass cytometry) allow for only limited analysis of a small number of targets. The current study represents the first comprehensive approach to spatial transcriptomics of high-grade serous ovarian carcinoma using intact tumor tissue. We selected a small cohort of patients with highly annotated high-grade serous ovarian carcinoma, categorized them by response to neoadjuvant chemotherapy (poor or excellent), and analyzed pre-treatment tumor tissue specimens. Our study uncovered extensive differences in tumor composition between the poor responders and excellent responders to chemotherapy, related to cell cluster organization and localization. This in-depth characterization of high-grade serous ovarian carcinoma tumor tissue from poor and excellent responders showed that spatial interactions between cell clusters may influence chemo-responsiveness more than cluster composition alone.
- Published
- 2022
- Full Text
- View/download PDF
8. Endothelial p130cas confers resistance to anti-angiogenesis therapy
- Author
-
Yunfei Wen, Anca Chelariu-Raicu, Sujanitha Umamaheswaran, Alpa M. Nick, Elaine Stur, Pahul Hanjra, Dahai Jiang, Nicholas B. Jennings, Xiuhui Chen, Sara Corvigno, Deanna Glassman, Gabriel Lopez-Berestein, Jinsong Liu, Mien-Chie Hung, and Anil K. Sood
- Subjects
p130cas ,VEGFR2 ,autophagy ,TNKS1BP1 ,adaptive resistance ,anti-angiogenic therapy ,Biology (General) ,QH301-705.5 - Abstract
Summary: Anti-angiogenic therapies, such as anti-VEGF antibodies (AVAs), have shown promise in clinical settings. However, adaptive resistance to such therapies occurs frequently. We use orthotopic ovarian cancer models with AVA-adaptive resistance to investigate the underlying mechanisms. Genomic profiling of AVA-resistant tumors guides us to endothelial p130cas. We find that bevacizumab induces cleavage of VEGFR2 in endothelial cells by caspase-10 and that VEGFR2 fragments internalize into the nucleus and autophagosomes. Nuclear VEGFR2 and p130cas fragments, together with TNKS1BP1 (tankyrase-1-binding protein), initiate endothelial cell death. Blockade of autophagy in AVA-resistant endothelial cells retains VEGFR2 at the membrane with bevacizumab treatment. Targeting host p130cas with RGD (Arg-Gly-Asp)-tagged nanoparticles or genomic ablation of vascular p130cas in p130casflox/floxTie2Cre mice significantly extends the survival of mice with AVA-resistant ovarian tumors. Higher vascular p130cas is associated with shorter survival of individuals with ovarian cancer. Our findings identify opportunities for new strategies to overcome adaptive resistance to AVA therapy.
- Published
- 2022
- Full Text
- View/download PDF
9. RNA-binding protein FXR1 drives cMYC translation by recruiting eIF4F complex to the translation start site
- Author
-
Jasmine George, Yongsheng Li, Ishaque P. Kadamberi, Deepak Parashar, Shirng-Wern Tsaih, Prachi Gupta, Anjali Geethadevi, Changliang Chen, Chandrima Ghosh, Yunguang Sun, Sonam Mittal, Ramani Ramchandran, Hallgeir Rui, Gabriel Lopez-Berestein, Cristian Rodriguez-Aguayo, Gustavo Leone, Janet S. Rader, Anil K. Sood, Madhusudan Dey, Sunila Pradeep, and Pradeep Chaluvally-Raghavan
- Subjects
FXR1 ,ovarian cancer ,cMYC ,SUnSET ,eIFs ,ARE ,Biology (General) ,QH301-705.5 - Abstract
Summary: Fragile X-related protein-1 (FXR1) gene is highly amplified in patients with ovarian cancer, and this amplification is associated with increased expression of both FXR1 mRNA and protein. FXR1 expression directly associates with the survival and proliferation of cancer cells. Surface sensing of translation (SUnSET) assay demonstrates that FXR1 enhances the overall translation in cancer cells. Reverse-phase protein array (RPPA) reveals that cMYC is the key target of FXR1. Mechanistically, FXR1 binds to the AU-rich elements (ARE) present within the 3′ untranslated region (3′UTR) of cMYC and stabilizes its expression. In addition, the RGG domain in FXR1 interacts with eIF4A1 and eIF4E proteins. These two interactions of FXR1 result in the circularization of cMYC mRNA and facilitate the recruitment of eukaryotic translation initiation factors to the translation start site. In brief, we uncover a mechanism by which FXR1 promotes cMYC levels in cancer cells.
- Published
- 2021
- Full Text
- View/download PDF
10. Circular RNAs in Cancer
- Author
-
Duc-Hiep Bach, Sang Kook Lee, and Anil K. Sood
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
Circular RNAs (circRNAs) are a class of single-stranded closed RNA molecules that are formed by precursor mRNA back-splicing or skipping events of thousands of genes in eukaryotes as covalently closed continuous loops. High-throughput sequencing and bioinformatics approaches have uncovered the broad expression of circRNAs across species. Their high stability, abundance, and evolutionary conservation among species points to their distinct properties and diverse cellular functions as efficient microRNAs and protein sponges; they also play important roles in modulating transcription and splicing. Additionally, most circRNAs are aberrantly expressed in pathological conditions and in a tissue-specific manner such as development and progression of cancer. Herein, we highlight the characteristics, functions, and mechanisms of action of circRNAs in cancer; we also provide an overview of recent progress in the circRNA field and future application of circRNAs as cancer biomarkers and novel therapeutic targets. Keywords: circular RNAs, cancer biomarkers, hallmarks of cancer, exosomes, oncogenic function, platelets, malignant tumors, hematological malignancies, biogenesis, biological functions
- Published
- 2019
- Full Text
- View/download PDF
11. Pan-cancer genomic analysis links 3’UTR DNA methylation with increased gene expression in T cellsResearch in context
- Author
-
Michael H. McGuire, Shelley M. Herbrich, Santosh K. Dasari, Sherry Y. Wu, Ying Wang, Rajesha Rupaimoole, Gabriel Lopez-Berestein, Keith A. Baggerly, and Anil K. Sood
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Investigations into the function of non-promoter DNA methylation have yielded new insights into the epigenetic regulation of gene expression. However, integrated genome-wide non-promoter DNA methylation and gene expression analyses across a wide number of tumour types and corresponding normal tissues have not been performed. Methods: To investigate the impact of non-promoter DNA methylation on cancer pathogenesis, we performed a large-scale analysis of gene expression and DNA methylation profiles, finding enrichment in the 3’UTR DNA methylation positively correlated with gene expression. Filtering for genes in which 3’UTR DNA methylation strongly correlated with gene expression yielded a list of genes enriched for functions involving T cell activation. Findings: The important immune checkpoint gene Havcr2 showed a substantial increase in 3’UTR DNA methylation upon T cell activation and subsequent upregulation of gene expression in mice. Furthermore, this increase in Havcr2 gene expression was abrogated by treatment with decitabine. Interpretation: These findings indicate that the 3’UTR is a functionally relevant DNA methylation site. Additionally, we show a potential novel mechanism of HAVCR2 regulation in T cells, providing new insights for modulating immune checkpoint blockade. Keywords: 3’UTR, Epigenetics, Immune checkpoint, Methylation
- Published
- 2019
- Full Text
- View/download PDF
12. PTGER3 induces ovary tumorigenesis and confers resistance to cisplatin therapy through up-regulation Ras-MAPK/Erk-ETS1-ELK1/CFTR1 axisResearch in context
- Author
-
Cristian Rodriguez-Aguayo, Emine Bayraktar, Cristina Ivan, Burcu Aslan, Junhua Mai, Guangan He, Lingegowda S. Mangala, Dahai Jiang, Archana S. Nagaraja, Bulent Ozpolat, Arturo Chavez-Reyes, Mauro Ferrari, Rahul Mitra, Zahid H. Siddik, Haifa Shen, Xianbin Yang, Anil K. Sood, and Gabriel Lopez-Berestein
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Inflammatory mediator prostaglandin E2–prostaglandin E2 receptor EP3 (PTGER3) signaling is critical for tumor-associated angiogenesis, tumor growth, and chemoresistance. However, the mechanism underlying these effects in ovarian cancer is not known. Methods: An association between higher tumoral expression of PTGER3 and shorter patient survival in the ovarian cancer dataset of The Cancer Genome Atlas prompted investigation of the antitumor effects of PTGER3 downmodulation. PTGER3 mRNA and protein levels were higher in cisplatin-resistant ovarian cancer cells than in their cisplatin-sensitive counterparts. Findings: Silencing of PTGER3 via siRNA in cancer cells was associated with decreased cell growth and less invasiveness, as well as cell-cycle arrest and increased apoptosis, mediated through the Ras-MAPK/Erk-ETS1-ELK1/CFTR1 axis. Furthermore, sustained PTGER3 silencing with multistage vector and liposomal 2’-F-phosphorodithioate-siRNA–mediated silencing of PTGER3 combined with cisplatin resulted in robust antitumor effects in cisplatin-resistant ovarian cancer models. Interpretation: These findings identify PTGER3 as a potential therapeutic target in chemoresistant ovarian cancers expressing high levels of this oncogenic protein. Fund: National Institutes of Health/National Cancer Institute, USA. Keywords: PTGER3, Ovarian cancer, RNA interference, Chemically modified siRNA, Cisplatin resistance, ETS1, ELK1, CFTR
- Published
- 2019
- Full Text
- View/download PDF
13. Extensive three-dimensional intratumor proteomic heterogeneity revealed by multiregion sampling in high-grade serous ovarian tumor specimens
- Author
-
Allison L. Hunt, Nicholas W. Bateman, Waleed Barakat, Sasha Makohon-Moore, Brian L. Hood, Kelly A. Conrads, Ming Zhou, Valerie Calvert, Mariaelena Pierobon, Jeremy Loffredo, Tracy J. Litzi, Julie Oliver, Dave Mitchell, Glenn Gist, Christine Rojas, Brian Blanton, Emma L. Robinson, Kunle Odunsi, Anil K. Sood, Yovanni Casablanca, Kathleen M. Darcy, Craig D. Shriver, Emanuel F. Petricoin, Uma N.M. Rao, G. Larry Maxwell, and Thomas P. Conrads
- Subjects
Oncology ,Cancer systems biology ,Proteomics ,Transcriptomics ,Science - Abstract
Summary: Enriched tumor epithelium, tumor-associated stroma, and whole tissue were collected by laser microdissection from thin sections across spatially separated levels of ten high-grade serous ovarian carcinomas (HGSOCs) and analyzed by mass spectrometry, reverse phase protein arrays, and RNA sequencing. Unsupervised analyses of protein abundance data revealed independent clustering of an enriched stroma and enriched tumor epithelium, with whole tumor tissue clustering driven by overall tumor “purity.” Comparing these data to previously defined prognostic HGSOC molecular subtypes revealed protein and transcript expression from tumor epithelium correlated with the differentiated subtype, whereas stromal proteins (and transcripts) correlated with the mesenchymal subtype. Protein and transcript abundance in the tumor epithelium and stroma exhibited decreased correlation in samples collected just hundreds of microns apart. These data reveal substantial tumor microenvironment protein heterogeneity that directly bears on prognostic signatures, biomarker discovery, and cancer pathophysiology and underscore the need to enrich cellular subpopulations for expression profiling.
- Published
- 2021
- Full Text
- View/download PDF
14. Distinct T cell receptor repertoire diversity of clinically defined high-grade serous ovarian cancer treatment subgroups
- Author
-
Sanghoon Lee, Li Zhao, Latasha D. Little, Shannon N. Westin, Amir A. Jazarei, Nicole D. Fleming, Jianhua Zhang, P. Andrew Futreal, and Anil K. Sood
- Subjects
Biological Sciences ,Immunology ,Cancer ,Science - Abstract
Summary: In patients with high-grade serous ovarian cancer (HGSC), it is unclear which genomic features are related to complete gross resection (R0), which is typically associated with better clinical outcomes, or response to neoadjuvant chemotherapy (NACT). In this study, we evaluated T cell receptor (TCR) repertoire diversity in primary and metastatic tumor samples (n = 90) from clinically well-annotated patients with HGSC who achieved R0 or received NACT with excellent or poor response based on a laparoscopic triage algorithm. TCR sequencing followed by an integrative analysis with comprehensive multi-omics data identified higher TCR diversity (e.g., higher number of unique productive sequences and less clonal relatedness) in the R0 than NACT groups. We found enrichment of specific TCRβ genes usage, distinct mutual exclusiveness and co-occurrence pattern of TCRβ genes among the groups. We also found significantly positive correlations between clonal relatedness and neoantigens, copy number variations, and mutation load in the groups.
- Published
- 2021
- Full Text
- View/download PDF
15. Gain-of-function p53 protein transferred via small extracellular vesicles promotes conversion of fibroblasts to a cancer-associated phenotype
- Author
-
Shaolin Ma, Michael H. McGuire, Lingegowda S. Mangala, Sanghoon Lee, Elaine Stur, Wen Hu, Emine Bayraktar, Alejandro Villar-Prados, Cristina Ivan, Sherry Y. Wu, Akira Yokoi, Santosh K. Dasari, Nicholas B. Jennings, Jinsong Liu, Gabriel Lopez-Berestein, Prahlad Ram, and Anil K. Sood
- Subjects
small EVs ,p53 ,CAFs ,HSP90 ,Nrf2 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Tumor and stromal interactions consist of reciprocal signaling through cytokines, growth factors, direct cell-cell interactions, and extracellular vesicles (EVs). Small EVs (≤200 nm) have been considered critical messengers of cellular communication during tumor development. Here, we demonstrate that gain-of-function (GOF) p53 protein can be packaged into small EVs and transferred to fibroblasts. GOF p53 protein is selectively bound by heat shock protein 90 (HSP90), a chaperone protein, and packaged into small EVs. Inhibition of HSP90 activity blocks packaging of GOF, but not wild-type, p53 in small EVs. GOF p53-containing small EVs result in their conversion to cancer-associated fibroblasts. In vivo studies reveal that GOF p53-containing small EVs can enhance tumor growth and promote fibroblast transformation into a cancer-associated phenotype. These findings provide a better understanding of the complex interactions between cancer and stromal cells and may have therapeutic implications.
- Published
- 2021
- Full Text
- View/download PDF
16. Role of tissue-factor bearing extracellular vesicles released from ovarian cancer cells in platelet aggregation in vitro and venous thrombosis in mice
- Author
-
Tomoyuki Sasano, Min Soon Cho, Cristian Rodriguez-Aguayo, Emine Bayraktar, Mana Taki, Vahid Afshar-Kharghan, and Anil K. Sood
- Subjects
Extracellular vesicles ,Cell-derived microparticles ,Tissue factor ,Ovarian neoplasms ,Thrombosis ,Platelet aggregation ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Objective: Venous thromboembolism (VTE) contributes to morbidity and mortality in women with ovarian cancer. Underlying mechanisms of venous thrombosis in ovarian cancer are not well-understood. The aim of this study was to identify the potential role of tissue factor (TF)-bearing extracellular vesicles (EVs) originated from cancer cells in venous thrombosis in ovarian cancer models. Methods: We examined the expression of TF on EVs generated by ovarian cancer cells and the effect of TF-positive EVs on platelet aggregation. Furthermore, we performed TF-knockdown or induced TF-overexpression in ovarian cancer cell lines and examined the effects of EVs obtained from these cells on platelet aggregation. We examined the effect of TF-bearing EVs originated from ovarian cancer cells on venous thrombosis in a mouse model of inferior vena cava (IVC) stenosis. Results: TF was expressed in several ovarian cancer cell lines. EVs derived from ovarian cancer cell lines expressing TF promoted platelet aggregation. TF-knockdown in TF-high CAOV3 and OVCAR8 ovarian cancer cells delayed platelet aggregation induced by their EVs in vitro. Conversely, TF overexpression in TF-low A2780 and HeyA8 cells shortened platelet aggregation time induced by their EVs. EVs from TF-overexpressing A2780 cells enhanced thrombosis formation in the IVC stenosis model and resulted in a larger clot burden as compared to EVs from A2780 control cells. Conclusions: TF expression in ovarian cancer cell-derived EVs promoted platelet aggregation and thrombosis in preclinical models. These findings may have implications for reducing VTE rates in women with ovarian cancer.
- Published
- 2021
- Full Text
- View/download PDF
17. Sustained Adrenergic Activation of YAP1 Induces Anoikis Resistance in Cervical Cancer Cells
- Author
-
Yang Li, Shanshan Yang, Nouara C. Sadaoui, Wei Hu, Santosh K. Dasari, Lingegowda S. Mangala, Yunjie Sun, Shuangtao Zhao, Linghua Wang, Yuan Liu, Lois M. Ramondetta, Ke Li, Chong Lu, Yu Kang, Steve W. Cole, Susan K. Lutgendorf, and Anil K. Sood
- Subjects
Biological Sciences ,Cancer ,Science - Abstract
Summary: Chronic stress-related hormones modulate tumor pathogenesis at multiple levels; however, the molecular pathways involved in stress and cervical cancer progression are not well understood. We established a preclinical orthotopic mouse model of cervical cancer and used the model to show that daily restraint stress increased tumor growth and metastatic tumor burden. Exposure to norepinephrine significantly protected cervical cancer cells from anoikis. We demonstrated that YAP1 was dephosphorylated and translocated from the cytoplasm to the nucleus by norepinephrine, a process initiated by ADRB2/cAMP/protein kinase A activation. Furthermore, anoikis resistance and YAP1 activation induced by norepinephrine could be rescued by a broad β-adrenergic receptor antagonist, propranolol. Collectively, our results provide a pivotal molecular pathway for disrupting pro-tumor neuroendocrine signaling in cervical cancer.
- Published
- 2020
- Full Text
- View/download PDF
18. Corrigendum to ‘Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer’ [EBioMedicine 38 (2018) 100–112]
- Author
-
Pinar Kanlikilicer, Recep Bayraktar, Merve Denizli, Mohammed H. Rashed, Cristina Ivan, Burcu Aslan, Rahul Mitra, Kubra Karagoz, Emine Bayraktar, Xinna Zhang, Cristian Rodriguez-Aguayo, Amr Ahmed El-Arabey, Nermin Kahraman, Seyda Baydogan, Ozgur Ozkayar, Michael L. Gatza, Bulent Ozpolat, George A. Calin, Anil K. Sood, and Gabriel Lopez-Berestein
- Subjects
Medicine ,Medicine (General) ,R5-920 - Published
- 2020
- Full Text
- View/download PDF
19. Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancerResearch in context
- Author
-
Pinar Kanlikilicer, Recep Bayraktar, Merve Denizli, Mohammed H. Rashed, Cristina Ivan, Burcu Aslan, Rahul Mitra, Kubra Karagoz, Emine Bayraktar, Xinna Zhang, Cristian Rodriguez-Aguayo, Amr Ahmed El-Arabey, Nermin Kahraman, Seyda Baydogan, Ozgur Ozkayar, Michael L. Gatza, Bulent Ozpolat, George A. Calin, Anil K. Sood, and Gabriel Lopez-Berestein
- Subjects
Medicine ,Medicine (General) ,R5-920 - Abstract
Background: Circulating miRNAs are known to play important roles in intercellular communication. However, the effects of exosomal miRNAs on cells are not fully understood. Methods: To investigate the role of exosomal miR-1246 in ovarian cancer (OC) microenvironment, we performed RPPA as well as many other in vitro functional assays in ovarian cancer cells (sensitive; HeyA8, Skov3ip1, A2780 and chemoresistant; HeyA8-MDR, Skov3-TR, A2780-CP20). Therapeutic effect of miR-1246 inhibitor treatment was tested in OC animal model. We showed the effect of OC exosomal miR-1246 uptake on macrophages by co-culture experiments. Findings: Substantial expression of oncogenic miR-1246 OC exosomes was found. We showed that Cav1 gene, which is the direct target of miR-1246, is involved in the process of exosomal transfer. A significantly worse overall prognosis were found for OC patients with high miR-1246 and low Cav1 expression based on TCGA data. miR-1246 expression were significantly higher in paclitaxel-resistant OC exosomes than in their sensitive counterparts. Overexpression of Cav1 and anti-miR-1246 treatment significantly sensitized OC cells to paclitaxel. We showed that Cav1 and multi drug resistance (MDR) gene is involved in the process of exosomal transfer. Our proteomic approach also revealed that miR-1246 inhibits Cav1 and acts through PDGFβ receptor at the recipient cells to inhibit cell proliferation. miR-1246 inhibitor treatment in combination with chemotherapy led to reduced tumor burden in vivo. Finally, we demonstrated that when OC cells are co-cultured with macrophages, they are capable of transferring their oncogenic miR-1246 to M2-type macrophages, but not M0-type macrophages. Interpretation: Our results suggest that cancer exosomes may contribute to oncogenesis by manipulating neighboring infiltrating immune cells. This study provide a new mechanistic therapeutic approach to overcome chemoresistance and tumor progression through exosomal miR-1246 in OC patients. Keywords: Exosome, oncomiR, miR-1246, Ovarian cancer, Cav1, P-gp
- Published
- 2018
- Full Text
- View/download PDF
20. ZRANB1 Is an EZH2 Deubiquitinase and a Potential Therapeutic Target in Breast Cancer
- Author
-
Peijing Zhang, Zhenna Xiao, Shouyu Wang, Mutian Zhang, Yongkun Wei, Qinglei Hang, Jongchan Kim, Fan Yao, Cristian Rodriguez-Aguayo, Baochau N. Ton, Minjung Lee, Yumeng Wang, Zhicheng Zhou, Liyong Zeng, Xiaoyu Hu, Sarah E. Lawhon, Ashley N. Siverly, Xiaohua Su, Jia Li, Xiaoping Xie, Xuhong Cheng, Liang-Chiu Liu, Hui-Wen Chang, Shu-Fen Chiang, Gabriel Lopez-Berestein, Anil K. Sood, Junjie Chen, M. James You, Shao-Cong Sun, Han Liang, Yun Huang, Xianbin Yang, Deqiang Sun, Yutong Sun, Mien-Chie Hung, and Li Ma
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Although EZH2 enzymatic inhibitors have shown antitumor effects in EZH2-mutated lymphoma and ARID1A-mutated ovarian cancer, many cancers do not respond because EZH2 can promote cancer independently of its histone methyltransferase activity. Here we identify ZRANB1 as the EZH2 deubiquitinase. ZRANB1 binds, deubiquitinates, and stabilizes EZH2. Depletion of ZRANB1 in breast cancer cells results in EZH2 destabilization and growth inhibition. Systemic delivery of ZRANB1 small interfering RNA (siRNA) leads to marked antitumor and antimetastatic effects in preclinical models of triple-negative breast cancer (TNBC). Intriguingly, a small-molecule inhibitor of ZRANB1 destabilizes EZH2 and inhibits the viability of TNBC cells. In patients with breast cancer, ZRANB1 levels correlate with EZH2 levels and poor survival. These findings suggest the therapeutic potential for targeting the EZH2 deubiquitinase ZRANB1. : Many cancer cells are sensitive to depletion of EZH2 but resistant to EZH2 inhibitors, due to EZH2’s enzyme-independent cancer-promoting function. Zhang et al. identify ZRANB1 as an EZH2 deubiquitinase and a potential anticancer target. Keywords: ZRANB1, EZH2, deubiquitinase, breast cancer
- Published
- 2018
- Full Text
- View/download PDF
21. HN1L Promotes Triple-Negative Breast Cancer Stem Cells through LEPR-STAT3 Pathway
- Author
-
Yi Liu, Dong Soon Choi, Jianting Sheng, Joe E. Ensor, Diana Hwang Liang, Cristian Rodriguez-Aguayo, Amanda Polley, Steve Benz, Olivier Elemento, Akanksha Verma, Yang Cong, Helen Wong, Wei Qian, Zheng Li, Sergio Granados-Principal, Gabriel Lopez-Berestein, Melissa D. Landis, Roberto R. Rosato, Bhuvanesh Dave, Stephen Wong, Dario Marchetti, Anil K. Sood, and Jenny C. Chang
- Subjects
HN1L ,LEPR ,STAT3 ,TNBC ,cancer stem cells ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Here, we show that HEMATOLOGICAL AND NEUROLOGICAL EXPRESSED 1-LIKE (HN1L) is a targetable breast cancer stem cell (BCSC) gene that is altered in 25% of whole breast cancer and significantly correlated with shorter overall or relapse-free survival in triple-negative breast cancer (TNBC) patients. HN1L silencing reduced the population of BCSCs, inhibited tumor initiation, resensitized chemoresistant tumors to docetaxel, and hindered cancer progression in multiple TNBC cell line-derived xenografts. Additionally, gene signatures associated with HN1L correlated with shorter disease-free survival of TNBC patients. We defined HN1L as a BCSC transcription regulator for genes involved in the LEPR-STAT3 signaling axis as HN1L binds to a putative consensus upstream sequence of STAT3, LEPTIN RECEPTOR, and MIR-150. Our data reveal that BCSCs in TNBC depend on the transcription regulator HN1L for the sustained activation of the LEPR-STAT3 pathway, which makes it a potentially important target for both prognosis and BCSC therapy.
- Published
- 2018
- Full Text
- View/download PDF
22. Therapeutic Targeting of AXL Receptor Tyrosine Kinase Inhibits Tumor Growth and Intraperitoneal Metastasis in Ovarian Cancer Models
- Author
-
Pinar Kanlikilicer, Bulent Ozpolat, Burcu Aslan, Recep Bayraktar, Nilgun Gurbuz, Cristian Rodriguez-Aguayo, Emine Bayraktar, Merve Denizli, Vianey Gonzalez-Villasana, Cristina Ivan, Ganesh L.R. Lokesh, Paola Amero, Silvia Catuogno, Monika Haemmerle, Sherry Yen-Yao Wu, Rahul Mitra, David G. Gorenstein, David E. Volk, Vittorio de Franciscis, Anil K. Sood, and Gabriel Lopez-Berestein
- Subjects
AXL ,aptamer ,metastasis ,ovarian cancer ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Despite substantial improvements in the treatment strategies, ovarian cancer is still the most lethal gynecological malignancy. Identification of drug treatable therapeutic targets and their safe and effective targeting is critical to improve patient survival in ovarian cancer. AXL receptor tyrosine kinase (RTK) has been proposed to be an important therapeutic target for metastatic and advanced-stage human ovarian cancer. We found that AXL-RTK expression is associated with significantly shorter patient survival based on the The Cancer Genome Atlas patient database. To target AXL-RTK, we developed a chemically modified serum nuclease-stable AXL aptamer (AXL-APTAMER), and we evaluated its in vitro and in vivo antitumor activity using in vitro assays as well as two intraperitoneal animal models. AXL-aptamer treatment inhibited the phosphorylation and the activity of AXL, impaired the migration and invasion ability of ovarian cancer cells, and led to the inhibition of tumor growth and number of intraperitoneal metastatic nodules, which was associated with the inhibition of AXL activity and angiogenesis in tumors. When combined with paclitaxel, in vivo systemic (intravenous [i.v.]) administration of AXL-aptamer treatment markedly enhanced the antitumor efficacy of paclitaxel in mice. Taken together, our data indicate that AXL-aptamers successfully target in vivo AXL-RTK and inhibit its AXL activity and tumor growth and progression, representing a promising strategy for the treatment of ovarian cancer.
- Published
- 2017
- Full Text
- View/download PDF
23. Differential Effects of EGFL6 on Tumor versus Wound Angiogenesis
- Author
-
Kyunghee Noh, Lingegowda S. Mangala, Hee-Dong Han, Ningyan Zhang, Sunila Pradeep, Sherry Y. Wu, Shaolin Ma, Edna Mora, Rajesha Rupaimoole, Dahai Jiang, Yunfei Wen, Mian M.K. Shahzad, Yasmin Lyons, MinSoon Cho, Wei Hu, Archana S. Nagaraja, Monika Haemmerle, Celia S.L. Mak, Xiuhui Chen, Kshipra M. Gharpure, Hui Deng, Wei Xiong, Charles V. Kingsley, Jinsong Liu, Nicholas Jennings, Michael J. Birrer, Richard R. Bouchard, Gabriel Lopez-Berestein, Robert L. Coleman, Zhiqiang An, and Anil K. Sood
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: 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. : Noh et al. identify EGFL6 as an angiogenic target that is selectively present in tumor endothelial cells in a hypoxic tumor microenvironment. EGFL6 blockade exerts robust anti-angiogenic and anti-tumor effects without affecting wound healing. These findings suggest an important approach for effectively targeting tumor angiogenesis. Keywords: tumor endothelial cells, ovarian cancer, chitosan nanoparticles, tumor vasculature, wound healing
- Published
- 2017
- Full Text
- View/download PDF
24. miRNA551b-3p Activates an Oncostatin Signaling Module for the Progression of Triple-Negative Breast Cancer
- Author
-
Deepak Parashar, Anjali Geethadevi, Miriam Ragle Aure, Jyotsna Mishra, Jasmine George, Changliang Chen, Manoj K. Mishra, Andliena Tahiri, Wei Zhao, Bindu Nair, Yiling Lu, Lingegowda S. Mangala, Cristian Rodriguez-Aguayo, Gabriel Lopez-Berestein, Amadou K.S. Camara, Mingyu Liang, Janet S. Rader, Ramani Ramchandran, Ming You, Anil K. Sood, Vessela N. Kristensen, Gordon B. Mills, Sunila Pradeep, and Pradeep Chaluvally-Raghavan
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Genomic amplification of 3q26.2 locus leads to the increased expression of microRNA 551b-3p (miR551b-3p) in triple-negative breast cancer (TNBC). Our results demonstrate that miR551b-3p translocates to the nucleus with the aid of importin-8 (IPO8) and activates STAT3 transcription. As a consequence, miR551b upregulates the expression of oncostatin M receptor (OSMR) and interleukin-31 receptor-α (IL-31RA) as well as their ligands OSM and IL-31 through STAT3 transcription. We defined this set of genes induced by miR551b-3p as the “oncostatin signaling module,” which provides oncogenic addictions in cancer cells. Notably, OSM is highly expressed in TNBC, and the elevated expression of OSM associates with poor outcome in estrogen-receptor-negative breast cancer patients. Conversely, targeting miR551b with anti-miR551b-3p reduced the expression of the OSM signaling module and reduced tumor growth, as well as migration and invasion of breast cancer cells. : Parashar et al. demonstrate that the mature microRNA-551b-3p translocates to the nucleus for transcriptional activation of STAT3 gene. As a consequence, miR551b activates an autocrine signaling loop through the upregulation of oncostatin family genes, including oncostatin, and its receptors for the growth and metastasis of triple-negative breast cancer. Keywords: miR551b, oncostatin, STAT3, OSMR, miRNA-therapy, IPO8, RNAi
- Published
- 2019
- Full Text
- View/download PDF
25. Role of CTGF in Sensitivity to Hyperthermia in Ovarian and Uterine Cancers
- Author
-
Hiroto Hatakeyama, Sherry Y. Wu, Yasmin A. Lyons, Sunila Pradeep, Wanqin Wang, Qian Huang, Karem A. Court, Tao Liu, Song Nie, Cristian Rodriguez-Aguayo, Fangrong Shen, Yan Huang, Takeshi Hisamatsu, Takashi Mitamura, Nicholas Jennings, Jeajun Shim, Piotr L. Dorniak, Lingegowda S. Mangala, Marco Petrillo, Vladislav A. Petyuk, Athena A. Schepmoes, Anil K. Shukla, Madeline Torres-Lugo, Ju-Seog Lee, Karin D. Rodland, Anna Fagotti, Gabriel Lopez-Berestein, Chun Li, and Anil K. Sood
- Subjects
hyperthermia ,ovarian cancer ,CTGF ,DOPC-liposome ,thermosensitivity ,copper sulfide nanoparticle ,Biology (General) ,QH301-705.5 - Abstract
Even though hyperthermia is a promising treatment for cancer, the relationship between specific temperatures and clinical benefits and predictors of sensitivity of cancer to hyperthermia is poorly understood. Ovarian and uterine tumors have diverse hyperthermia sensitivities. Integrative analyses of the specific gene signatures and the differences in response to hyperthermia between hyperthermia-sensitive and -resistant cancer cells identified CTGF as a key regulator of sensitivity. CTGF silencing sensitized resistant cells to hyperthermia. CTGF small interfering RNA (siRNA) treatment also sensitized resistant cancers to localized hyperthermia induced by copper sulfide nanoparticles and near-infrared laser in orthotopic ovarian cancer models. CTGF silencing aggravated energy stress induced by hyperthermia and enhanced apoptosis of hyperthermia-resistant cancers.
- Published
- 2016
- Full Text
- View/download PDF
26. microRNA Therapeutics in Cancer — An Emerging Concept
- Author
-
Maitri Y. Shah, Alessandra Ferrajoli, Anil K Sood, Gabriel Lopez-Berestein, and George A. Calin
- Subjects
microRNA ,Non-coding RNA ,Cancer ,Therapeutics ,Clinical trials ,Medicine ,Medicine (General) ,R5-920 - Abstract
MicroRNAs (miRNAs) are an evolutionarily conserved class of small, regulatory non-coding RNAs that negatively regulate protein coding gene and other non-coding transcripts expression. miRNAs have been established as master regulators of cellular processes, and they play a vital role in tumor initiation, progression and metastasis. Further, widespread deregulation of microRNAs have been reported in several cancers, with several microRNAs playing oncogenic and tumor suppressive roles. Based on these, miRNAs have emerged as promising therapeutic tools for cancer management. In this review, we have focused on the roles of miRNAs in tumorigenesis, the miRNA-based therapeutic strategies currently being evaluated for use in cancer, and the advantages and current challenges to their use in the clinic.
- Published
- 2016
- Full Text
- View/download PDF
27. Direct Upregulation of STAT3 by MicroRNA-551b-3p Deregulates Growth and Metastasis of Ovarian Cancer
- Author
-
Pradeep Chaluvally-Raghavan, Kang Jin Jeong, Sunila Pradeep, Andreia Machado Silva, Shuangxing Yu, Wenbin Liu, Tyler Moss, Cristian Rodriguez-Aguayo, Dong Zhang, Prahlad Ram, Jinsong Liu, Yiling Lu, Gabriel Lopez-Berestein, George A. Calin, Anil K. Sood, and Gordon B. Mills
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: 3q26.2 amplification in high-grade serous ovarian cancer leads to increased expression of mature microRNA miR551b-3p, which is associated with poor clinical outcome. Importantly, miR551b-3p contributes to resistance to apoptosis and increased survival and proliferation of cancer cells in vitro and in vivo. miR551b-3p upregulates STAT3 protein levels, and STAT3 is required for the effects of miR551b-3p on cell proliferation. Rather than decreasing levels of target mRNA as expected, we demonstrate that miR551b-3p binds a complementary sequence on the STAT3 promoter, recruiting RNA polymerase II and the TWIST1 transcription factor to activate STAT3 transcription, and thus directly upregulates STAT3 expression. Furthermore, anti-miR551b reduced STAT3 expression in ovarian cancer cells in vitro and in vivo and reduced ovarian cancer growth in vivo. Together, our data demonstrate a role for miR551b-3p in transcriptional activation. Thus, miR551b-3p represents a promising candidate biomarker and therapeutic target in ovarian cancer. : Chaluvally-Raghavan et al. find that miR551b-3p affects transcription of STAT3. The authors find that miR551b-3p binds to the STAT3 promoter and facilitates RNA polymerase II and TWIST1 transcription factor recruitment, which in turn activates STAT3 transcription.
- Published
- 2016
- Full Text
- View/download PDF
28. Pan-Cancer Analysis of lncRNA Regulation Supports Their Targeting of Cancer Genes in Each Tumor Context
- Author
-
Hua-Sheng Chiu, Sonal Somvanshi, Ektaben Patel, Ting-Wen Chen, Vivek P. Singh, Barry Zorman, Sagar L. Patil, Yinghong Pan, Sujash S. Chatterjee, Anil K. Sood, Preethi H. Gunaratne, Pavel Sumazin, Samantha J. Caesar-Johnson, John A. Demchok, Ina Felau, Melpomeni Kasapi, Martin L. Ferguson, Carolyn M. Hutter, Heidi J. Sofia, Roy Tarnuzzer, Zhining Wang, Liming Yang, Jean C. Zenklusen, Jiashan (Julia) Zhang, Sudha Chudamani, Jia Liu, Laxmi Lolla, Rashi Naresh, Todd Pihl, Qiang Sun, Yunhu Wan, Ye Wu, Juok Cho, Timothy DeFreitas, Scott Frazer, Nils Gehlenborg, Gad Getz, David I. Heiman, Jaegil Kim, Michael S. Lawrence, Pei Lin, Sam Meier, Michael S. Noble, Gordon Saksena, Doug Voet, Hailei Zhang, Brady Bernard, Nyasha Chambwe, Varsha Dhankani, Theo Knijnenburg, Roger Kramer, Kalle Leinonen, Yuexin Liu, Michael Miller, Sheila Reynolds, Ilya Shmulevich, Vesteinn Thorsson, Wei Zhang, Rehan Akbani, Bradley M. Broom, Apurva M. Hegde, Zhenlin Ju, Rupa S. Kanchi, Anil Korkut, Jun Li, Han Liang, Shiyun Ling, Wenbin Liu, Yiling Lu, Gordon B. Mills, Kwok-Shing Ng, Arvind Rao, Michael Ryan, Jing Wang, John N. Weinstein, Jiexin Zhang, Adam Abeshouse, Joshua Armenia, Debyani Chakravarty, Walid K. Chatila, Ino de Bruijn, Jianjiong Gao, Benjamin E. Gross, Zachary J. Heins, Ritika Kundra, Konnor La, Marc Ladanyi, Augustin Luna, Moriah G. Nissan, Angelica Ochoa, Sarah M. Phillips, Ed Reznik, Francisco Sanchez-Vega, Chris Sander, Nikolaus Schultz, Robert Sheridan, S. Onur Sumer, Yichao Sun, Barry S. Taylor, Jioajiao Wang, Hongxin Zhang, Pavana Anur, Myron Peto, Paul Spellman, Christopher Benz, Joshua M. Stuart, Christopher K. Wong, Christina Yau, D. Neil Hayes, Joel S. Parker, Matthew D. Wilkerson, Adrian Ally, Miruna Balasundaram, Reanne Bowlby, Denise Brooks, Rebecca Carlsen, Eric Chuah, Noreen Dhalla, Robert Holt, Steven J.M. Jones, Katayoon Kasaian, Darlene Lee, Yussanne Ma, Marco A. Marra, Michael Mayo, Richard A. Moore, Andrew J. Mungall, Karen Mungall, A. Gordon Robertson, Sara Sadeghi, Jacqueline E. Schein, Payal Sipahimalani, Angela Tam, Nina Thiessen, Kane Tse, Tina Wong, Ashton C. Berger, Rameen Beroukhim, Andrew D. Cherniack, Carrie Cibulskis, Stacey B. Gabriel, Galen F. Gao, Gavin Ha, Matthew Meyerson, Steven E. Schumacher, Juliann Shih, Melanie H. Kucherlapati, Raju S. Kucherlapati, Stephen Baylin, Leslie Cope, Ludmila Danilova, Moiz S. Bootwalla, Phillip H. Lai, Dennis T. Maglinte, David J. Van Den Berg, Daniel J. Weisenberger, J. Todd Auman, Saianand Balu, Tom Bodenheimer, Cheng Fan, Katherine A. Hoadley, Alan P. Hoyle, Stuart R. Jefferys, Corbin D. Jones, Shaowu Meng, Piotr A. Mieczkowski, Lisle E. Mose, Amy H. Perou, Charles M. Perou, Jeffrey Roach, Yan Shi, Janae V. Simons, Tara Skelly, Matthew G. Soloway, Donghui Tan, Umadevi Veluvolu, Huihui Fan, Toshinori Hinoue, Peter W. Laird, Hui Shen, Wanding Zhou, Michelle Bellair, Kyle Chang, Kyle Covington, Chad J. Creighton, Huyen Dinh, HarshaVardhan Doddapaneni, Lawrence A. Donehower, Jennifer Drummond, Richard A. Gibbs, Robert Glenn, Walker Hale, Yi Han, Jianhong Hu, Viktoriya Korchina, Sandra Lee, Lora Lewis, Wei Li, Xiuping Liu, Margaret Morgan, Donna Morton, Donna Muzny, Jireh Santibanez, Margi Sheth, Eve Shinbrot, Linghua Wang, Min Wang, David A. Wheeler, Liu Xi, Fengmei Zhao, Julian Hess, Elizabeth L. Appelbaum, Matthew Bailey, Matthew G. Cordes, Li Ding, Catrina C. Fronick, Lucinda A. Fulton, Robert S. Fulton, Cyriac Kandoth, Elaine R. Mardis, Michael D. McLellan, Christopher A. Miller, Heather K. Schmidt, Richard K. Wilson, Daniel Crain, Erin Curley, Johanna Gardner, Kevin Lau, David Mallery, Scott Morris, Joseph Paulauskis, Robert Penny, Candace Shelton, Troy Shelton, Mark Sherman, Eric Thompson, Peggy Yena, Jay Bowen, Julie M. Gastier-Foster, Mark Gerken, Kristen M. Leraas, Tara M. Lichtenberg, Nilsa C. Ramirez, Lisa Wise, Erik Zmuda, Niall Corcoran, Tony Costello, Christopher Hovens, Andre L. Carvalho, Ana C. de Carvalho, José H. Fregnani, Adhemar Longatto-Filho, Rui M. Reis, Cristovam Scapulatempo-Neto, Henrique C.S. Silveira, Daniel O. Vidal, Andrew Burnette, Jennifer Eschbacher, Beth Hermes, Ardene Noss, Rosy Singh, Matthew L. Anderson, Patricia D. Castro, Michael Ittmann, David Huntsman, Bernard Kohl, Xuan Le, Richard Thorp, Chris Andry, Elizabeth R. Duffy, Vladimir Lyadov, Oxana Paklina, Galiya Setdikova, Alexey Shabunin, Mikhail Tavobilov, Christopher McPherson, Ronald Warnick, Ross Berkowitz, Daniel Cramer, Colleen Feltmate, Neil Horowitz, Adam Kibel, Michael Muto, Chandrajit P. Raut, Andrei Malykh, Jill S. Barnholtz-Sloan, Wendi Barrett, Karen Devine, Jordonna Fulop, Quinn T. Ostrom, Kristen Shimmel, Yingli Wolinsky, Andrew E. Sloan, Agostino De Rose, Felice Giuliante, Marc Goodman, Beth Y. Karlan, Curt H. Hagedorn, John Eckman, Jodi Harr, Jerome Myers, Kelinda Tucker, Leigh Anne Zach, Brenda Deyarmin, Hai Hu, Leonid Kvecher, Caroline Larson, Richard J. Mural, Stella Somiari, Ales Vicha, Tomas Zelinka, Joseph Bennett, Mary Iacocca, Brenda Rabeno, Patricia Swanson, Mathieu Latour, Louis Lacombe, Bernard Têtu, Alain Bergeron, Mary McGraw, Susan M. Staugaitis, John Chabot, Hanina Hibshoosh, Antonia Sepulveda, Tao Su, Timothy Wang, Olga Potapova, Olga Voronina, Laurence Desjardins, Odette Mariani, Sergio Roman-Roman, Xavier Sastre, Marc-Henri Stern, Feixiong Cheng, Sabina Signoretti, Andrew Berchuck, Darell Bigner, Eric Lipp, Jeffrey Marks, Shannon McCall, Roger McLendon, Angeles Secord, Alexis Sharp, Madhusmita Behera, Daniel J. Brat, Amy Chen, Keith Delman, Seth Force, Fadlo Khuri, Kelly Magliocca, Shishir Maithel, Jeffrey J. Olson, Taofeek Owonikoko, Alan Pickens, Suresh Ramalingam, Dong M. Shin, Gabriel Sica, Erwin G. Van Meir, Hongzheng Zhang, Wil Eijckenboom, Ad Gillis, Esther Korpershoek, Leendert Looijenga, Wolter Oosterhuis, Hans Stoop, Kim E. van Kessel, Ellen C. Zwarthoff, Chiara Calatozzolo, Lucia Cuppini, Stefania Cuzzubbo, Francesco DiMeco, Gaetano Finocchiaro, Luca Mattei, Alessandro Perin, Bianca Pollo, Chu Chen, John Houck, Pawadee Lohavanichbutr, Arndt Hartmann, Christine Stoehr, Robert Stoehr, Helge Taubert, Sven Wach, Bernd Wullich, Witold Kycler, Dawid Murawa, Maciej Wiznerowicz, Ki Chung, W. Jeffrey Edenfield, Julie Martin, Eric Baudin, Glenn Bubley, Raphael Bueno, Assunta De Rienzo, William G. Richards, Steven Kalkanis, Tom Mikkelsen, Houtan Noushmehr, Lisa Scarpace, Nicolas Girard, Marta Aymerich, Elias Campo, Eva Giné, Armando López Guillermo, Nguyen Van Bang, Phan Thi Hanh, Bui Duc Phu, Yufang Tang, Howard Colman, Kimberley Evason, Peter R. Dottino, John A. Martignetti, Hani Gabra, Hartmut Juhl, Teniola Akeredolu, Serghei Stepa, Dave Hoon, Keunsoo Ahn, Koo Jeong Kang, Felix Beuschlein, Anne Breggia, Michael Birrer, Debra Bell, Mitesh Borad, Alan H. Bryce, Erik Castle, Vishal Chandan, John Cheville, John A. Copland, Michael Farnell, Thomas Flotte, Nasra Giama, Thai Ho, Michael Kendrick, Jean-Pierre Kocher, Karla Kopp, Catherine Moser, David Nagorney, Daniel O’Brien, Brian Patrick O’Neill, Tushar Patel, Gloria Petersen, Florencia Que, Michael Rivera, Lewis Roberts, Robert Smallridge, Thomas Smyrk, Melissa Stanton, R. Houston Thompson, Michael Torbenson, Ju Dong Yang, Lizhi Zhang, Fadi Brimo, Jaffer A. Ajani, Ana Maria Angulo Gonzalez, Carmen Behrens, Jolanta Bondaruk, Russell Broaddus, Bogdan Czerniak, Bita Esmaeli, Junya Fujimoto, Jeffrey Gershenwald, Charles Guo, Alexander J. Lazar, Christopher Logothetis, Funda Meric-Bernstam, Cesar Moran, Lois Ramondetta, David Rice, Anil Sood, Pheroze Tamboli, Timothy Thompson, Patricia Troncoso, Anne Tsao, Ignacio Wistuba, Candace Carter, Lauren Haydu, Peter Hersey, Valerie Jakrot, Hojabr Kakavand, Richard Kefford, Kenneth Lee, Georgina Long, Graham Mann, Michael Quinn, Robyn Saw, Richard Scolyer, Kerwin Shannon, Andrew Spillane, Jonathan Stretch, Maria Synott, John Thompson, James Wilmott, Hikmat Al-Ahmadie, Timothy A. Chan, Ronald Ghossein, Anuradha Gopalan, Douglas A. Levine, Victor Reuter, Samuel Singer, Bhuvanesh Singh, Nguyen Viet Tien, Thomas Broudy, Cyrus Mirsaidi, Praveen Nair, Paul Drwiega, Judy Miller, Jennifer Smith, Howard Zaren, Joong-Won Park, Nguyen Phi Hung, Electron Kebebew, W. Marston Linehan, Adam R. Metwalli, Karel Pacak, Peter A. Pinto, Mark Schiffman, Laura S. Schmidt, Cathy D. Vocke, Nicolas Wentzensen, Robert Worrell, Hannah Yang, Marc Moncrieff, Chandra Goparaju, Jonathan Melamed, Harvey Pass, Natalia Botnariuc, Irina Caraman, Mircea Cernat, Inga Chemencedji, Adrian Clipca, Serghei Doruc, Ghenadie Gorincioi, Sergiu Mura, Maria Pirtac, Irina Stancul, Diana Tcaciuc, Monique Albert, Iakovina Alexopoulou, Angel Arnaout, John Bartlett, Jay Engel, Sebastien Gilbert, Jeremy Parfitt, Harman Sekhon, George Thomas, Doris M. Rassl, Robert C. Rintoul, Carlo Bifulco, Raina Tamakawa, Walter Urba, Nicholas Hayward, Henri Timmers, Anna Antenucci, Francesco Facciolo, Gianluca Grazi, Mirella Marino, Roberta Merola, Ronald de Krijger, Anne-Paule Gimenez-Roqueplo, Alain Piché, Simone Chevalier, Ginette McKercher, Kivanc Birsoy, Gene Barnett, Cathy Brewer, Carol Farver, Theresa Naska, Nathan A. Pennell, Daniel Raymond, Cathy Schilero, Kathy Smolenski, Felicia Williams, Carl Morrison, Jeffrey A. Borgia, Michael J. Liptay, Mark Pool, Christopher W. Seder, Kerstin Junker, Larsson Omberg, Mikhail Dinkin, George Manikhas, Domenico Alvaro, Maria Consiglia Bragazzi, Vincenzo Cardinale, Guido Carpino, Eugenio Gaudio, David Chesla, Sandra Cottingham, Michael Dubina, Fedor Moiseenko, Renumathy Dhanasekaran, Karl-Friedrich Becker, Klaus-Peter Janssen, Julia Slotta-Huspenina, Mohamed H. Abdel-Rahman, Dina Aziz, Sue Bell, Colleen M. Cebulla, Amy Davis, Rebecca Duell, J. Bradley Elder, Joe Hilty, Bahavna Kumar, James Lang, Norman L. Lehman, Randy Mandt, Phuong Nguyen, Robert Pilarski, Karan Rai, Lynn Schoenfield, Kelly Senecal, Paul Wakely, Paul Hansen, Ronald Lechan, James Powers, Arthur Tischler, William E. Grizzle, Katherine C. Sexton, Alison Kastl, Joel Henderson, Sima Porten, Jens Waldmann, Martin Fassnacht, Sylvia L. Asa, Dirk Schadendorf, Marta Couce, Markus Graefen, Hartwig Huland, Guido Sauter, Thorsten Schlomm, Ronald Simon, Pierre Tennstedt, Oluwole Olabode, Mark Nelson, Oliver Bathe, Peter R. Carroll, June M. Chan, Philip Disaia, Pat Glenn, Robin K. Kelley, Charles N. Landen, Joanna Phillips, Michael Prados, Jeffry Simko, Karen Smith-McCune, Scott VandenBerg, Kevin Roggin, Ashley Fehrenbach, Ady Kendler, Suzanne Sifri, Ruth Steele, Antonio Jimeno, Francis Carey, Ian Forgie, Massimo Mannelli, Michael Carney, Brenda Hernandez, Benito Campos, Christel Herold-Mende, Christin Jungk, Andreas Unterberg, Andreas von Deimling, Aaron Bossler, Joseph Galbraith, Laura Jacobus, Michael Knudson, Tina Knutson, Deqin Ma, Mohammed Milhem, Rita Sigmund, Andrew K. Godwin, Rashna Madan, Howard G. Rosenthal, Clement Adebamowo, Sally N. Adebamowo, Alex Boussioutas, David Beer, Thomas Giordano, Anne-Marie Mes-Masson, Fred Saad, Therese Bocklage, Lisa Landrum, Robert Mannel, Kathleen Moore, Katherine Moxley, Russel Postier, Joan Walker, Rosemary Zuna, Michael Feldman, Federico Valdivieso, Rajiv Dhir, James Luketich, Edna M. Mora Pinero, Mario Quintero-Aguilo, Carlos Gilberto Carlotti, Jr., Jose Sebastião Dos Santos, Rafael Kemp, Ajith Sankarankuty, Daniela Tirapelli, James Catto, Kathy Agnew, Elizabeth Swisher, Jenette Creaney, Bruce Robinson, Carl Simon Shelley, Eryn M. Godwin, Sara Kendall, Cassaundra Shipman, Carol Bradford, Thomas Carey, Andrea Haddad, Jeffey Moyer, Lisa Peterson, Mark Prince, Laura Rozek, Gregory Wolf, Rayleen Bowman, Kwun M. Fong, Ian Yang, Robert Korst, W. Kimryn Rathmell, J. Leigh Fantacone-Campbell, Jeffrey A. Hooke, Albert J. Kovatich, Craig D. Shriver, John DiPersio, Bettina Drake, Ramaswamy Govindan, Sharon Heath, Timothy Ley, Brian Van Tine, Peter Westervelt, Mark A. Rubin, Jung Il Lee, Natália D. Aredes, and Armaz Mariamidze
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Long noncoding RNAs (lncRNAs) are commonly dysregulated in tumors, but only a handful are known to play pathophysiological roles in cancer. We inferred lncRNAs that dysregulate cancer pathways, oncogenes, and tumor suppressors (cancer genes) by modeling their effects on the activity of transcription factors, RNA-binding proteins, and microRNAs in 5,185 TCGA tumors and 1,019 ENCODE assays. Our predictions included hundreds of candidate onco- and tumor-suppressor lncRNAs (cancer lncRNAs) whose somatic alterations account for the dysregulation of dozens of cancer genes and pathways in each of 14 tumor contexts. To demonstrate proof of concept, we showed that perturbations targeting OIP5-AS1 (an inferred tumor suppressor) and TUG1 and WT1-AS (inferred onco-lncRNAs) dysregulated cancer genes and altered proliferation of breast and gynecologic cancer cells. Our analysis indicates that, although most lncRNAs are dysregulated in a tumor-specific manner, some, including OIP5-AS1, TUG1, NEAT1, MEG3, and TSIX, synergistically dysregulate cancer pathways in multiple tumor contexts. : Chiu et al. present a pan-cancer analysis of lncRNA regulatory interactions. They suggest that the dysregulation of hundreds of lncRNAs target and alter the expression of cancer genes and pathways in each tumor context. This implies that hundreds of lncRNAs can alter tumor phenotypes in each tumor context. Keywords: lncRNA, regulation, modulation, cancer gene, pan-cancer, noncoding RNA, microRNA, RNA-binding proteins, interactome
- Published
- 2018
- Full Text
- View/download PDF
29. Contributors
- Author
-
Floor J. Backes, Lindsey B. Beffa, Caroline C. Billingsley, Michael J. Birrer, Kristin Bixel, Teresa K.L. Boitano, Wendy R. Brewster, Dana M. Chase, Shaina Bruce, Angelena Crown, Christina S. Chu, Daniel L. Clarke-Pearson, Robert A. Ross, Brian Crosland, Joshua G. Cohen, Robert L. Coleman, Paul A. DiSilvestro, Oliver Dorigo, Linda R. Duska, Ramez Nassef Eskander, Mary L. Gemignani, Camille Catherine Gunderson, Andrea R. Hagemann, Thomas J. Herzog, Travis R. Korenaga, Warner K. Huh, Lindsay Kuroki, Katherine Kurnit, Robert S. Mannel, L. Stewart Massad, Cara A. Mathews, David S. Miller, Bradley J. Monk, David G. Mutch, Rachita Nikam, JoAnn V. Pinkerton, Matthew Powell, Dominique L. Rash, Lisa M. Landrum, Kari L. Ring, Malte Renz, Brandon Roane, Stephen C. Rubin, Ritu Salani, Jane Satero, Anil K. Sood, John T. Soper, Elizabeth Christina Stock, C. James Sung, Krishnansu Sujata Tewari, Michael D. Toboni, Katherine Tucker, Joan L. Walker, Jaclyn A. Wall, Christina Washington, Lari B. Wenzel, Shannon N. Westin, Catheryn M. Yashar, William T. Creasman, and Rosemary E. Zuna
- Published
- 2023
- Full Text
- View/download PDF
30. Long Noncoding RNA Ceruloplasmin Promotes Cancer Growth by Altering Glycolysis
- Author
-
Rajesha Rupaimoole, Jaehyuk Lee, Monika Haemmerle, Hui Ling, Rebecca A. Previs, Sunila Pradeep, Sherry Y. Wu, Cristina Ivan, Manuela Ferracin, Jennifer B. Dennison, Niki M. Zacharias Millward, Archana S. Nagaraja, Kshipra M. Gharpure, Michael McGuire, Nidhin Sam, Guillermo N. Armaiz-Pena, Nouara C. Sadaoui, Cristian Rodriguez-Aguayo, George A. Calin, Ronny I. Drapkin, Jeffery Kovacs, Gordon B. Mills, Wei Zhang, Gabriel Lopez-Berestein, Pratip K. Bhattacharya, and Anil K. Sood
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Long noncoding RNAs (lncRNAs) significantly influence the development and regulation of genome expression in cells. Here, we demonstrate the role of lncRNA ceruloplasmin (NRCP) in cancer metabolism and elucidate functional effects leading to increased tumor progression. NRCP was highly upregulated in ovarian tumors, and knockdown of NRCP resulted in significantly increased apoptosis, decreased cell proliferation, and decreased glycolysis compared with control cancer cells. In an orthotopic mouse model of ovarian cancer, siNRCP delivered via a liposomal carrier significantly reduced tumor growth compared with control treatment. We identified NRCP as an intermediate binding partner between STAT1 and RNA polymerase II, leading to increased expression of downstream target genes such as glucose-6-phosphate isomerase. Collectively, we report a previously unrecognized role of the lncRNA NRCP in modulating cancer metabolism. As demonstrated, DOPC nanoparticle-incorporated siRNA-mediated silencing of this lncRNA in vivo provides therapeutic avenue toward modulating lncRNAs in cancer.
- Published
- 2015
- Full Text
- View/download PDF
31. The RNA-Binding Protein DDX1 Promotes Primary MicroRNA Maturation and Inhibits Ovarian Tumor Progression
- Author
-
Cecil Han, Yunhua Liu, Guohui Wan, Hyun Jin Choi, Luqing Zhao, Cristina Ivan, Xiaoming He, Anil K. Sood, Xinna Zhang, and Xiongbin Lu
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Posttranscriptional maturation is a critical step in microRNA (miRNA) biogenesis that determines mature miRNA levels. In addition to core components (Drosha and DGCR8 [DiGeorge syndrome critical region gene 8]) in the microprocessor, regulatory RNA-binding proteins may confer the specificity for recruiting and processing of individual primary miRNAs (pri-miRNAs). Here, we identify DDX1 as a regulatory protein that promotes the expression of a subset of miRNAs, including five members in the microRNA-200 (miR-200) family and four miRNAs in an eight-miRNA signature of a mesenchymal ovarian cancer subtype. A majority of DDX1-dependent miRNAs are induced after DNA damage. This induction is facilitated by the ataxia telangiectasia mutated (ATM)-mediated phosphorylation of DDX1. Inhibiting DDX1 promotes ovarian tumor growth and metastasis in a syngeneic mouse model. Analysis of The Cancer Genome Atlas (TCGA) reveals that low DDX1 levels are associated with poor clinical outcome in patients with serous ovarian cancer. These findings suggest that DDX1 is a key modulator in miRNA maturation and ovarian tumor suppression.
- Published
- 2014
- Full Text
- View/download PDF
32. Antagonism of Tumoral Prolactin Receptor Promotes Autophagy-Related Cell Death
- Author
-
Yunfei Wen, Behrouz Zand, Bulent Ozpolat, Miroslaw J. Szczepanski, Chunhua Lu, Erkan Yuca, Amy R. Carroll, Neslihan Alpay, Chandra Bartholomeusz, Ibrahim Tekedereli, Yu Kang, Rajesha Rupaimoole, Chad V. Pecot, Heather J. Dalton, Anadulce Hernandez, Anna Lokshin, Susan K. Lutgendorf, Jinsong Liu, Walter N. Hittelman, Wen Y. Chen, Gabriel Lopez-Berestein, Marta Szajnik, Naoto T. Ueno, Robert L. Coleman, and Anil K. Sood
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Therapeutic upregulation of macroautophagy in cancer cells provides an alternative mechanism for cell death. Prolactin (PRL) and its receptor (PRLR) are considered attractive therapeutic targets because of their roles as growth factors in tumor growth and progression. We utilized G129R, an antagonist peptide of PRL, to block activity of the tumoral PRL/PRLR axis, which resulted in inhibition of tumor growth in orthotopic models of human ovarian cancer. Prolonged treatment with G129R induced the accumulation of redundant autolysosomes in 3D cancer spheroids, leading to a type II programmed cell death. This inducible autophagy was a noncanonical beclin-1-independent pathway and was sustained by an astrocytic phosphoprotein (PEA-15) and protein kinase C zeta interactome. Lower levels of tumoral PRL/PRLR in clinical samples were associated with longer patient survival. Our findings provide an understanding of the mechanisms of tumor growth inhibition through targeting PRL/PRLR and may have clinical implications. : Pharmacological manipulation of autophagy represents a new therapeutic opportunity for cancer. Wen et al. show that blockade of the tumoral PRL/PRLR axis with an antagonist, G129R, induces prolonged autophagy. This inducible autophagy is sustained by the PEA-15 and PKC zeta interactome and leads to type II programmed cell death. There was an inverse correlation between tumoral PRL/PRLR expression and survival of ovarian cancer patients. This study reveals a previously unrecognized mechanism related to targeting the tumoral PRL/PRLR pathway.
- Published
- 2014
- Full Text
- View/download PDF
33. Autocrine Effects of Tumor-Derived Complement
- Author
-
Min Soon Cho, Hernan G. Vasquez, Rajesha Rupaimoole, Sunila Pradeep, Sherry Wu, Behrouz Zand, Hee-Dong Han, Cristian Rodriguez-Aguayo, Justin Bottsford-Miller, Jie Huang, Takahito Miyake, Hyun-Jin Choi, Heather J. Dalton, Cristina Ivan, Keith Baggerly, Gabriel Lopez-Berestein, Anil K. Sood, and Vahid Afshar-Kharghan
- Subjects
Biology (General) ,QH301-705.5 - Abstract
We describe a role for the complement system in enhancing cancer growth. Cancer cells secrete complement proteins that stimulate tumor growth upon activation. Complement promotes tumor growth via a direct autocrine effect that is partially independent of tumor-infiltrating cytotoxic T cells. Activated C5aR and C3aR signal through the PI3K/AKT pathway in cancer cells, and silencing the PI3K or AKT gene in cancer cells eliminates the progrowth effects of C5aR and C3aR stimulation. In patients with ovarian or lung cancer, higher tumoral C3 or C5aR mRNA levels were associated with decreased overall survival. These data identify a role for tumor-derived complement proteins in promoting tumor growth, and they therefore have substantial clinical and therapeutic implications.
- Published
- 2014
- Full Text
- View/download PDF
34. Biologic Effects of Dopamine on Tumor Vasculature in Ovarian Carcinoma
- Author
-
Myrthala Moreno-Smith, Sun Joo Lee, Chunhua Lu, Archana S Nagaraja, Guangan He, Rajesha Rupaimoole, Hee Dong Han, Nicholas B Jennings, Ju-Won Roh, Masato Nishimura, Yu Kang, Julie K Allen, Guillermo N Armaiz, Koji Matsuo, Mian M K Shahzad, Justin Bottsford-Miller, Robert R Langley, Steve W Cole, Susan K Lutgendorf, Zahid H Siddik, and Anil K Sood
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Chronic sympathetic nervous system activation results in increased angiogenesis and tumor growth in orthotopic mouse models of ovarian carcinoma. However, the mechanistic effects of such activation on the tumor vasculature are not well understood. Dopamine (DA), an inhibitory catecholamine, regulates the functions of normal and abnormal blood vessels. Here, we examined whether DA, an inhibitory catecholamine, could block the effects of chronic stress on tumor vasculature and tumor growth. Exogenous administration of DA not only decreased tumor microvessel density but also increased pericyte coverage of tumor vessels following daily restraint stress in mice. Daily restraint stress resulted in significantly increased tumor growth in the SKOV3ip1 and HeyA8 ovarian cancer models. DA treatment blocked stress-mediated increases in tumor growth and increased pericyte coverage of tumor endothelial cells. Whereas the antiangiogenic effect of DA is mediated by dopamine receptor 2 (DR2), our data indicate that DA, through DR1, stimulates vessel stabilization by increasing pericyte recruitment to tumor endothelial cells. DA significantly stimulated migration of mouse 10T1/2 pericyte-like cells in vitro and increased cyclic adenosine mono-phosphate (cAMP) levels in these cells. Moreover, DA or the DR1 agonist SKF 82958 increased platinum concentration in SKOV3ip1 tumor xenografts following cisplatin administration. In conclusion, DA stabilizes tumor blood vessels through activation of pericyte cAMP-protein kinase A signaling pathway by DR1. These findings could have implications for blocking the stimulatory effects of chronic stress on tumor growth.
- Published
- 2013
- Full Text
- View/download PDF
35. Interleukin-1β Promotes Ovarian Tumorigenesis through a p53/NF-κB-Mediated Inflammatory Response in Stromal Fibroblasts
- Author
-
Isaiah Gregory Schauer, Jing Zhang, Zhen Xing, Xiaoqing Guo, Imelda Mercado-Uribe, Anil K. Sood, Peng Huang, and Jinsong Liu
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Cancer has long been considered a disease that mimics an “unhealed wound,” with oncogene-induced secretory activation signals from epithelial cancer cells facilitating stromal fibroblast, endothelial, and inflammatory cell participation in tumor progression. However, the underlying mechanisms that orchestrate cooperative interaction between malignant epithelium and the stroma remain largely unknown. Here, we identified interleukin-1β (IL-1β) as a stromal-acting chemokine secreted by ovarian cancer cells, which suppresses p53 protein expression in cancer-associated fibroblasts (CAFs). Elevated expression of IL-1β and cognate receptor IL-1R1 in ovarian cancer epithelial cells and CAFs independently predicted reduced overall patient survival, as did repressed nuclear p53 in ovarian CAFs. Knockdown of p53 expression in ovarian fibroblasts significantly enhanced the expression and secretion of chemokines IL-8, growth regulated oncogene-alpha (GRO-α), IL-6, IL-1β, and vascular endothelial growth factor (VEGF), significantly increased in vivo mouse xenograft ovarian cancer tumor growth, and was entirely dependent on interaction with, and transcriptional up-regulation of, nuclear factor-kappaB (NF-κB) p65. Our results have uncovered a previously unrecognized circuit whereby epithelial cancer cells use IL-1β as a communication factor instructing stromal fibroblasts through p53 to generate a protumorigenic inflammatory microenvironment. Attenuation of p53 protein expression in stromal fibroblasts generates critical protumorigenic functionality, reminiscent of the role that oncogenic p53 mutations play in cancer cells. These findings implicate CAFs as an important target for blocking inflammation in the tumor microenvironment and reducing tumor growth.
- Published
- 2013
- Full Text
- View/download PDF
36. Cancer-Associated Fibroblasts and Their Putative Role in Potentiating the Initiation and Development of Epithelial Ovarian Cancer
- Author
-
Isaiah G. Schauer, Anil K. Sood, Samuel Mok, and Jinsong Liu
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The progression of ovarian cancer, from cell transformation through invasion of normal tissue, relies on communication between tumor cells and their adjacent stromal microenvironment. Through a natural selection process, an autocrine-paracrine communication loop establishes reciprocal reinforcement of growth and migration signals. Thus, the cancer-activated stromal response is similar to an off-switch-defective form of the normal, universal response needed to survive insult or injury. It is becoming clearer within the cancer literature base that tumor stroma plays a bimodal role in cancer development: it impedes neoplastic growth in normal tissue while encouraging migration and tumor growth in a co-opted desmoplastic response during tumor progression. In this review, we discuss this reciprocal influence that ovarian cancer epithelial cells may have on ovarian stromal cell-reactive phenotype, stromal cell behavior, disrupted signaling networks, and tumor suppressor status in the stroma, within the context of cancer fibroblast studies from alternate cancer tissue settings. We focus on the exchange of secreted factors, in particular interleukin 1β and SDF-1α, between activated fibroblasts and cancer cells as a key area for future investigation and therapeutic development. A better understanding of the bidirectional reliance of early epithelial cancer cells on activated stromal cells could lead to the identification of novel diagnostic stromal markers and targets for therapy.
- Published
- 2011
- Full Text
- View/download PDF
37. Targeted Delivery of Small Interfering RNA Using Reconstituted High-Density Lipoprotein Nanoparticles
- Author
-
Mian M.K. Shahzad, Lingegowda S. Mangala, Hee Dong Han, Chunhua Lu, Justin Bottsford-Miller, Masato Nishimura, Edna M. Mora, Jeong-Won Lee, Rebecca L. Stone, Chad V. Pecot, Duangmani Thanapprapasr, Ju-Won Roh, Puja Gaur, Maya P. Nair, Yun-Yong Park, Nirupama Sabnis, Michael T. Deavers, Ju-Seog Lee, Lee M. Ellis, Gabriel Lopez-Berestein, Walter J. McConathy, Laszlo Prokai, Andras G. Lacko, and Anil K. Sood
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
RNA interference holds tremendous potential as a therapeutic approach, especially in the treatment of malignant tumors. However, efficient and biocompatible delivery methods are needed for systemic delivery of small interfering RNA (siRNA). To maintain a high level of growth, tumor cells scavenge high-density lipoprotein (HDL) particles by overexpressing its receptor: scavenger receptor type B1 (SR-B1). In this study, we exploited this cellular characteristic to achieve efficient siRNA delivery and established a novel formulation of siRNA by incorporating it into reconstituted HDL (rHDL) nanoparticles. Here, we demonstrate that rHDL nanoparticles facilitate highly efficient systemic delivery of siRNA in vivo, mediated by the SR-B1. Moreover, in therapeutic proof-of-concept studies, these nanoparticles were effective in silencing the expression of two proteins that are key to cancer growth and metastasis (signal transducer and activator of transcription 3 and focal adhesion kinase) in orthotopic mouse models of ovarian and colorectal cancer. These data indicate that an rHDL nanoparticle is a novel and highly efficient siRNA carrier, and therefore, this novel technology could serve as the foundation for new cancer therapeutic approaches.
- Published
- 2011
- Full Text
- View/download PDF
38. List of contributors
- Author
-
Arnold P. Advincula, Jamie N. Bakkum-Gamez, Genevieve Bouchard-Fortier, Anne Burke, Leslie H. Clark, Robert L. Coleman, Allan Covens, Deborah S. Cowley, Anne R. Davis, Mary Segars Dolan, Sarah K. Dotters-Katz, Nataki C. Douglas, Sean C. Dowdy, Linda O. Eckert, Michael Fialkow, Eric J. Forman, Michael Frumovitz, Paola Alvarez Gehrig, David M. Gershenson, Jennifer Bushman Gilner, Laura J. Havrilesky, Cherie C. Hill, Hye-Chun Hur, Anuja Jhingran, James M. Kelley, Anna C. Kirby, Jeffrey A. Kuller, Eduardo Lara-Torre, Gretchen M. Lentz, Roger A. Lobo, Karen H. Lu, Vicki Mendiratta, Larissa A. Meyer, Jane L. Miller, Andra Nica, Jaclyn D. Nunziato, James W. Orr, Amanda Padro, Natacha Phoolcharoen, Thomas M. Price, Beth W. Rackow, Pedro T. Ramirez, Licia Raymond, Eleanor H.J. Rhee, Katherine Rivlin, David T. Rock, Timothy Ryntz, Mila Pontremoli Salcedo, Gloria Salvo, Samith Sandadi, Kathleen M. Schmeler, Judith A. Smith, Pamela T. Soliman, Anil K. Sood, Premal H. Thaker, Mireille Truong, Jenna Turocy, Fidel A. Valea, Catherine H. Watson, Shannon N. Westin, and Zev Williams
- Published
- 2022
- Full Text
- View/download PDF
39. RNA delivery for cancer gene therapy
- Author
-
Selin Oncul, Paola Amero, Cristian Rodriguez-Aguayo, Anil K. Sood, George A. Calin, and Gabriel Lopez-Berestein
- Published
- 2022
- Full Text
- View/download PDF
40. Contributors
- Author
-
Mohamad-Gabriel Alameh, Marice Alcantara, Jessica Alluin, Paola Amero, Virginia Arechavala-Gomeza, George A. Calin, David R. Corey, Hagen Cramer, Niels Dammes, Daniel W. Drolet, James Frederiksen, Amy D. Gelinas, Nagy A. Habib, Inbal Hazan-Halevy, Kevin Holm, Nebojsa Janjic, Krystal C. Johnson, Samantha T. Johnson, Edo Kon, Marcin Kortylewski, Dalit Landesman-Milo, Marc M. Lemaitre, Christopher Lincoln, Gabriel Lopez-Berestein, Sean A. Lynch, Selin Oncul, Rachel M. Ostroff, Dan Peer, Cristian Rodriguez-Aguayo, John C. Rohloff, Daniel Rossi, John J. Rossi, Thomas M. Rupp, Daniel J. Schneider, Patricia Soblechero-Martín, Min-Sun Song, Anil K. Sood, Yu-Lin Su, Bruce A. Sullenger, Jon Voutila, Drew Weissman, Preston Williams, Chunsong Yu, Haixiang Yu, Zhuoran Zhang, and Jiehua Zhou
- Published
- 2022
- Full Text
- View/download PDF
41. Tumor-Selective Response to Antibody-Mediated Targeting of αvβ3 Integrin in Ovarian Cancer
- Author
-
Charles N. Landen, Tae-Jin Kim, Yvonne G. Lin, William M. Merritt, Aparna A. Kamat, Liz Y. Han, Whitney A. Spannuth, Alpa M. Nick, Nicholas B. Jennnings, Michael S. Kinch, David Tice, and Anil K. Sood
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The αvβ3 integrin is expressed on proliferating endothelial cells and some cancer cells, but its expression on ovarian cancer cells and its potential as a therapeutic target are unknown. In this study, expression of the αvβ3 integrin on ovarian cancer cell lines and murine endothelial cells was tested, and the effect of a fully humanized monoclonal antibody against αvβ3, Abegrin (etaracizumab), on cell invasion, viability, tumor growth, and the Akt pathway were examined in vitro and in vivo. We found that etaracizumab recognizes αvβ3 on the ovarian cancer cell lines SKOV3ip1, HeyA8, and A2780ip2 (at low levels) but not on murine endothelial cells. Etaracizumab treatment decreased ovarian cancer proliferation and invasion. In vivo, tumor-bearing mice treated with etaracizumab alone gave variable results. There was no effect on A2780ip2 growth, but a 36% to 49% tumor weight reduction in the SKOV3ip1 and HeyA8 models was found (P < .05). However, combined etaracizumab and paclitaxel was superior to paclitaxel in the SKOV3ip1 and A2780ip2 models (by 51–73%, P < .001) but not in the HeyA8 model. Treatment with etaracizumab was then noted to decrease p-Akt and p-mTOR in SKOV3ip1, but not in HeyA8, which is Akt-independent. Tumors resected after therapy showed that etaracizumab treatment reduced the proliferating cell nuclear antigen index but not microvessel density. This study identifies tumor cell αvβ3 integrin as an attractive target and defines the Akt pathway as a predictor of response to function-blocking antibody.
- Published
- 2008
- Full Text
- View/download PDF
42. miRNA551b-3p Activates an Oncostatin Signaling Module for the Progression of Triple-Negative Breast Cancer
- Author
-
Ramani Ramchandran, Deepak Parashar, Gordon B. Mills, Anjali Geethadevi, Andliena Tahiri, Ming You, Anil K. Sood, Jyotsna Mishra, Wei Zhao, Sunila Pradeep, Yiling Lu, Pradeep Chaluvally-Raghavan, Cristian Rodriguez-Aguayo, Gabriel Lopez-Berestein, Lingegowda S. Mangala, Janet S. Rader, Manoj K. Mishra, Jasmine George, Miriam Ragle Aure, Amadou K.S. Camara, Vessela N. Kristensen, Changliang Chen, Bindu Nair, and Mingyu Liang
- Subjects
0301 basic medicine ,STAT3 Transcription Factor ,Transcriptional Activation ,Transcription, Genetic ,Carcinogenesis ,Mice, Nude ,Triple Negative Breast Neoplasms ,Oncostatin M ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Transcription (biology) ,RNA interference ,Cell Movement ,Cell Line, Tumor ,microRNA ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Neoplasm Invasiveness ,Molecular Targeted Therapy ,STAT3 ,lcsh:QH301-705.5 ,Triple-negative breast cancer ,Cell Proliferation ,Cell Nucleus ,biology ,Oncostatin M receptor ,medicine.disease ,beta Karyopherins ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,lcsh:Biology (General) ,Cancer cell ,Cancer research ,biology.protein ,Disease Progression ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Summary: Genomic amplification of 3q26.2 locus leads to the increased expression of microRNA 551b-3p (miR551b-3p) in triple-negative breast cancer (TNBC). Our results demonstrate that miR551b-3p translocates to the nucleus with the aid of importin-8 (IPO8) and activates STAT3 transcription. As a consequence, miR551b upregulates the expression of oncostatin M receptor (OSMR) and interleukin-31 receptor-α (IL-31RA) as well as their ligands OSM and IL-31 through STAT3 transcription. We defined this set of genes induced by miR551b-3p as the “oncostatin signaling module,” which provides oncogenic addictions in cancer cells. Notably, OSM is highly expressed in TNBC, and the elevated expression of OSM associates with poor outcome in estrogen-receptor-negative breast cancer patients. Conversely, targeting miR551b with anti-miR551b-3p reduced the expression of the OSM signaling module and reduced tumor growth, as well as migration and invasion of breast cancer cells. : Parashar et al. demonstrate that the mature microRNA-551b-3p translocates to the nucleus for transcriptional activation of STAT3 gene. As a consequence, miR551b activates an autocrine signaling loop through the upregulation of oncostatin family genes, including oncostatin, and its receptors for the growth and metastasis of triple-negative breast cancer. Keywords: miR551b, oncostatin, STAT3, OSMR, miRNA-therapy, IPO8, RNAi
- Published
- 2019
43. Extensive three-dimensional intratumor proteomic heterogeneity revealed by multiregion sampling in high-grade serous ovarian tumor specimens
- Author
-
Kelly A. Conrads, Kathleen M. Darcy, Brian Blanton, Allison L. Hunt, Craig D. Shriver, Thomas P. Conrads, Uma N. M. Rao, Jeremy Loffredo, Emma L. Robinson, Nicholas W. Bateman, Yovanni Casablanca, Waleed Barakat, Julie Oliver, Tracy J. Litzi, Anil K. Sood, G. Larry Maxwell, Christine Rojas, Mariaelena Pierobon, Ming Zhou, Brian L. Hood, Emanuel F. Petricoin, Glenn Gist, Sasha C. Makohon-Moore, Dave Mitchell, Valerie S. Calvert, and Kunle Odunsi
- Subjects
Proteomics ,Tumor microenvironment ,Multidisciplinary ,Stromal cell ,Cancer systems biology ,Science ,Biology ,Molecular biology ,Epithelium ,Article ,Gene expression profiling ,Serous fluid ,medicine.anatomical_structure ,Stroma ,Oncology ,medicine ,Transcriptomics ,Laser capture microdissection - Abstract
Summary Enriched tumor epithelium, tumor-associated stroma, and whole tissue were collected by laser microdissection from thin sections across spatially separated levels of ten high-grade serous ovarian carcinomas (HGSOCs) and analyzed by mass spectrometry, reverse phase protein arrays, and RNA sequencing. Unsupervised analyses of protein abundance data revealed independent clustering of an enriched stroma and enriched tumor epithelium, with whole tumor tissue clustering driven by overall tumor “purity.” Comparing these data to previously defined prognostic HGSOC molecular subtypes revealed protein and transcript expression from tumor epithelium correlated with the differentiated subtype, whereas stromal proteins (and transcripts) correlated with the mesenchymal subtype. Protein and transcript abundance in the tumor epithelium and stroma exhibited decreased correlation in samples collected just hundreds of microns apart. These data reveal substantial tumor microenvironment protein heterogeneity that directly bears on prognostic signatures, biomarker discovery, and cancer pathophysiology and underscore the need to enrich cellular subpopulations for expression profiling., Graphical abstract, Highlights • LMD was used to investigate 3-D molecular heterogeneity in ovarian cancer tissue • Diverse molecular profiles were identified from 3-D spatially separated samples • Molecular heterogeneity impacts HGSOC prognostic sub-type assignment • Proteomic heterogeneity analysis web portal deployed at www.lmdomics.org, Oncology; Cancer systems biology; Proteomics; Transcriptomics
- Published
- 2021
44. Cyclosporin A Reverses Chemoresistance in Patients With Gynecologic Malignancies
- Author
-
Anil K. Sood, Joel I. Sorosky, Robert C. Squatrito, Jeffrey S. Skilling, Barrie Anderson, and Richard E. Buller
- Subjects
multidrug resistance ,cyclosporine ,resistance modulation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Multidrug resistance is a major obstacle in successful systemic therapy of gynecologic malignancies. The objectives of this study are to evaluate the activity of cyclosporin A used to overcome drug resistance in a variety of gynecologic malignancies. Forty women (29 with ovarian cancer, 7 with uterine cancer, 3 with cervical cancer, and 1 with choriocarcinoma) were treated with cyclosporin A, 4 mg/kg intravenously, 6 hours before and 18 hours after the specific chemotherapeutic agent, to which the tumor had developed drug resistance. All patients had shown resistance to the chemotherapy agent used in combination with cyclosporin A. All patients had been heavily pretreated (mean, 2.8 previous chemotherapy regimens). Overall, among 38 available patients with gynecologic malignancies, a 29% objective response rate was observed. Twenty-six (65%) of all patients received three or more cycles of cyclosporin A. There was a 25% response rate for patients with ovarian cancer patients and 50% for those with uterine cancer. There were no responses among the three patients with cervical cancer, and the patient with choriocarcinoma had a complete response. All patients were evaluable for toxicity. Leukopenia and nausea were the most common toxic reactions, but in most cases they were transient, and only three patients required a treatment delay. The most common grade 3 or 4 toxicity was thrombocytopenia, which was observed in 22% of the patients. Cyclosporin A is well tolerated and has significant potential for reversal of chemoresistance in heavily pretreated patients with ovarian and uterine malignancies.
- Published
- 1999
- Full Text
- View/download PDF
45. Therapeutic Silencing of Bcl-2 by Systemically Administered siRNA Nanotherapeutics Inhibits Tumor Growth by Autophagy and Apoptosis and Enhances the Efficacy of Chemotherapy in Orthotopic Xenograft Models of ER (−) and ER (+) Breast Cancer
- Author
-
Ibrahim Tekedereli, S Neslihan Alpay, Ugur Akar, Erkan Yuca, Cristian Ayugo-Rodriguez, He-Dong Han, Anil K Sood, Gabriel Lopez-Berestein, and Bulent Ozpolat
- Subjects
apoptosis ,autophagy ,Bcl-2 ,breast cancer ,chemotherapy ,estrogen receptor ,gene silencing ,liposome ,nanoparticles ,nanotherapeutics ,programmed cell deathsiRNA ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Bcl-2 is overexpressed in about a half of human cancers and 50–70% of breast cancer patients, thereby conferring resistance to conventional therapies and making it an excellent therapeutic target. Small interfering RNA (siRNA) offers novel and powerful tools for specific gene silencing and molecularly targeted therapy. Here, we show that therapeutic silencing of Bcl-2 by systemically administered nanoliposomal (NL)-Bcl-2 siRNA (0.15 mg siRNA/kg, intravenous) twice a week leads to significant antitumor activity and suppression of growth in both estrogen receptor-negative (ER(−)) MDA-MB-231 and ER-positive (+) MCF7 breast tumors in orthotopic xenograft models (P < 0.05). A single intravenous injection of NL-Bcl-2-siRNA provided robust and persistent silencing of the target gene expression in xenograft tumors. NL-Bcl-2-siRNA treatment significantly increased the efficacy of chemotherapy when combined with doxorubicin in both MDA-MB-231 and MCF-7 animal models (P < 0.05). NL-Bcl-2-siRNA treatment-induced apoptosis and autophagic cell death, and inhibited cyclin D1, HIF1α and Src/Fak signaling in tumors. In conclusion, our data provide the first evidence that in vivo therapeutic targeting Bcl-2 by systemically administered nanoliposomal-siRNA significantly inhibits growth of both ER(−) and ER(+) breast tumors and enhances the efficacy of chemotherapy, suggesting that therapeutic silencing of Bcl-2 by siRNA is a viable approach in breast cancers.
- Published
- 2013
- Full Text
- View/download PDF
46. Corrigendum to ‘Exosomal miRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer’ [EBioMedicine 38 (2018) 100–112]
- Author
-
Anil K. Sood, Nermin Kahraman, Seyda Baydogan, Cristian Rodriguez-Aguayo, Michael L. Gatza, Burcu Aslan, George A. Calin, Bulent Ozpolat, Ozgur Ozkayar, Gabriel Lopez-Berestein, Mohammed H. Rashed, Amr Ahmed El-Arabey, Xinna Zhang, Cristina Ivan, Pinar Kanlikilicer, Recep Bayraktar, Emine Bayraktar, Merve Denizli, Kubra Karagoz, and Rahul Mitra
- Subjects
ATP Binding Cassette Transporter, Subfamily B ,Caveolin 1 ,lcsh:Medicine ,Apoptosis ,Exosomes ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Receptor, Platelet-Derived Growth Factor beta ,Mice ,Cell Line, Tumor ,microRNA ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Macrophage ,Cell Proliferation ,Chemo resistance ,Ovarian Neoplasms ,lcsh:R5-920 ,business.industry ,Gene Expression Profiling ,Macrophages ,lcsh:R ,General Medicine ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,MicroRNAs ,Drug Resistance, Neoplasm ,Cancer research ,Female ,RNA Interference ,Corrigendum ,Ovarian cancer ,business ,lcsh:Medicine (General) ,Signal Transduction - Abstract
Circulating miRNAs are known to play important roles in intercellular communication. However, the effects of exosomal miRNAs on cells are not fully understood.To investigate the role of exosomal miR-1246 in ovarian cancer (OC) microenvironment, we performed RPPA as well as many other in vitro functional assays in ovarian cancer cells (sensitive; HeyA8, Skov3ip1, A2780 and chemoresistant; HeyA8-MDR, Skov3-TR, A2780-CP20). Therapeutic effect of miR-1246 inhibitor treatment was tested in OC animal model. We showed the effect of OC exosomal miR-1246 uptake on macrophages by co-culture experiments.Substantial expression of oncogenic miR-1246 OC exosomes was found. We showed that Cav1 gene, which is the direct target of miR-1246, is involved in the process of exosomal transfer. A significantly worse overall prognosis were found for OC patients with high miR-1246 and low Cav1 expression based on TCGA data. miR-1246 expression were significantly higher in paclitaxel-resistant OC exosomes than in their sensitive counterparts. Overexpression of Cav1 and anti-miR-1246 treatment significantly sensitized OC cells to paclitaxel. We showed that Cav1 and multi drug resistance (MDR) gene is involved in the process of exosomal transfer. Our proteomic approach also revealed that miR-1246 inhibits Cav1 and acts through PDGFβ receptor at the recipient cells to inhibit cell proliferation. miR-1246 inhibitor treatment in combination with chemotherapy led to reduced tumor burden in vivo. Finally, we demonstrated that when OC cells are co-cultured with macrophages, they are capable of transferring their oncogenic miR-1246 to M2-type macrophages, but not M0-type macrophages.Our results suggest that cancer exosomes may contribute to oncogenesis by manipulating neighboring infiltrating immune cells. This study provide a new mechanistic therapeutic approach to overcome chemoresistance and tumor progression through exosomal miR-1246 in OC patients.
- Published
- 2020
47. Carcinoma of the Ovaries and Fallopian Tubes
- Author
-
Robert L. Coleman, Koji Matsuo, S.N. Westin, Premal H. Thaker, Anil K. Sood, and Jinsong Liu
- Subjects
Oncology ,endocrine system ,medicine.medical_specialty ,endocrine system diseases ,business.industry ,medicine.disease ,female genital diseases and pregnancy complications ,Internal medicine ,Gynecologic cancer ,Overall survival ,medicine ,Serous ovarian cancer ,Carcinoma ,Ovarian cancer ,business - Abstract
Ovarian cancer remains the deadliest gynecologic cancer. Although there are many subtypes of ovarian cancer, high-grade serous ovarian cancer accounts for more than 70% of ovarian cancer deaths, and overall survival has not changed significantly for several decades. An improved understanding of the precursor sites has allowed for new prevention strategies. Moreover, rapidly growing knowledge regarding the molecular and cellular features of ovarian cancer has resulted in new and effective therapeutic approaches.
- Published
- 2020
- Full Text
- View/download PDF
48. Contributors
- Author
-
James L. Abbruzzese, Omar Abdel-Wahab, Ghassan K. Abou-Alfa, Janet L. Abrahm, Jeffrey S. Abrams, Jeremy S. Abramson, Dara L. Aisner, Michelle Alonso-Basanta, Jesus Anampa, Megan E. Anderson, Emmanuel S. Antonarakis, Richard Aplenc, Frederick R. Appelbaum, Luiz H. Araujo, Ammar Asban, Edward Ashwood, Farrukh T. Awan, Juliet L. Aylward, Arjun V. Balar, Courtney J. Balentine, Stefan K. Barta, Nancy Bartlett, Karen Basen-Engquist, Lynda Kwon Beaupin, Ross S. Berkowitz, Donald A. Berry, Therese Bevers, John F. Boggess, Julie R. Brahmer, Janet Brown, Karen Brown, Powel Brown, Ilene Browner, Paul A. Bunn, William R. Burns, John C. Byrd, Karen Cadoo, David P. Carbone, H. Ballentine Carter, Jorge J. Castillo, Alfred E. Chang, Eric Chang, Stephen J. Chanock, Claudia I. Chapuy, Vikash P. Chauhan, Herbert Chen, Ronald C. Chen, Nai-Kong V. Cheung, Jennifer H. Choe, Michaele C. Christian, Paul M. Cinciripini, Michael F. Clarke, Robert E. Coleman, Robert L. Coleman, Adriana M. Coletta, Jerry M. Collins, Jean M. Connors, Michael Cools, Kevin R. Coombes, Jorge Cortes, Mauro W. Costa, Anne Covey, Kenneth H. Cowan, Christopher H. Crane, Jeffrey Crawford, Kristy Crooks, Daniel J. Culkin, Brian G. Czito, Piero Dalerba, Josep Dalmau, Mai Dang, Michael D'Angelica, Kurtis D. Davies, Myrtle Davis, Nicolas Dea, Ana De Jesus-Acosta, Angelo M. DeMarzo, Theodore L. DeWeese, Maximilian Diehn, Subba R. Digumarthy, Angela Dispenzieri, Khanh T. Do, Konstantin Dobrenkov, Jeffrey S. Dome, James H. Doroshow, Jay F. Dorsey, Marianne Dubard-Gault, Steven G. DuBois, Dan G. Duda, Malcolm Dunlop, Linda R. Duska, Madeleine Duvic, Imane El Dika, Hashem El-Serag, Jeffrey M. Engelmann, David S. Ettinger, Lola A. Fashoyin-Aje, Eric R. Fearon, James M. Ford, Wilbur A. Franklin, Phoebe E. Freer, Boris Freidlin, Alison G. Freifeld, Terence W. Friedlander, Debra L. Friedman, Arian F. Fuller, Lorenzo Galluzzi, Mark C. Gebhardt, Daniel J. George, Mark B. Geyer, Amato J. Giaccia, Mark R. Gilbert, Whitney Goldner, Donald P. Goldstein, Annekathryn Goodman, Karyn A. Goodman, Kathleen Gordon, Laura Graeff-Armas, Alexander J. Greenstein, Stuart A. Grossman, Stephan Grupp, Arjun Gupta, Irfanullah Haider, Missak Haigentz, John D. Hainsworth, Benjamin E. Haithcock, Christopher L. Hallemeier, Samir Hanash, Aphrothiti J. Hanrahan, James Harding, Michael R. Harrison, Muneer G. Hasham, Ernest Hawk, Jonathan Hayman, Jonathan E. Heinlen, N. Lynn Henry, Joseph Herman, Brian P. Hobbs, Ingunn Holen, Leora Horn, Neil S. Horowitz, Steven M. Horwitz, Odette Houghton, Scott C. Howard, Clifford A. Hudis, Stephen P. Hunger, Arti Hurria, David H. Ilson, Annie Im, Gopa Iyer, Elizabeth M. Jaffee, Reshma Jagsi, Rakesh K. Jain, William Jarnagin, Aminah Jatoi, Anuja Jhingran, David H. Johnson, Brian Johnston, Patrick G. Johnston, Kevin D. Judy, Lisa A. Kachnic, Orit Kaidar-Person, Sanjeeva Kalva, Deborah Y. Kamin, Hagop Kantarjian, Giorgos Karakousis, Maher Karam-Hage, Nadine M. Kaskas, Michael B. Kastan, Nora Katabi, Daniel R. Kaul, Scott R. Kelley, Nancy Kemeny, Erin E. Kent, Oliver Kepp, Simon Khagi, Joshua E. Kilgore, D. Nathan Kim, Bette K. Kleinschmidt-DeMasters, Edward L. Korn, Guido Kroemer, Geoffrey Y. Ku, Shivaani Kummar, Bonnie Ky, Daniel A. Laheru, Paul F. Lambert, Mark Lawler, Jennifer G. Le-Rademacher, John Y.K. Lee, Nancy Y. Lee, Susanna L. Lee, Jonathan E. Leeman, Andreas Linkermann, Jinsong Liu, Simon Lo, Jason W. Locasale, Charles L. Loprinzi, Maeve Lowery, Emmy Ludwig, Matthew A. Lunning, Robert A. Lustig, Mitchell Machtay, Crystal Mackall, David A. Mahvi, David M. Mahvi, Amit Maity, Neil Majithia, Marcos Malumbres, Karen Colbert Maresso, John D. Martin, Koji Matsuo, Natalie H. Matthews, Lauren Mauro, R. Samuel Mayer, Worta McCaskill-Stevens, Megan A. McNamara, Neha Mehta-Shah, Robert E. Merritt, Matthew I. Milowsky, Lori M. Minasian, Tara C. Mitchell, Demytra Mitsis, Michelle Mollica, Margaret Mooney, Farah Moustafa, Lida Nabati, Jarushka Naidoo, Amol Narang, Heidi Nelson, William G. Nelson, Suzanne Nesbit, Mark Niglas, Tracey O'Connor, Kenneth Offit, Mihaela Onciu, Eileen M. O’Reilly, Elaine A. Ostrander, Lisa Pappas-Taffer, Drew Pardoll, Jae H. Park, Anery Patel, Anish J. Patel, Steven R. Patierno, Steven Z. Pavletic, Peter C. Phillips, Miriam D. Post, Amy A. Pruitt, Christiane Querfeld, Vance A. Rabius, S. Vincent Rajkumar, Mohammad O. Ramadan, Erinn B. Rankin, Sushanth Reddy, Michael A. Reid, Scott Reznik, Tina Rizack, Jason D. Robinson, Leslie Robinson-Bostom, Carlos Rodriguez-Galindo, Paul B. Romesser, Steven T. Rosen, Myrna R. Rosenfeld, Nadia Rosenthal, Meredith Ross, Julia H. Rowland, Anthony H. Russell, Michael S. Sabel, Arjun Sahgal, Ryan D. Salinas, Erin E. Salo-Mullen, Manuel Salto-Tellez, Sydney M. Sanderson, John T. Sandlund, Victor M. Santana, Michelle Savage, Eric C. Schreiber, Lynn Schuchter, Liora Schultz, Michael V. Seiden, Morgan M. Sellers, Payal D. Shah, Jinru Shia, Konstantin Shilo, Eric Small, Angela B. Smith, Stephen N. Snow, David B. Solit, Anil K. Sood, Enrique Soto-Perez-de-Celis, Joseph A. Sparano, Vladimir S. Spiegelman, Sheri L. Spunt, Zsofia K. Stadler, David P. Steensma, Richard M. Stone, Steven Kent Stranne, Kelly Stratton, Bill Sugden, Andrew M. Swanson, Martin S. Tallman, James E. Talmadge, David T. Teachey, Catalina V. Teba, Ayalew Tefferi, Bin Tean Teh, Joyce M.C. Teng, Joel E. Tepper, Premal H. Thaker, Aaron P. Thrift, Arthur-Quan Tran, Grace Triska, Donald Trump, Kenneth Tsai, Chia-Lin Tseng, Diane Tseng, Sandra Van Schaeybroeck, Brian A. Van Tine, Erin R. Vanness, Gauri Varadhachary, Marileila Varella-Garcia, Richard L. Wahl, Michael F. Walsh, Thomas Wang, Jared Weiss, Irving L. Weissman, Shannon N. Westin, Jeffrey D. White, Richard Wilson, Richard J. Wong, Gary S. Wood, Yaohui G. Xu, Meng Xu-Welliver, Shlomit Yust-Katz, Timothy Zagar, Elaine M. Zeman, Tian Zhang, and James A. Zwiebel
- Published
- 2020
- Full Text
- View/download PDF
49. Direct Upregulation of STAT3 by MicroRNA-551b-3p Deregulates Growth and Metastasis of Ovarian Cancer
- Author
-
Gabriel Lopez-Berestein, Anil K. Sood, Tyler J. Moss, Sunila Pradeep, Kang Jin Jeong, Yiling Lu, Gordon B. Mills, Pradeep Chaluvally-Raghavan, Jinsong Liu, Andreia M. Silva, Prahlad T. Ram, George A. Calin, Dong Zhang, Shuangxing Yu, Wenbin Liu, and Cristian Rodriguez-Aguayo
- Subjects
0301 basic medicine ,Transcription, Genetic ,RNA polymerase II ,Carcinoma, Ovarian Epithelial ,Metastasis ,Twist transcription factor ,0302 clinical medicine ,Neoplasms, Glandular and Epithelial ,Cell Self Renewal ,Neoplasm Metastasis ,Promoter Regions, Genetic ,lcsh:QH301-705.5 ,Ovarian Neoplasms ,biology ,Nuclear Proteins ,Tumor Burden ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Female ,RNA Polymerase II ,STAT3 Transcription Factor ,Cell Survival ,Down-Regulation ,Mice, Nude ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,Spheroids, Cellular ,microRNA ,medicine ,Gene silencing ,Animals ,Humans ,Gene Silencing ,Transcription factor ,Cell Proliferation ,Twist-Related Protein 1 ,Gene Amplification ,medicine.disease ,MicroRNAs ,030104 developmental biology ,lcsh:Biology (General) ,Cancer cell ,Cancer research ,biology.protein ,Ovarian cancer - Abstract
Summary: 3q26.2 amplification in high-grade serous ovarian cancer leads to increased expression of mature microRNA miR551b-3p, which is associated with poor clinical outcome. Importantly, miR551b-3p contributes to resistance to apoptosis and increased survival and proliferation of cancer cells in vitro and in vivo. miR551b-3p upregulates STAT3 protein levels, and STAT3 is required for the effects of miR551b-3p on cell proliferation. Rather than decreasing levels of target mRNA as expected, we demonstrate that miR551b-3p binds a complementary sequence on the STAT3 promoter, recruiting RNA polymerase II and the TWIST1 transcription factor to activate STAT3 transcription, and thus directly upregulates STAT3 expression. Furthermore, anti-miR551b reduced STAT3 expression in ovarian cancer cells in vitro and in vivo and reduced ovarian cancer growth in vivo. Together, our data demonstrate a role for miR551b-3p in transcriptional activation. Thus, miR551b-3p represents a promising candidate biomarker and therapeutic target in ovarian cancer. : Chaluvally-Raghavan et al. find that miR551b-3p affects transcription of STAT3. The authors find that miR551b-3p binds to the STAT3 promoter and facilitates RNA polymerase II and TWIST1 transcription factor recruitment, which in turn activates STAT3 transcription.
- Published
- 2016
50. Contributors
- Author
-
Sheri A. Babb, Floor J. Backes, Emma L. Barber, Wendy R. Brewster, Dana M. Chase, Christina S. Chu, Daniel L. Clarke-Pearson, David E. Cohn, Robert L. Coleman, Larry J. Copeland, Patricia A. Cronin, William T. Creasman, Philip J. Di Saia, Kemi M. Doll, Eric L. Eisenhauer, Jeffrey M. Fowler, Mary L. Gemignani, Camille C. Gunderson, Chad A. Hamilton, Thomas J. Herzog, Erica R. Hope, Marilyn Huang, Lisa M. Landrum, Robert S. Mannel, Charlotte S. Marcus, L. Stewart Massad, Cara A. Mathews, G. Larry Maxwell, D. Scott McMeekin, David Scott Miller, Bradley J. Monk, David G. Mutch, Emily R. Penick, Stephen C. Rubin, Ritu Salani, Brian M. Slomovitz, Anil K. Sood, John T. Soper, Krishnansu S. Tewari, Joan L. Walker, Lari B. Wenzel, Shannon N. Westin, Siu-Fun Wong, Catheryn M. Yashar, and Rosemary E. Zuna
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.