1
|
Siskar AN, Hanzlik E, Cardenas MF, Eldomery MK, Pinto S, Tinkle CL, Zhang Q, Li X, Lin T, Dhanda SK, Reis G, Li D, Raghavan R, Vortmeyer A, Karajannis MA, Robinson GW, Onar-Thomas A, Blackburn PR, Wheeler DA, Chiang J. FOXR2 activation is not exclusive of CNS neuroblastoma. Neuro Oncol 2025:noaf076. [PMID: 40237561 DOI: 10.1093/neuonc/noaf076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND FOXR2 activation is regarded as pathognomonic for CNS neuroblastoma (NB). However, a comprehensive understanding of the landscape for CNS tumors exhibiting FOXR2 activation is lacking. METHODS Histopathologic, molecular, imaging, and clinical data of 42 CNS tumors with FOXR2 overexpression identified through screening institutional datasets and published institutional cases were analyzed. RESULTS Among the 42 tumors, 21 (50.0%) were high-grade gliomas (HGGs), and 18 (42.9%) were embryonal tumors. The HGGs included ten H3 K27M-mutant diffuse midline gliomas (DMGs) and eight radiation-associated tumors. The embryonal tumors included 11 CNS NBs and six pineoblastomas (PBs). FOXR2 expression was similar between CNS NB and other tumor types (P = 0.82). HGGs with FOXR2 overexpression, unlike NBs and PBs, displayed diverse concomitant genetic alterations. The most common mechanisms of FOXR2 activation involved structural alterations causing promoter donation and enhancer hijacking from active genes essential for brain development, followed by alternative promoter activation or truncated LINE-1 retrotransposition. The preferential activation mechanism varied by tumor type. All but two aberrant FOXR2 transcripts incorporated non-canonical, non-coding exons. Gene set enrichment analysis demonstrated shared downstream effects of FOXR2 activation at the epigenome and transcriptome levels across tumor types. DMGs and PBs with FOXR2 overexpression were aggressive, with 0% 2-year overall survival, whereas CNS NBs responded well to combined chemotherapy and radiation. CONCLUSIONS CNS tumors with FOXR2 overexpression manifest significant histological, molecular, imaging, and clinical diversity. While HGGs and PBs with FOXR2 overexpression demonstrated inferior prognosis, CNS NBs showed favorable outcomes. Integrating histologic and molecular diagnostic approaches is imperative for accurate prognostication and optimal therapeutic decision-making.
Collapse
Affiliation(s)
- Alexa N Siskar
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Emily Hanzlik
- Department of Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Maria F Cardenas
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mohammad K Eldomery
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Soniya Pinto
- Department of Radiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Christopher L Tinkle
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Qunyu Zhang
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoyu Li
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tong Lin
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sandeep K Dhanda
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gerald Reis
- Memorial Healthcare System, Hollywood, FL, USA
| | - Daphne Li
- Pediatric Neurosurgery, Advocate Children's Hospital, Park Ridge, IL, USA
| | - Ravi Raghavan
- Department of Pathology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Alexander Vortmeyer
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Giles W Robinson
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Patrick R Blackburn
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David A Wheeler
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jason Chiang
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
2
|
Royston HN, Hampton AB, Bhagat D, Pinto EF, Emerson MD, Funato K. A human embryonic stem cell-based model reveals the cell of origin of FOXR2-activated CNS neuroblastoma. Neurooncol Adv 2024; 6:vdae144. [PMID: 39220247 PMCID: PMC11364937 DOI: 10.1093/noajnl/vdae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Background FOXR2-activated central nervous system (CNS) neuroblastoma (CNS NB-FOXR2) is a recently identified subtype of brain tumor characterized by the elevated expression of the transcription factor FOXR2 mainly due to genomic rearrangements. However, the precise pathogenic mechanisms, including the cell type of origin, remain elusive. Methods A gene expression analysis of patient tumors was performed to identify putative cell types of origin. Based on this prediction, a new human embryonic stem cell-based model was developed to validate the origin and to examine the molecular and cellular mechanisms underlying the formation of CNS NB-FOXR2. Results Our data showed that CNS NB-FOXR2 tumors express a high level of lineage marker genes associated with the medial ganglionic eminence (MGE), a transient structure located in the developing ventral forebrain. Our model confirmed the cell-type-specific effect of FOXR2 on the proliferation and in vivo tumorigenicity. Additionally, we found that FOXR2 overexpression activated the MEK/ERK signaling pathway through a suppression of the endogenous RAS inhibitor DIRAS3. The MEK inhibitor trametinib suppressed the proliferation of FOXR2-expressing MGE progenitors more than nonexpressing cells. Conclusions Our study collectively demonstrates that MGE progenitors are the cell of origin of CNS NB-FOXR2 and that FOXR2 activates the MEK/ERK signaling pathway, providing a potential therapeutic target.
Collapse
Affiliation(s)
- Hitomi N Royston
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
| | - Autumn B Hampton
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
| | - Dhruv Bhagat
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
| | - Evonne F Pinto
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
| | - Miriam D Emerson
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
| | - Kosuke Funato
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
3
|
Gharbaran R. Insights into the molecular roles of FOXR2 in the pathology of primary pediatric brain tumors. Crit Rev Oncol Hematol 2023; 192:104188. [PMID: 37879492 DOI: 10.1016/j.critrevonc.2023.104188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/23/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023] Open
Abstract
Forkhead box gene R2 (FOXR2) belongs to the family of FOX genes which codes for highly conserved transcription factors (TFs) with critical roles in biological processes ranging from development to organogenesis to metabolic and immune regulation to cellular homeostasis. A number of FOX genes are associated with cancer development and progression and poor prognosis. A growing body of evidence suggests that FOXR2 is an oncogene. Studies suggested important roles for FOXR2 in cancer cell growth, metastasis, and drug resistance. Recent studies showed that FOXR2 is overexpressed by a subset of newly identified entities of embryonal tumors. This review discusses the role(s) FOXR2 plays in the pathology of pediatric brain cancers and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Rajendra Gharbaran
- Biological Sciences Department, Bronx Community College/City University of New York, 2155 University Avenue, Bronx, NY 10453, USA.
| |
Collapse
|
4
|
Song J, Li L, Fang Y, Lin Y, Wu L, Wan W, Wei G, Hua F, Ying J. FOXN Transcription Factors: Regulation and Significant Role in Cancer. Mol Cancer Ther 2023; 22:1028-1039. [PMID: 37566097 DOI: 10.1158/1535-7163.mct-23-0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/29/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023]
Abstract
A growing number of studies have demonstrated that cancer development is closely linked to abnormal gene expression, including alterations in the transcriptional activity of transcription factors. The Forkhead box class N (FOXN) proteins FOXN1-6 form a highly conserved class of transcription factors, which have been shown in recent years to be involved in the regulation of malignant progression in a variety of cancers. FOXNs mediate cell proliferation, cell-cycle progression, cell differentiation, metabolic homeostasis, embryonic development, DNA damage repair, tumor angiogenesis, and other critical biological processes. Therefore, transcriptional dysregulation of FOXNs can directly affect cellular physiology and promote cancer development. Numerous studies have demonstrated that the transcriptional activity of FOXNs is regulated by protein-protein interactions, microRNAs (miRNA), and posttranslational modifications (PTM). However, the mechanisms underlying the molecular regulation of FOXNs in cancer development are unclear. Here, we reviewed the molecular regulatory mechanisms of FOXNs expression and activity, their role in the malignant progression of tumors, and their value for clinical applications in cancer therapy. This review may help design experimental studies involving FOXN transcription factors, and enhance their therapeutic potential as antitumor targets.
Collapse
Affiliation(s)
- Jiali Song
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Longshan Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yang Fang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Luojia Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Wei Wan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Gen Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| |
Collapse
|
5
|
Martins Gama J, Caetano Oliveira R, Teixeira P, Silva F, Abrantes C, Figueiredo P, Simões S, Rebelo O, Cipriano MA. An Immunohistochemical Study of Breast Cancer Brain Metastases: The Role of CD44 and AKT in the Prognosis. Appl Immunohistochem Mol Morphol 2023; 31:318-323. [PMID: 37093706 DOI: 10.1097/pai.0000000000001119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 03/02/2023] [Indexed: 04/25/2023]
Abstract
Breast cancer is a major health burden, and up to one-third of patients with breast cancer develop brain metastases, which are linked to a very poor prognosis. Few biomarkers are available to predict the prognosis of patients with metastases. Assessment by immunohistochemistry may be used as a tool to predict the behavior of these tumors. A retrospective transversal study including 114 patients (diagnosed between 2000 and 2016) with breast cancer brain metastases was carried out using archival biological material from 114 patients with breast cancer brain metastases. Expression of CD44, HER2, ER, PR, CA9, PDL-1, CD133, ALDH1, PTEN, AKT, PI3K, and AR markers was assessed by immunohistochemistry. The overexpression of CD44 and AKT was associated with worse overall survival ( P =0.047 and P =0,034, respectively), on univariate analysis, in the cohort of parenchymal and bone metastases; the impact of AKT expression was also evident in the parenchymal cohort on uni ( P =0.021) and multivariate analysis ( P =0.027). The remaining markers did not exhibit a statistical correlation. Immunohistochemistry markers such as CD44 and AKT may have a prognostic impact on survival in patients with breast cancer brain metastases. The conjugation with other markers may help with the stratification of patients and therapy.
Collapse
Affiliation(s)
| | - Rui Caetano Oliveira
- Department of Pathology
- Clinical Academic Center of Coimbra (CACC)
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Byophysics, Faculty of Medicine, University of Coimbra
| | | | | | - Carlos Abrantes
- Department of Pathology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, Portugal
| | - Paulo Figueiredo
- Department of Pathology, Instituto Português de Oncologia de Coimbra Francisco Gentil, Coimbra, Portugal
| | - Sandra Simões
- Neuropathology Laboratory, Department of Neurology, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário Coimbra
| | - Olinda Rebelo
- Neuropathology Laboratory, Department of Neurology, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário Coimbra
| | | |
Collapse
|
6
|
Schmitt-Hoffner F, van Rijn S, Toprak UH, Mauermann M, Rosemann F, Heit-Mondrzyk A, Hübner JM, Camgöz A, Hartlieb S, Pfister SM, Henrich KO, Westermann F, Kool M. FOXR2 Stabilizes MYCN Protein and Identifies Non- MYCN-Amplified Neuroblastoma Patients With Unfavorable Outcome. J Clin Oncol 2021; 39:3217-3228. [PMID: 34110923 PMCID: PMC8500564 DOI: 10.1200/jco.20.02540] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clinical outcomes of patients with neuroblastoma range from spontaneous tumor regression to fatality. Hence, understanding the mechanisms that cause tumor progression is crucial for the treatment of patients. In this study, we show that FOXR2 activation identifies a subset of neuroblastoma tumors with unfavorable outcome and we investigate the mechanism how FOXR2 relates to poor outcome in patients.
Collapse
Affiliation(s)
- Felix Schmitt-Hoffner
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Sjoerd van Rijn
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Umut H Toprak
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Monika Mauermann
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Felix Rosemann
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Anke Heit-Mondrzyk
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jens-Martin Hübner
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Aylin Camgöz
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Sabine Hartlieb
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kai-Oliver Henrich
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Westermann
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcel Kool
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
7
|
Liao CW, Zheng C, Wang L. Down-regulation of FOXR2 inhibits hypoxia-driven ROS-induced migration and invasion of thyroid cancer cells via regulation of the hedgehog pathway. Clin Exp Pharmacol Physiol 2020; 47:1076-1082. [PMID: 32068268 DOI: 10.1111/1440-1681.13286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 02/08/2020] [Accepted: 02/14/2020] [Indexed: 12/15/2022]
Abstract
Forkhead box R2 (FOXR2), a new member of the FOX family, is involved in a wide range of biological processes such as embryogenesis, differentiation, transformation and metabolic homeostasis. Recently, FOXR2 has been reported to be aberrantly expressed in a variety of cancers and correlated with cancer development. However, the specific role of FOXR2 in thyroid cancer (TC) remains unclear. In this study, we showed that FOXR2 was highly expressed in TC tissues and cell lines. Moreover, down-regulation of FOXR2 inhibited hypoxia-induced reactive oxygen species (ROS) production and migration/invasion of TC cells. We also found that the hedgehog pathway was responsible for the partial mechanisms underlying the inhibitory effect. Taken together, these findings indicated that down-regulation of FOXR2 inhibits hypoxia-driven ROS-induced migration and invasion of TC cells via regulation of the hedgehog pathway. Thus, FOXR2 may hold great potential for TC treatment.
Collapse
Affiliation(s)
- Chong-Wu Liao
- First Department of General Surgery, Xi'an Central Hospital, Xi'an, China
| | - Chen Zheng
- First Department of General Surgery, Xi'an Central Hospital, Xi'an, China
| | - Le Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Transposon Insertion Mutagenesis in Mice for Modeling Human Cancers: Critical Insights Gained and New Opportunities. Int J Mol Sci 2020; 21:ijms21031172. [PMID: 32050713 PMCID: PMC7036786 DOI: 10.3390/ijms21031172] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
Transposon mutagenesis has been used to model many types of human cancer in mice, leading to the discovery of novel cancer genes and insights into the mechanism of tumorigenesis. For this review, we identified over twenty types of human cancer that have been modeled in the mouse using Sleeping Beauty and piggyBac transposon insertion mutagenesis. We examine several specific biological insights that have been gained and describe opportunities for continued research. Specifically, we review studies with a focus on understanding metastasis, therapy resistance, and tumor cell of origin. Additionally, we propose further uses of transposon-based models to identify rarely mutated driver genes across many cancers, understand additional mechanisms of drug resistance and metastasis, and define personalized therapies for cancer patients with obesity as a comorbidity.
Collapse
|
9
|
Zhang Y, Yang W, Li D, Yang JY, Guan R, Yang MQ. Toward the precision breast cancer survival prediction utilizing combined whole genome-wide expression and somatic mutation analysis. BMC Med Genomics 2018; 11:104. [PMID: 30454048 DOI: 10.1109/bibm.2017.8217762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023] Open
Abstract
BACKGROUND Breast cancer is the most common type of invasive cancer in woman. It accounts for approximately 18% of all cancer deaths worldwide. It is well known that somatic mutation plays an essential role in cancer development. Hence, we propose that a prognostic prediction model that integrates somatic mutations with gene expression can improve survival prediction for cancer patients and also be able to reveal the genetic mutations associated with survival. METHOD Differential expression analysis was used to identify breast cancer related genes. Genetic algorithm (GA) and univariate Cox regression analysis were applied to filter out survival related genes. DAVID was used for enrichment analysis on somatic mutated gene set. The performance of survival predictors were assessed by Cox regression model and concordance index(C-index). RESULTS We investigated the genome-wide gene expression profile and somatic mutations of 1091 breast invasive carcinoma cases from The Cancer Genome Atlas (TCGA). We identified 118 genes with high hazard ratios as breast cancer survival risk gene candidates (log rank p < 0.0001 and c-index = 0.636). Multiple breast cancer survival related genes were found in this gene set, including FOXR2, FOXD1, MTNR1B and SDC1. Further genetic algorithm (GA) revealed an optimal gene set consisted of 88 genes with higher c-index (log rank p < 0.0001 and c-index = 0.656). We validated this gene set on an independent breast cancer data set and achieved a similar performance (log rank p < 0.0001 and c-index = 0.614). Moreover, we revealed 25 functional annotations, 15 gene ontology terms and 14 pathways that were significantly enriched in the genes that showed distinct mutation patterns in the different survival risk groups. These functional gene sets were used as new features for the survival prediction model. In particular, our results suggested that the Fanconi anemia pathway had an important role in breast cancer prognosis. CONCLUSIONS Our study indicated that the expression levels of the gene signatures remain the effective indicators for breast cancer survival prediction. Combining the gene expression information with other types of features derived from somatic mutations can further improve the performance of survival prediction. The pathways that were associated with survival risk suggested by our study can be further investigated for improving cancer patient survival.
Collapse
Affiliation(s)
- Yifan Zhang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - William Yang
- Department of Computer Science, Carnegie Mellon University School of Computer Science, 5000 Forbes Ave, Pittsburgh, 24105, USA
| | - Dan Li
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Jack Y Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Renchu Guan
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Mary Qu Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA.
| |
Collapse
|
10
|
Zhang Y, Yang W, Li D, Yang JY, Guan R, Yang MQ. Toward the precision breast cancer survival prediction utilizing combined whole genome-wide expression and somatic mutation analysis. BMC Med Genomics 2018; 11:104. [PMID: 30454048 PMCID: PMC6245494 DOI: 10.1186/s12920-018-0419-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Breast cancer is the most common type of invasive cancer in woman. It accounts for approximately 18% of all cancer deaths worldwide. It is well known that somatic mutation plays an essential role in cancer development. Hence, we propose that a prognostic prediction model that integrates somatic mutations with gene expression can improve survival prediction for cancer patients and also be able to reveal the genetic mutations associated with survival. Method Differential expression analysis was used to identify breast cancer related genes. Genetic algorithm (GA) and univariate Cox regression analysis were applied to filter out survival related genes. DAVID was used for enrichment analysis on somatic mutated gene set. The performance of survival predictors were assessed by Cox regression model and concordance index(C-index). Results We investigated the genome-wide gene expression profile and somatic mutations of 1091 breast invasive carcinoma cases from The Cancer Genome Atlas (TCGA). We identified 118 genes with high hazard ratios as breast cancer survival risk gene candidates (log rank p < 0.0001 and c-index = 0.636). Multiple breast cancer survival related genes were found in this gene set, including FOXR2, FOXD1, MTNR1B and SDC1. Further genetic algorithm (GA) revealed an optimal gene set consisted of 88 genes with higher c-index (log rank p < 0.0001 and c-index = 0.656). We validated this gene set on an independent breast cancer data set and achieved a similar performance (log rank p < 0.0001 and c-index = 0.614). Moreover, we revealed 25 functional annotations, 15 gene ontology terms and 14 pathways that were significantly enriched in the genes that showed distinct mutation patterns in the different survival risk groups. These functional gene sets were used as new features for the survival prediction model. In particular, our results suggested that the Fanconi anemia pathway had an important role in breast cancer prognosis. Conclusions Our study indicated that the expression levels of the gene signatures remain the effective indicators for breast cancer survival prediction. Combining the gene expression information with other types of features derived from somatic mutations can further improve the performance of survival prediction. The pathways that were associated with survival risk suggested by our study can be further investigated for improving cancer patient survival. Electronic supplementary material The online version of this article (10.1186/s12920-018-0419-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yifan Zhang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - William Yang
- Department of Computer Science, Carnegie Mellon University School of Computer Science, 5000 Forbes Ave, Pittsburgh, 24105, USA
| | - Dan Li
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Jack Y Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Renchu Guan
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA
| | - Mary Qu Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Ph.D. Program of University of Arkansas at Little Rock and Univ. of Arkansas Medical Sciences, 2801 S. Univ. Ave, Little Rock, 72204, USA.
| |
Collapse
|
11
|
Leng X, Ma J, Liu Y, Shen S, Yu H, Zheng J, Liu X, Liu L, Chen J, Zhao L, Ruan X, Xue Y. Mechanism of piR-DQ590027/MIR17HG regulating the permeability of glioma conditioned normal BBB. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:246. [PMID: 30305135 PMCID: PMC6180493 DOI: 10.1186/s13046-018-0886-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
Background The blood-brain barrier (BBB) strongly restricts the entry of anti-glioma drugs into tumor tissues and thus decreases chemotherapy efficacy. Malignant gliomas are highly invasive tumours that use the perivascular space for invasion and co-opt existing vessels as satellite tumor form. Because regulation of the effect of noncoding RNA on BBB function is attracting growing attention, we investigated the effects of noncoding RNA on the permeability of glioma conditioned normal BBB and the mechanism involved using PIWI-associated RNA piR-DQ590027 as a starting point. Methods The mRNA levels of MIR17HG, miR-153, miR-377, ZO-1, occludin, and claudin-5 were determined using real-time PCR. Transient cell transfection was performed using Lipofectamine 3000 reagent. TEER and HRP flux were applied to measure the permeability of glioma conditioned normal BBB. Western blotting and immunofluorescence assays were used to measure ZO-1, occludin, and claudin-5 levels. Reporter vector construction and a luciferase reporter assay were performed to detect the binding sites of MIR17HG and piR-DQ590027, MIR17HG and miR-153 (miR-377), and FOXR2 and miR-153 (miR-377). RNA immunoprecipitation was used to test the interaction between miR-153 (miR-377) and its target proteins. Chromatin immunoprecipitation was performed to detect the interaction between the transcription factor FOXR2 and ZO-1, occludin, and claudin-5. Results piR-DQ590027 was expressed at low levels in glioma-conditioned ECs (GECs) of the in vitro glioma conditioned normal BBB model. Overexpression of piR-DQ590027 down-regulated the expressions of ZO-1, occludin, and claudin-5 and increased the permeability of glioma conditioned normal BBB. MIR17HG had high expression in GECs but miR-153 and miR-377 had low expression. piR-DQ590027 bound to and negatively regulated MIR17HG. FOXR2 was a downstream target of miR-153 and miR-377; MIR17HG bound separately to miR-153 and miR-377 and negatively regulated their ability to mediate FOXR2 expression. FOXR2 associated with the promoter regions of ZO-1, occludin, and claudin-5 in GECs to promote their transcription. Conclusion The piR-DQ590027/MIR17HG/miR-153 (miR-377)/FOXR2 pathway plays an important role in regulating glioma conditioned normal BBB permeability and provides a new target for the comprehensive treatment of glioma. Electronic supplementary material The online version of this article (10.1186/s13046-018-0886-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xue Leng
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Shuyuan Shen
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Hai Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Jiajia Chen
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Lini Zhao
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xuelei Ruan
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China. .,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
12
|
Li B, Huang W, Cao N, Lou G. Forkhead‐box R2 promotes metastasis and growth by stimulating angiogenesis and activating hedgehog signaling pathway in ovarian cancer. J Cell Biochem 2018; 119:7780-7789. [PMID: 29943836 DOI: 10.1002/jcb.27148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/18/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Bing Li
- Department of Gynecology Harbin Medical University Cancer Hospital Harbin China
| | - Wei Huang
- Department of Gynecology Harbin Medical University Cancer Hospital Harbin China
| | - Ning Cao
- Department of Gynecology Harbin Medical University Cancer Hospital Harbin China
| | - Ge Lou
- Department of Gynecology Harbin Medical University Cancer Hospital Harbin China
| |
Collapse
|
13
|
Wang J, Li W, Zhao Y, Kang D, Fu W, Zheng X, Pang X, Du G. Members of FOX family could be drug targets of cancers. Pharmacol Ther 2017; 181:183-196. [PMID: 28830838 DOI: 10.1016/j.pharmthera.2017.08.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
FOX families play important roles in biological processes, including metabolism, development, differentiation, proliferation, apoptosis, migration, invasion and longevity. Here we are focusing on roles of FOX members in cancers, FOX members and drug resistance, FOX members and stem cells. Finally, FOX members as drug targets of cancer treatment were discussed. Future perspectives of FOXC1 research were described in the end.
Collapse
Affiliation(s)
- Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China
| | - Wan Li
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China
| | - Ying Zhao
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China
| | - De Kang
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China
| | - Weiqi Fu
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China
| | - Xiangjin Zheng
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China
| | - Xiaocong Pang
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, 100050 Beijing, China.
| |
Collapse
|
14
|
miR-202 Suppresses Cell Proliferation by Targeting FOXR2 in Endometrial Adenocarcinoma. DISEASE MARKERS 2017; 2017:2827435. [PMID: 28827892 PMCID: PMC5554569 DOI: 10.1155/2017/2827435] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/06/2017] [Accepted: 06/18/2017] [Indexed: 12/23/2022]
Abstract
Background MicroRNA-202 (miR-202) has been reported to be aberrantly regulated in several cancers. The aim of this study is to explore the functional role of miR-202 in EAC tumor growth. Material and Methods miR-202 expression was detected by qRT-PCR. TargetScan and luciferase reporter assay were used to elucidate the candidate target gene of miR-202. The FOXR2 protein level was assessed by Western blot and immunohistochemistry. Survival analysis was explored for FOXR2 expression in EAC patients. Results miR-202 expression was significantly decreased in EAC tissues (P < 0.01) compared with that in control tissues. And the downregulate miR-202 was significantly associated with poor prognosis (P < 0.01). Re-expression of miR-202 dramatically suppressed cell proliferation in vitro and tumor growth in vivo. FOXR2 was identified as a direct target of miR-202. In EAC tissues, FOXR2 was upregulated and the increased FOXR2 was significantly associated with poor prognosis. In miR-202-transfected cells, the FOXR2 expression was inversely changed. The analysis of FOXR2 protein expression and miR-202 transcription in EAC tissues showed negative correlation (R = −0.429). Conclusion miR-202 may function as a tumor suppressor in EAC tumor growth by targeting FOXR2 oncogene, which may provide new insights into the molecular mechanism and new targets for treatment of EAC.
Collapse
|
15
|
Lu SQ, Qiu Y, Dai WJ, Zhang XY. FOXR2 Promotes the Proliferation, Invasion, and Epithelial-Mesenchymal Transition in Human Colorectal Cancer Cells. Oncol Res 2017; 25:681-689. [PMID: 28548041 PMCID: PMC7841062 DOI: 10.3727/096504016x14771034190471] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Forkhead box R2 (FOXR2), a member of the FOX gene family, has not been very well investigated for its role in cancer. A recent study has shown that FOXR2 is highly expressed in breast cancer samples and is associated with poor prognosis. In addition, FOXR2 was identified as an oncogene in medulloblastoma. Nevertheless, whether FOXR2 plays a role in colorectal cancer (CRC) remains unclear. In the present study, we conducted several in vitro and in vivo studies to investigate the expression and effect of FOXR2 in CRC. The study results demonstrated that FOXR2 was upregulated in CRC tissues and cells. Downregulation of FOXR2 inhibited CRC cell proliferation, invasion, and the epithelial-mesenchymal transition (EMT) phenotype in vitro and also suppressed CRC cell growth and metastasis in vivo. Furthermore, downregulation of FOXR2 remarkably reduced the protein expression of Shh, Gli1, and Ptch1 in SW480 cells. Taken together, our data suggested that FOXR2 significantly promoted proliferation, invasion, and EMT of CRC cells. All these findings provided evidence for the role of FOXR2 as an oncogene in CRC development.
Collapse
Affiliation(s)
- Sheng-Qiang Lu
- *Department of Anesthesiology, Hubei Cancer Hospital, Wuhan, P.R. China
| | - Yan Qiu
- †Department of Clinical Laboratory, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, P.R. China
| | - Wei-Jie Dai
- ‡Department of Gastroenterology, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, P.R. China
| | - Xiao-Yu Zhang
- §Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University and The Second People’s Hospital of Huai’an, Huai’an, P.R. China
| |
Collapse
|
16
|
FoxR2 promotes glioma proliferation by suppression of the p27 pathway. Oncotarget 2017; 8:56255-56266. [PMID: 28915588 PMCID: PMC5593559 DOI: 10.18632/oncotarget.17447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 04/14/2017] [Indexed: 02/06/2023] Open
Abstract
FoxR2 plays an important role in the development of many human tumors. However, the effects of FoxR2 on tumorigenicity of human glioma remain unclear. In this study, we investigated the roles of FoxR2 in cell proliferation and invasion of glioma. We found that overexpression of FoxR2 promoted the proliferation, migration and invasion of glioma cells. Knockout of FoxR2 induced G1 arrest by decreasing the expression levels of cyclin D1, cyclin E and p-Rb. Mechanistically, upregulation of FoxR2 increased the level and activity of MMP-2 and decreased the expression of p27. Furthermore, overexpression of FoxR2 decreased the nuclear accumulation of p27. Taken together, these results indicate that upregulation of FoxR2 may confer enhanced tumorigenicity in glioma cells.
Collapse
|
17
|
Knockdown of FOXR2 suppresses the tumorigenesis, growth and metastasis of prostate cancer. Biomed Pharmacother 2017; 87:471-475. [PMID: 28068638 DOI: 10.1016/j.biopha.2016.12.120] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 01/05/2023] Open
Abstract
Fork-head box R2 (FOXR2), a member of FOX protein family, was reported to play important roles in the development and progression of cancers. However, the expression and function of FOXR2 in prostate cancer remain unclear. In this study, we investigated the role of FOXR2 in prostate cancer and cancer progression including the molecular mechanism that drives FOXR2-mediated oncogenesis. Our results showed that FOXR2 was overexpressed in prostate cancer cell lines. The in vitro experiments demonstrated that knockdown of FOXR2 significantly repressed the proliferation, migration and invasiveness of prostate cancer cells. Furthermore, the in vivo experiments indicated that knockdown of FOXR2 significantly attenuated prostate cancer growth. Finally, knockdown of FOXR2 significantly down-regulated the protein expression levels of β-catenin, cyclinD1 and c-Myc in DU-145 cells. Taken together, our results demonstrated for the first time that FOXR2 plays a critical role in cell proliferation and invasion, at least in part, through inhibiting the Wnt/β-catenin signaling pathway during prostate cancer progression. Thus, FOXR2 may be an attractive therapeutic target for the treatment of prostate cancer.
Collapse
|
18
|
Wang X, He B, Gao Y, Li Y. FOXR2 contributes to cell proliferation and malignancy in human hepatocellular carcinoma. Tumour Biol 2016; 37:10459-67. [PMID: 26846213 DOI: 10.1007/s13277-016-4923-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/27/2016] [Indexed: 02/06/2023] Open
Abstract
Forkhead box R2 (FOXR2), a member of forkhead box (FOX) family, has been identified as an oncogene in medulloblastoma and breast cancer recently. However, the expression and function of FOXR2 in hepatocellular carcinoma cell (HCC) are still unclear. Here, we report that FOXR2 is frequently upregulated in 25/42 (59.5 %) of HCC specimens compared with neighboring non-cancerous tissues in messenger RNA (mRNA) level and further confirmed by immunohistochemistry analysis in protein level. Cellular function analyses revealed that FOXR2 promoted cell growth and colony formation, whereas knockdown of FOXR2 by RNA inference inhibited cell growth and decreased the growth ability of HCC cells in soft agar. Moreover, we also found FOXR2 overexpression facilitated the development of tumor xenografts in nude mice model. In addition, we validated β-catenin, Skp2, c-Myc, and Gli-1 as the potential downstream effectors of FOXR2 in the regulation of cell proliferation and malignancy by quantitative real-time PCR analysis. Collectively, our data suggest that FOXR2 promotes cell proliferation and malignancy in HCC and could be a novel promising therapeutic target for this disease.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Oncology and Hematology, Shanghai East Hospital, Tongji University School of Medicine, 150 Ji-Mo Road, Shanghai, 200120, China.,Department of Oncology, East Hospital, Dalian Medical University, Shanghai, 200120, China
| | - Bin He
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yong Gao
- Department of Oncology and Hematology, Shanghai East Hospital, Tongji University School of Medicine, 150 Ji-Mo Road, Shanghai, 200120, China
| | - Yandong Li
- Department of Oncology and Hematology, Shanghai East Hospital, Tongji University School of Medicine, 150 Ji-Mo Road, Shanghai, 200120, China.
| |
Collapse
|