1
|
Hsu CC, Yao X, Chen SY, Tsuo TC, Wang IC. The conformation of FOXM1 homodimers in vivo is crucial for regulating transcriptional activities. Nucleic Acids Res 2024; 52:13625-13643. [PMID: 39535028 DOI: 10.1093/nar/gkae988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 09/24/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Conformational changes in a transcription factor can significantly affect its transcriptional activity. The activated form of the FOXM1 transcription factor regulates the transcriptional network of genes essential for cell cycle progression and carcinogenesis. However, the mechanism and impact of FOXM1 conformational change on its transcriptional activity in vivo throughout the cell cycle progression remain unexplored. Here, we demonstrate that FOXM1 proteins form novel intermolecular homodimerizations in vivo, and these conformational changes in FOXM1 homodimers impact activity during the cell cycle. Specifically, during the G1 phase, FOXM1 undergoes autorepressive homodimerization, wherein the αβα motif in the C-terminal transcriptional activation domain interacts with the ββαβ motif in the N-terminal repression domain, as evidenced by FRET imaging. Phosphorylation of the αβα motif by PLK1 at S715/S724 disrupts ββαβ-αβα hydrophobic interactions, thereby facilitating a conserved αβα motif switch binding partner to the novel intrinsically disordered regions, leading to FOXM1 autostimulatory homodimerization persisting from the S phase to the G2/M phase in vivo. Furthermore, we identified a minimal ββαβ motif peptide that effectively inhibits cancer cell proliferation both in cell culture and in a mouse tumor model, suggesting a promising autorepression approach for targeting FOXM1 in cancer therapy.
Collapse
Affiliation(s)
- Chia-Chan Hsu
- Institute of Biotechnology, National Tsing Hua University, No. 101, Sec. 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Xiang Yao
- Institute of Biotechnology, National Tsing Hua University, No. 101, Sec. 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Shang-Yao Chen
- Institute of Biotechnology, National Tsing Hua University, No. 101, Sec. 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Tsui-Chun Tsuo
- National Institute of Environmental Health Sciences, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, Miaoli 350401, Taiwan
| | - I-Ching Wang
- Institute of Biotechnology, National Tsing Hua University, No. 101, Sec. 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Department of Life Sciences, National Tsing Hua University, No. 101, Sec. 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Brain Research Center, National Tsing Hua University, No. 101, Sec. 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| |
Collapse
|
2
|
Chen T, Ni M, Wang H, Xue F, Jiang T, Wu X, Li C, Liang S, Hong L, Wu Q. The Reparative Effect of FOXM1 in Pulmonary Disease. Lung 2024; 203:1. [PMID: 39601876 DOI: 10.1007/s00408-024-00773-4] [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: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
FOXM1, a key member of the FOX transcription factor family, maintains cell homeostasis by accurately controlling diverse biological processes, such as proliferation, cell cycle progression, differentiation, DNA damage repair, tissue homeostasis, angiogenesis, apoptosis, redox signaling, and drug resistance. In recent years, an increasing number of studies have focused on the role of FOXM1 in the occurrence of multiple diseases and various pathophysiological processes. In the field of pulmonary diseases, FOXM1 has a certain reparative effect by promoting cell proliferation, regulating cell cycle, antifibrosis, participating in inflammation regulation, and synergizing with other signaling pathways. On the basis of the repair properties of FOXM1, this review explores its therapeutic potential in acute lung injury/acute respiratory distress syndrome, asthma, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, pulmonary arterial hypertension, lung cancer, and other lung diseases, with the goal of providing a new perspective for the analysis of FOXM1-related mechanism of action and the expansion of clinical treatment strategies.
Collapse
Affiliation(s)
- Tianhao Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Ming Ni
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Hao Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Fei Xue
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Tao Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Xuanpeng Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Chenxi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Shuhao Liang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Leyu Hong
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China
| | - Qifei Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Yan Ta West Road No. 277, Xi'an, 710061, Shaanxi, China.
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.
| |
Collapse
|
3
|
Ito T, Tanaka Y, Kaku-Ito Y, Oda Y, Nakahara T. FOXM1: a new therapeutic target of extramammary Paget disease. Sci Rep 2024; 14:4048. [PMID: 38374400 PMCID: PMC10876583 DOI: 10.1038/s41598-024-54773-8] [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: 10/03/2023] [Accepted: 02/16/2024] [Indexed: 02/21/2024] Open
Abstract
Extramammary Paget disease (EMPD) is a rare skin cancer that primarily affects older individuals predominantly in areas with apocrine sweat glands. Although most early EMPD lesions are indolent, patients with metastatic EMPD have a poor prognosis due to the lack of effective systemic treatment. In this study, we investigated the role of forkhead box M1 (FOXM1), a potent transcription factor, in EMPD and assessed the potential of FOXM1 as a therapeutic target. Immunohistochemistry of 112 primary and 17 metastatic EMPD samples revealed that FOXM1 expression increased with tumor progression. Patients in whom FOXM1 was expressed in more than 10% of tumor cells had significantly shorter disease-specific survival than the other patients (p = 0.0397). In in vitro studies using our newly established EMPD cell line, KS-EMPD-1, we found high expression of FOXM1. Knockdown of FOXM1 impaired tumor cell viability, migration, and invasion. Inhibition of FOXM1 using thiostrepton also reduced tumor cell viability in a dose-dependent manner. These findings suggest that FOXM1 is a promising therapeutic target for patients with EMPD.
Collapse
Affiliation(s)
- Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Yuka Tanaka
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yumiko Kaku-Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
4
|
Zhang N, Gao Y, Bian Q, Wang Q, Shi Y, Zhao Z, Yu H. The role of fascin-1 in the pathogenesis, diagnosis and management of respiratory related cancers. Front Oncol 2022; 12:948110. [PMID: 36033434 PMCID: PMC9404296 DOI: 10.3389/fonc.2022.948110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Human cancer statistics report that respiratory related cancers such as lung, laryngeal, oral and nasopharyngeal cancers account for a large proportion of tumors, and tumor metastasis remains the major reason for patient death. The metastasis of tumor cells requires actin cytoskeleton remodeling, in which fascin-1 plays an important role. Fascin-1 can cross-link F-actin microfilaments into bundles and form finger-like cell protrusions. Some studies have shown that fascin-1 is overexpressed in human tumors and is associated with tumor growth, migration and invasion. The role of fascin-1 in respiratory related cancers is not very clear. The main purpose of this study was to provide an updated literature review on the role of fascin-1 in the pathogenesis, diagnosis and management of respiratory related cancers. These studies suggested that fascin-1 can serve as an emerging biomarker and potential therapeutic target, and has attracted widespread attention.
Collapse
Affiliation(s)
- Naibin Zhang
- Department of biochemistry, Jining Medical University, Jining, China
| | - Yankun Gao
- Department of biochemistry, Jining Medical University, Jining, China
| | - Qiang Bian
- Collaborative Innovation Center, Jining Medical University, Jining, China
- Department of Pathophysiology, Weifang Medical University, Weifang, China
| | - Qianqian Wang
- Department of biochemistry, Jining Medical University, Jining, China
| | - Ying Shi
- Department of biochemistry, Jining Medical University, Jining, China
| | - Zhankui Zhao
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Honglian Yu
- Department of biochemistry, Jining Medical University, Jining, China
- Collaborative Innovation Center, Jining Medical University, Jining, China
- *Correspondence: Honglian Yu,
| |
Collapse
|
5
|
Meng RY, Jin H, Nguyen TV, Chai OH, Park BH, Kim SM. Ursolic Acid Accelerates Paclitaxel-Induced Cell Death in Esophageal Cancer Cells by Suppressing Akt/FOXM1 Signaling Cascade. Int J Mol Sci 2021; 22:11486. [PMID: 34768915 PMCID: PMC8584129 DOI: 10.3390/ijms222111486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/17/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022] Open
Abstract
Ursolic acid (UA), a pentacyclic triterpenoid extracted from various plants, inhibits cell growth, metastasis, and tumorigenesis in various cancers. Chemotherapy resistance and the side effects of paclitaxel (PTX), a traditional chemotherapy reagent, have limited the curative effect of PTX in esophageal cancer. In this study, we investigate whether UA promotes the anti-tumor effect of PTX and explore the underlying mechanism of their combined effect in esophageal squamous cell carcinoma (ESCC). Combination treatment with UA and PTX inhibited cell proliferation and cell growth more effectively than either treatment alone by inducing more significant apoptosis, as indicated by increased sub-G1 phase distribution and protein levels of cleaved-PARP and cleaved caspase-9. Similar to the cell growth suppressive effect, the combination of UA and PTX significantly inhibited cell migration by targeting uPA, MMP-9, and E-cadherin in ESCC cells. In addition, combination treatment with UA and PTX significantly activated p-GSK-3β and suppressed the activation of Akt and FOXM1 in ESCC cells. Those effects were enhanced by the Akt inhibitor LY2940002 and inverted by the Akt agonist SC79. In an in vivo evaluation of a murine xenograft model of esophageal cancer, combination treatment with UA and PTX suppressed tumor growth significantly better than UA or PTX treatment alone. Thus, UA effectively potentiates the anti-tumor efficacy of PTX by targeting the Akt/FOXM1 cascade since combination treatment shows significantly more anti-tumor potential than PTX alone both in vitro and in vivo. Combination treatment with UA and PTX could be a new strategy for curing esophageal cancer patients.
Collapse
Affiliation(s)
- Ruo Yu Meng
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea;
| | - Hua Jin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China;
| | - Thi Van Nguyen
- Department of Anatomy, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea; (T.V.N.); (O.-H.C.)
| | - Ok-Hee Chai
- Department of Anatomy, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea; (T.V.N.); (O.-H.C.)
| | - Byung-Hyun Park
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54907, Korea;
| | - Soo Mi Kim
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea;
| |
Collapse
|
6
|
Li R, Wang X, Zhao X, Zhang X, Chen H, Ma Y, Liu Y. Centromere protein F and Forkhead box M1 correlation with prognosis of non-small cell lung cancer. Oncol Lett 2020; 19:1368-1374. [PMID: 31966068 PMCID: PMC6956421 DOI: 10.3892/ol.2019.11232] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common histological type of lung cancer. Altered expression of centromere protein F (CENPF), a transient kinetochore protein, has been found in a variety of human cancers. However, its clinical significance in NSCLC remains unknown. In the present study the results of quantitative PCR and western blot analyses demonstrated that CENPF and Forkhead box M1 (FOXM1) were significantly higher in NSCLC tissues than in the non-cancerous controls at both transcriptional and translational levels. Immunohistochemical staining results showed 58.7% (44/75) and 64.0% (48/75) of NSCLC tissues displayed high expression of CENPF and FOXM1, respectively. CENPF protein expression showed a positive correlation with tumor size (P=0.0179), vital status (P=0.0008) and FOXM1 expression (P=0.0013) in NSCLC. Poor overall survival was correlated with high levels of CENPF and FOXM1 in NSCLC patients as evaluated by Kaplan-Meier and log rank test. Multivariate analyses showed that CENPF expression was an independent prognostic factor for NSCLC. In conclusion, our study provides evidence of the prognostic function of CENPF in NSCLC.
Collapse
Affiliation(s)
- Rui Li
- Department of Clinical Laboratory, Shenyang Fifth People's Hospital, Shenyang, Liaoning 110021, P.R. China
| | - Xia Wang
- Department of Clinical Laboratory, Shenyang Fifth People's Hospital, Shenyang, Liaoning 110021, P.R. China
| | - Xiaoqian Zhao
- Department of Clinical Laboratory, Shenyang Fifth People's Hospital, Shenyang, Liaoning 110021, P.R. China
| | - Xiaohong Zhang
- Department of Clinical Laboratory, Shenyang Fifth People's Hospital, Shenyang, Liaoning 110021, P.R. China
| | - Honghai Chen
- Department of Clinical Laboratory, Shenyang Fifth People's Hospital, Shenyang, Liaoning 110021, P.R. China
| | - Yue Ma
- Department of Clinical Laboratory, Shenyang Fifth People's Hospital, Shenyang, Liaoning 110021, P.R. China
| | - Yandong Liu
- Admin Office, Shenyang Fifth People's Hospital, Shenyang, Liaoning 110021, P.R. China
| |
Collapse
|
7
|
TRIM6 promotes colorectal cancer cells proliferation and response to thiostrepton by TIS21/FoxM1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:23. [PMID: 31992359 PMCID: PMC6988281 DOI: 10.1186/s13046-019-1504-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/12/2019] [Indexed: 01/26/2023]
Abstract
BACKGROUND Tripartite motif-containing proteins (TRIM) play a crucial role in carcinogenesis. Little attention has been focused on the possible functions of TRIM6 on carcinogenesis. METHODS The expression levels of TRIM6 were assessed in colorectal cancer (CRC) samples. TRIM6 expression was knocked down in CRC cell lines, and subjected to Cell counting kit-8 (CCK-8), bromodeoxyuridine (BrdU) incorporation and cell cycle assays. Immunoprecipitation and proteomics analysis was performed to identify potential associated proteins of TRIM6. RESULTS TRIM6 expression was up-regulated in CRC samples and TRIM6 expression may be an independent prognostic marker for CRC. Knocking down TRIM6 expression suppressed CRC cell proliferation, induced cell cycle arrested at G2/M phase and increased sensitivity to 5-fluorouracil and oxaliplatin. TIS21, an anti-proliferative protein involved in the regulation of G2/M arrest, was identified as an interaction partner of TRIM6. Moreover, CRC cells with TRIM6 overexpression showed decreased TIS21 protein stability. TIS21 ubiquitination was increased in CRC cells overexpressing TRIM6, but not in those overexpressing TRIM6 E3 catalytic mutant (C15A). Further, Lys5 was essential for TRIM6 mediated TIS21 ubiquitination. TIS21 overexpression reversed the induced effects of TRIM6 overexpression on CRC cell proliferation, and the levels of forkhead box M1 (FoxM1), phosphorylated FoxM1, Cyclin B1 and c-Myc. Thiostrepton, a specific inhibitor for FoxM1, was less effective in anti-proliferative activity against CRC cells with lower level of TRIM6 in vitro and in vivo. CONCLUSIONS Our study suggests that TRIM6 promotes the progression of CRC via TIS21/FoxM1.
Collapse
|
8
|
Bezzecchi E, Ronzio M, Dolfini D, Mantovani R. NF-YA Overexpression in Lung Cancer: LUSC. Genes (Basel) 2019; 10:genes10110937. [PMID: 31744190 PMCID: PMC6895822 DOI: 10.3390/genes10110937] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/04/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022] Open
Abstract
The CCAAT box is recognized by the trimeric transcription factor NF-Y, whose NF-YA subunit is present in two major splicing isoforms, NF-YAl (“long”) and NF-YAs (“short”). Little is known about the expression levels of NF-Y subunits in tumors, and nothing in lung cancer. By interrogating RNA-seq TCGA and GEO datasets, we found that, unlike NF-YB/NF-YC, NF-YAs is overexpressed in lung squamous cell carcinomas (LUSC). The ratio of the two isoforms changes from normal to cancer cells, with NF-YAs becoming predominant in the latter. NF-YA increased expression correlates with common proliferation markers. We partitioned all 501 TCGA LUSC tumors in the four molecular cohorts and verified that NF-YAs is similarly overexpressed. We analyzed global and subtype-specific RNA-seq data and found that CCAAT is the most abundant DNA matrix in promoters of genes overexpressed in all subtypes. Enriched Gene Ontology terms are cell-cycle and signaling. Survival curves indicate a worse clinical outcome for patients with increasing global amounts of NF-YA; same with hazard ratios with very high and, surprisingly, very low NF-YAs/NF-YAl ratios. We then analyzed gene expression in this latter cohort and identified a different, pro-migration signature devoid of CCAAT. We conclude that overexpression of the NF-Y regulatory subunit in LUSC has the scope of increasing CCAAT-dependent, proliferative (NF-YAshigh) or CCAAT-less, pro-migration (NF-YAlhigh) genes. The data further reinstate the importance of analysis of single isoforms of TFs involved in tumor development.
Collapse
|
9
|
Ahmed F. Integrated Network Analysis Reveals FOXM1 and MYBL2 as Key Regulators of Cell Proliferation in Non-small Cell Lung Cancer. Front Oncol 2019; 9:1011. [PMID: 31681566 PMCID: PMC6804573 DOI: 10.3389/fonc.2019.01011] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Loss of control on cell division is an important factor for the development of non-small cell lung cancer (NSCLC), however, its molecular mechanism and gene regulatory network are not clearly understood. This study utilized the systems bioinformatics approach to reveal the “driver-network” involve in tumorigenic processes in NSCLC. Methods: A meta-analysis of gene expression data of NSCLC was integrated with protein-protein interaction (PPI) data to construct an NSCLC network. MCODE and iRegulone were used to identify the local clusters and its upstream transcription regulators involve in NSCLC. Pair-wise gene expression correlation was performed using GEPIA. The survival analysis was performed by the Kaplan-Meier plot. Results: This study identified a local “driver-network” with highest MCODE score having 26 up-regulated genes involved in the process of cell proliferation in NSCLC. Interestingly, the “driver-network” is under the regulation of TFs FOXM1 and MYBL2 as well as miRNAs. Furthermore, the overexpression of member genes in “driver-network” and the TFs are associated with poor overall survival (OS) in NSCLC patients. Conclusion: This study identified a local “driver-network” and its upstream regulators responsible for the cell proliferation in NSCLC, which could be promising biomarkers and therapeutic targets for NSCLC treatment.
Collapse
Affiliation(s)
- Firoz Ahmed
- Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia.,University of Jeddah Center for Scientific and Medical Research, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
Zhang G, Wang W, Huang W, Xie X, Liang Z, Cao H. Cross-disease analysis identified novel common genes for both lung adenocarcinoma and lung squamous cell carcinoma. Oncol Lett 2019; 18:3463-3470. [PMID: 31516564 PMCID: PMC6732964 DOI: 10.3892/ol.2019.10678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 05/25/2019] [Indexed: 12/25/2022] Open
Abstract
Lung squamous cell carcinoma (LSCC) exhibits a number of similarities with lung adenocarcinoma (LA) in terms of copy number alterations. However, compared with LA, the range of genetic alterations in LSCC is less understood. In the present study, a large-scale literature-based search of LA-associated genes and LSCC-associated genes was performed to identify the genetic basis in common with these two diseases. For each of the LA-associated genes, a mega-analysis was performed to test its expression variations in LSCC using 11 RNA expression datasets, with significant genes identified using statistical analysis. Subsequently, a functional pathway analysis was performed to identify a possible association between any of the significant genes identified from the mega-analysis and LSCC, followed by a co-expression analysis. A multiple linear regression (MLR) model was employed to investigate the possible influence of sample size, country of origin and study date on gene expression in patients with LSCC. Disease-gene association data analysis identified 1,178 genes involved in LA, 334 in LSCC, with a significant overlap of 187 genes (P<1.02×−161). Mega-analysis revealed that three LA-associated genes, such as solute carrier family 2 member 1 (SLC2A1), endothelial PAS domain protein 1 (EPAS1) and cyclin-dependent kinase 4 (CDK4), were significantly associated with LSCC (P<1.60×10−8), with multiple potential pathways identified by functional pathway analysis, which were further validated by co-expression analysis. The present MLR analysis suggested that the country of origin was a significant factor for the levels of expression of all three genes in patients with LSCC (P<4.0×10−3). Collectively, the present results suggested that genes associated with LA should be further investigated for their association with LSCC. In addition, SLC2A1, EPAS1 and CDK4 may be novel risk genes associated with LA and LSCC.
Collapse
Affiliation(s)
- Guanghui Zhang
- Department of Cardiothoracic Surgery, Ningbo Fourth Hospital, Ningbo, Zhejiang 315037, P.R. China
| | - Weijie Wang
- Department of Cardiothoracic Surgery, Ningbo Fourth Hospital, Ningbo, Zhejiang 315037, P.R. China
| | - Weiyang Huang
- Department of Cardiothoracic Surgery, Ningbo Fourth Hospital, Ningbo, Zhejiang 315037, P.R. China
| | - Xiaoli Xie
- Department of Cardiothoracic Surgery, Ningbo Fourth Hospital, Ningbo, Zhejiang 315037, P.R. China
| | - Zhigang Liang
- Department of Thoracic Surgery, Ningbo First Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Hongbao Cao
- Statistical Genomics and Data Analysis Core, National Institutes of Health, Bethesda, MD 20852, USA
| |
Collapse
|
11
|
Tang C, Liu T, Wang K, Wang X, Xu S, He D, Zeng J. Transcriptional regulation of FoxM1 by HIF‑1α mediates hypoxia‑induced EMT in prostate cancer. Oncol Rep 2019; 42:1307-1318. [PMID: 31364741 PMCID: PMC6718104 DOI: 10.3892/or.2019.7248] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/26/2019] [Indexed: 01/06/2023] Open
Abstract
Hypoxia is a tumorigenesis-related microenvironment change which usually occurs in the earliest stage of prostate cancer (PCa) development. Accumulating evidence has demonstrated that hypoxia/hypoxia-inducing factor (HIF) is involved in the induction of epithelial-mesenchymal transition (EMT) and increased metastatic potential in PCa. However, the mechanism by which hypoxia/HIF regulates EMT remains unclear. In the present study, we demonstrated the molecular mechanisms of hypoxia-induced EMT in PCa, focusing on HIF-1α/Forkhead box M1 (FoxM1) signaling pathway. PCa PC3 and DU145 cell lines were used as the model system in vitro. Our data revealed that hypoxia induced EMT in PCa cells. Bioinformatics analysis identified the possible association between HIF-1α and FoxM1. Additionally, FoxM1 was significantly associated with PCa development and Gleason scores of PCa. Exposure to hypoxia resulted in the increased expression of HIF-1α and FoxM1. Genetic knockdown FoxM1 abolished hypoxia-induced EMT in PCa, while exogenous overexpression of FoxM1 facilitated hypoxia-induced EMT. Furthermore, the increase of FoxM1 during hypoxia was due to the transcriptional regulation on the FoxM1 promoter by HIF-1α. We also confirmed the binding site of HIF-1α on the FoxM1 promoter by different lengths promoter sequences. These findings provide new insights into how EMT is regulated in PCa under hypoxic stress. It is worthwhile to investigate in future that inhibition of FoxM1 as a potential target may be an effective therapeutic strategy against PCa.
Collapse
Affiliation(s)
- Cong Tang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
12
|
Li Y, Wu F, Tan Q, Guo M, Ma P, Wang X, Zhang S, Xu J, Luo P, Jin Y. The multifaceted roles of FOXM1 in pulmonary disease. Cell Commun Signal 2019; 17:35. [PMID: 30992007 PMCID: PMC6469073 DOI: 10.1186/s12964-019-0347-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/31/2019] [Indexed: 02/06/2023] Open
Abstract
Forkhead box M1 (FOXM1), a transcriptional regulator of G1/S and G2/M transition and M phase progression in the cell cycle, plays a principal role in many physiological and pathological processes. A growing number of studies have focused on the relationship between abnormal FOXM1 expression and pulmonary diseases, such as lung cancer, chronic obstructive pulmonary disease (COPD), asthma, acute lung injury (ALI), pulmonary fibrosis, and pulmonary arterial hypertension (PAH). These studies indicate that the FOXM1 regulatory network is a major predictor of poor outcomes, especially in lung cancer, and provide novel insight into various pulmonary diseases. For the first time, this review summarizes the mechanistic relationship between FOXM1 dysregulation and pulmonary diseases, the benefits of targeting abnormal FOXM1 expression, and the questions that remain to be addressed in the future.
Collapse
Affiliation(s)
- Yumei Li
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Feng Wu
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Xuan Wang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Shuai Zhang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Juanjuan Xu
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Ping Luo
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
13
|
Tight correlation between FoxM1 and FoxP3+ Tregs in gastric cancer and their clinical significance. Clin Exp Med 2018; 18:413-420. [PMID: 29804142 DOI: 10.1007/s10238-018-0505-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 05/02/2018] [Indexed: 12/25/2022]
Abstract
The aim of the present study was to investigate the expression of Forkhead box transcription M1 (FoxM1) and Forkhead box transcription P3 (FoxP3) in gastric cancer tissues in order to reveal any correlation between FoxM1, FoxP3 and clinicopathological parameters. Their clinical significance in gastric cancer was also investigated. Immunohistochemistry was used to detect the expression of FoxM1 and FoxP3 in gastric cancer and para-cancer tissues. The clinical significance of FoxM1 and FoxP3 in gastric cancer was explored, and the association between FoxM1 and FoxP3 was further analyzed. As a result, the overexpression of FoxM1 and FoxP3 was evident in gastric cancer (P < 0.001). FoxM1 overexpression was showed to be correlated with late AJCC stage (P = 0.025), while positive tumoral FoxP3 expression was associated with deeper invasion (P = 0.020), lymph node metastasis (P = 0.019) and later AJCC stage (P = 0.024). Overexpression of FoxM1 or FoxP3 was revealed to be negative prognostic factors for survival duration (P < 0.05), whereas only FoxM1 was shown to be independently associated with prognosisin gastric cancer after multivariate analysis (P = 0.020). A significant and positive correlation between FoxM1 and FoxP3 expressions was finally confirmed (P = 0.001). This significantly positive correlation between FoxM1 and FoxP3 prompts that FoxM1 may induce immune inhibition by recruiting FoxP3+ Tregs, leading to the progression of carcinogenesis, invasion and metastasis.
Collapse
|
14
|
Li L, Wu D, Yu Q, Li L, Wu P. Prognostic value of FOXM1 in solid tumors: a systematic review and meta-analysis. Oncotarget 2018; 8:32298-32308. [PMID: 28427178 PMCID: PMC5458285 DOI: 10.18632/oncotarget.15764] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
Accumulated studies have provided controversial evidences of the association between Forkhead Box M1 (FOXM1) expression and survival of human solid tumors. To address this inconsistency, we performed a meta-analysis with 23 studies identified from PubMed and Medline. We found elevated FOXM1-protein expression was significantly associated with worse 3-year overall survival (OS) (OR = 3.30, 95% CI = 2.56 to 4.25, P < 0.00001) 5-year OS (OR =3.35, 95% CI = 2.64 to 4.26, P < 0.00001) and 10-year OS (OR = 5.24, 95% CI = 2.61 to 10.52, P < 0.00001) of human solid tumors. Similar results were observed when disease free survival (DFS) were analyzed. Subgroup analysis showed that FOXM1 overexpression was associated with poor prognosis of colorectal cancer, gastric cancer, hepatic cancer, lung cancer and ovarian cancer. High expression level of FOXM1 was also associated with advanced tumor stage. In conclusion, elevated FOXM1 expression is associated with poor survival in most solid tumors. FOXM1 is a potential biomarker for prognosis prediction and a promising therapeutic target in human solid tumors.
Collapse
Affiliation(s)
- Lijun Li
- Department of Surgery, Hangzhou Xixi Hospital, Hangzhou, China
| | - Dang Wu
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Qun Yu
- Fourth Ward of Neurosurgery, Division of Nursing, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Lingdi Li
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, China
| | - Pin Wu
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Lv C, Zhao G, Sun X, Wang P, Xie N, Luo J, Tong T. Acetylation of FOXM1 is essential for its transactivation and tumor growth stimulation. Oncotarget 2018; 7:60366-60382. [PMID: 27542221 PMCID: PMC5312389 DOI: 10.18632/oncotarget.11332] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 07/10/2016] [Indexed: 11/25/2022] Open
Abstract
Forkhead box transcription factor M1 (FOXM1) plays crucial roles in a wide array of biological processes, including cell proliferation and differentiation, the cell cycle, and tumorigenesis by regulating the expression of its target genes. Elevated expression of FOXM1 is frequently observed in a multitude of malignancies. Here we show that FOXM1 can be acetylated by p300/CBP at lysines K63, K422, K440, K603 and K614 in vivo. This modification is essential for its transactivation on the target genes. Acetylation of FOXM1 increases during the S phase and remains high throughout the G2 and M phases, when FOXM1 transcriptional activity is required. We find that the acetylation-deficient FOXM1 mutant is less active and exhibits significantly weaker tumorigenic activities compared to wild-type FOXM1. Mechanistically, the acetylation of FOXM1 enhances its transcriptional activity by increasing its DNA binding affinity, protein stability, and phosphorylation sensitivity. In addition, we demonstrate that NAD-dependent histone deacetylase SIRT1 physically binds to and deacetylates FOXM1 in vivo. The deacetylation of FOXM1 by SIRT1 attenuates its transcriptional activity and decreases its protein stability. Together, our findings demonstrate that the reversible acetylation of FOXM1 by p300/CBP and SIRT1 modulates its transactivation function.
Collapse
Affiliation(s)
- Cuicui Lv
- Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Ganye Zhao
- Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Xinpei Sun
- Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Pan Wang
- Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Nan Xie
- Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| | - Jianyuan Luo
- Center for Medical Genetics, Department of Medical Genetics, Peking University Health Science Center, Beijing, China
| | - Tanjun Tong
- Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
16
|
Hamurcu Z, Ashour A, Kahraman N, Ozpolat B. FOXM1 regulates expression of eukaryotic elongation factor 2 kinase and promotes proliferation, invasion and tumorgenesis of human triple negative breast cancer cells. Oncotarget 2017; 7:16619-35. [PMID: 26918606 PMCID: PMC4941339 DOI: 10.18632/oncotarget.7672] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 01/06/2016] [Indexed: 12/14/2022] Open
Abstract
Eukaryotic elongation factor 2 kinase (eEF2K), an emerging molecular target for cancer therapy, contributes to cancer proliferation, cell survival, tumorigenesis, and invasion, disease progression and drug resistance. Although eEF2K is highly up-regulated in various cancers, the mechanism of gene regulation has not been elucidated. In this study, we examined the role of Forkhead Box M1 (FOXM1) proto-oncogenic transcription factor in triple negative breast cancer (TNBC) cells and the regulation of eEF2K. We found that FOXM1 is highly upregulated in TNBC and its knockdown by RNA interference (siRNA) significantly inhibited eEF2K expression and suppressed cell proliferation, colony formation, migration, invasion and induced apoptotic cell death, recapitulating the effects of eEF2K inhibition. Knockdown of FOXM1 inhibited regulators of cell cycle, migration/invasion and survival, including cyclin D1, Src and MAPK-ERK signaling pathways, respectively. We also demonstrated that FOXM1 (1B and 1C isoforms) directly binds to and transcriptionally regulates eEF2K gene expression by chromatin immunoprecipitation (ChIP) and luciferase gene reporter assays. Furthermore, in vivo inhibition of FOXM1 by liposomal siRNA-nanoparticles suppressed growth of MDA-MB-231 TNBC tumor xenografts in orthotopic models. In conclusion, our study provides the first evidence about the transcriptional regulation of eEF2K in TNBC and the role of FOXM1 in mediating breast cancer cell proliferation, survival, migration/invasion, progression and tumorgenesis and highlighting the potential of FOXM1/eEF2K axis as a molecular target in breast and other cancers.
Collapse
Affiliation(s)
- Zuhal Hamurcu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Faculty of Medicine, Department of Medical Biology, Erciyes University, Kayseri, Turkey.,Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Ahmed Ashour
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nermin Kahraman
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
17
|
He S, Liao B, Deng Y, Su C, Tuo J, Liu J, Yao S, Xu L. MiR-216b inhibits cell proliferation by targeting FOXM1 in cervical cancer cells and is associated with better prognosis. BMC Cancer 2017; 17:673. [PMID: 28978307 PMCID: PMC5628450 DOI: 10.1186/s12885-017-3650-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 09/21/2017] [Indexed: 02/07/2023] Open
Abstract
Background Our previous study showed FOXM1 expression was significantly up-regulated in cervical cancer, and was associated with poor prognosis. To clarify miRNAs-FOXM1 modulation pathways, in this study, we investigated the relationships between miR-216b and FOXM1 and the role of miR-216b in cell proliferation and prognosis of cervical cancer patients. Methods Western blotting and qPCR were used to determine expression of FOXM1, cell cycle related factors and miR-216b level. MiR-216b overexpression and inhibited cell models were constructed, and siRNA was used for FOXM1 silencing. Cell proliferation was analyzed by MTT and colony formation assay. Dual luciferase reporter assay system was used to clarify the relationships between miR-216b and FOXM1. Kaplan-Meier survival analysis was used to evaluate prognosis. Results MiR-216b was down-regulated in cervical cancer cells and tissues, and its ectopic expression could decrease cell proliferation. Western blotting analysis showed miR-216b can inhibit cell proliferation by regulating FOXM1-related cell cycle factors, suppressing cyclinD1, c-myc, LEF1 and p-Rb and enhancing p21 expression. Repressing of miR-216b stimulated cervical cancer cell proliferation, whereas silencing FOXM1 expression could reverse this effect. Western blotting and luciferase assay results proved FOXM1 is a direct target of miR-216b. Survival analysis showed higher level of miR-216b was associated with better prognosis in cervical cancer patients. Conclusions FOXM1 expression could be suppressed by miR-216b via direct binding to FOXM1 3′-UTR and miR-216b could inhibit cell proliferation by regulating FOXM1 related Wnt/β-catenin signal pathway. MiR-216b level is related to prognosis in cervical cancer patients and may serve as a potential prognostic marker. Electronic supplementary material The online version of this article (10.1186/s12885-017-3650-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shanyang He
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510700, China
| | - Bing Liao
- Department of Pathology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yalan Deng
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510700, China
| | - Chang Su
- Department of Hematology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiuling Tuo
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jun Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510700, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510700, China.
| | - Lin Xu
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
18
|
Nandi D, Cheema PS, Jaiswal N, Nag A. FoxM1: Repurposing an oncogene as a biomarker. Semin Cancer Biol 2017; 52:74-84. [PMID: 28855104 DOI: 10.1016/j.semcancer.2017.08.009] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 08/08/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022]
Abstract
The past few decades have witnessed a tremendous progress in understanding the biology of cancer, which has led to more comprehensive approaches for global gene expression profiling and genome-wide analysis. This has helped to determine more sophisticated prognostic and predictive signature markers for the prompt diagnosis and precise screening of cancer patients. In the search for novel biomarkers, there has been increased interest in FoxM1, an extensively studied transcription factor that encompasses most of the hallmarks of malignancy. Considering the attractive potential of this multifarious oncogene, FoxM1 has emerged as an important molecule implicated in initiation, development and progression of cancer. Bolstered with the skill to maneuver the proliferation signals, FoxM1 bestows resistance to contemporary anti-cancer therapy as well. This review sheds light on the large body of literature that has accumulated in recent years that implies that FoxM1 neoplastic functions can be used as a novel predictive, prognostic and therapeutic marker for different cancers. This assessment also highlights the key features of FoxM1 that can be effectively harnessed to establish FoxM1 as a strong biomarker in diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Deeptashree Nandi
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Pradeep Singh Cheema
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Neha Jaiswal
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|
19
|
Oxidative Stress Gene Expression Profile Correlates with Cancer Patient Poor Prognosis: Identification of Crucial Pathways Might Select Novel Therapeutic Approaches. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2597581. [PMID: 28770020 PMCID: PMC5523271 DOI: 10.1155/2017/2597581] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/30/2017] [Indexed: 12/17/2022]
Abstract
The role of altered redox status and high reactive oxygen species (ROS) is still controversial in cancer development and progression. Intracellular levels of ROS are elevated in cancer cells suggesting a role in cancer initiation and progression; on the contrary, ROS elevated levels may induce programmed cell death and have been associated with cancer suppression. Thus, it is crucial to consider the double-face of ROS, for novel therapeutic strategies targeting redox regulatory mechanisms. In this review, in order to derive cancer-type specific oxidative stress genes' profile and their potential prognostic role, we integrated a publicly available oxidative stress gene signature with patient survival data from the Cancer Genome Atlas database. Overall, we found several genes statistically significant associated with poor prognosis in the examined six tumor types. Among them, FoxM1 and thioredoxin reductase1 expression showed the same pattern in four out of six cancers, suggesting their specific critical role in cancer-related oxidative stress adaptation. Our analysis also unveiled an enriched cellular network, highlighting specific pathways, in which many genes are strictly correlated. Finally, we discussed novel findings on the correlation between oxidative stress and cancer stem cells in order to define those pathways to be prioritized in drug development.
Collapse
|
20
|
Maekawa A, Kohashi K, Kuda M, Iura K, Ishii T, Endo M, Nakatsura T, Iwamoto Y, Oda Y. Prognostic significance of FOXM1 expression and antitumor effect of FOXM1 inhibition in synovial sarcomas. BMC Cancer 2016; 16:511. [PMID: 27439614 PMCID: PMC4955131 DOI: 10.1186/s12885-016-2542-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 07/08/2016] [Indexed: 12/25/2022] Open
Abstract
Background Synovial sarcoma (SS) is a soft tissue sarcoma of unknown histogenesis. Most metastatic or unresectable cases are incurable. Novel antitumor agents and precise prognostication are needed for SS patients. The protein forkhead box M1 (FOXM1), which belongs to the FOX family of transcription factors, is considered to be an independent predictor of poor survival in many cancers and sarcomas, but the prognostic implications and oncogenic roles of FOXM1 in SS are poorly understood. Here we examined the correlation between FOXM1 expression and clinicopathologic and prognostic factors, and we investigated the efficacy of FOXM1 target therapy in SS cases. Methods Immunohistochemical study of 106 tumor specimens was conducted to evaluate their immunohistochemical expression of FOXM1. An in vitro study examined the antitumor effect of the FOXM1 inhibitor thiostrepton and small interference RNA (siRNA) on two SS cell lines. We also assessed the efficacy of the combined use of doxorubicin (DOX) and thiostrepton. Results Univariate and multivariate analyses revealed that FOXM1 expression was associated with poor prognosis in SS. The cDNA microarray analysis using clinical samples revealed that the expression of cell cycle-associated genes was correlated with FOXM1 expression. FOXM1 inhibition by thiostrepton showed significant antitumor activity on the SS cell lines in vitro. FOXM1 interruption by siRNA increased the chemosensitivity for DOX in both SS cell lines. Conclusion FOXM1 expression is a novel biomarker, and its inhibition is a potential treatment option for SS. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2542-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Akira Maekawa
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masaaki Kuda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kunio Iura
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takeaki Ishii
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Makoto Endo
- Departments of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yukihide Iwamoto
- Departments of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
21
|
Sun Q, Dong M, Chen Y, Zhang J, Qiao J, Guo X. Prognostic significance of FoxM1 expression in non-small cell lung cancer. J Thorac Dis 2016; 8:1269-73. [PMID: 27293846 DOI: 10.21037/jtd.2016.04.13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Various studies examined the relationship between FoxM1 overexpression with the clinical outcome in patients with non-small cell lung cancer (NSCLC), but yielded conflicting results. METHODS Electronic databases updated to Jan 01, 2015 were searched to find relevant studies. A meta-analysis was conducted with eligible studies which quantitatively evaluated the relationship between FoxM1 overexpression and survival of patients with NSCLC Survival data were aggregated and quantitatively analyzed. RESULTS We performed a meta-analysis of seven studies (n=860 patients) that evaluated the correlation between FoxM1 overexpression and survival in patients with NSCLC. Combined hazard ratios suggested that FoxM1 overexpression was associated with poor prognosis of overall survival (OS) (HR =1.73, 95% CI: 1.32-2.14) in patients with NSCLC. CONCLUSIONS FoxM1 overexpression indicates a poor prognosis for patients with NSCLC.
Collapse
Affiliation(s)
- Qing Sun
- Department of Oncology, Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Min Dong
- Department of Oncology, Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Yujuan Chen
- Department of Oncology, Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Jiawei Zhang
- Department of Oncology, Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Jinpeng Qiao
- Department of Oncology, Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Xuedan Guo
- Department of Oncology, Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi 214002, China
| |
Collapse
|
22
|
Ito T, Kohashi K, Yamada Y, Iwasaki T, Maekawa A, Kuda M, Hoshina D, Abe R, Furue M, Oda Y. Prognostic Significance of Forkhead Box M1 (FOXM1) Expression and Antitumor Effect of FOXM1 Inhibition in Angiosarcoma. J Cancer 2016; 7:823-30. [PMID: 27162541 PMCID: PMC4860799 DOI: 10.7150/jca.14461] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/15/2016] [Indexed: 01/06/2023] Open
Abstract
Background: The prognosis of angiosarcoma is poor and a novel treatment option for the disease is desired. The aim of this study was to investigate the prognostic significance of Forkhead box M1 (FOXM1), a transcription factor that regulates cell-cycle progression and various crucial processes in tumor progression, and its potential as a new therapeutic target. Methods: We investigated 125 angiosarcoma clinical samples (94 primary lesions and 31 metastatic lesions in 94 patients) and a human angiosarcoma cell line (HAMON) using immunohistochemical staining and molecular biological approaches. FOXM1 expression in angiosarcoma samples was also compared with that in Kaposi's sarcomas (n = 13), epithelioid hemangioendotheliomas (n = 13) and benign hemangiomas (n = 10). Results: Patients with FOXM1-overexpressing angiosarcoma had significantly shorter survival (both for disease-specific survival [DSS] and event-free survival [EFS]) than other patients (5-year DSS, 23.5% vs. 47.1%, P = 0.013; and 5-year EFS, 5.5% vs. 28.7%, P = 0.004). FOXM1 overexpression was also an independent prognostic factor for both DSS and EFS in Cox multivariate analyses (hazard ratio [HR] 2.84, 95% confidence interval [CI] 1.10-5.81, P = 0.039; and HR 4.16, 95%CI 2.03-8.67, P = 0.0001, respectively). FOXM1 inhibition using both small interfering RNA and a specific inhibitor (thiostrepton) suppressed cell proliferation of the angiosarcoma cell line. Furthermore, FOXM1 inhibition improved the chemosensitivity to docetaxel in vitro. Conclusions: FOXM1 inhibition may be a potential therapeutic option for angiosarcoma.
Collapse
Affiliation(s)
- Takamichi Ito
- 1. Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan;; 2. Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichi Kohashi
- 1. Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Yamada
- 1. Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Iwasaki
- 1. Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Maekawa
- 1. Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaaki Kuda
- 1. Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daichi Hoshina
- 3. Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Riichiro Abe
- 3. Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masutaka Furue
- 2. Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- 1. Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Tahmasbpour E, Ghanei M, Qazvini A, Vahedi E, Panahi Y. Gene expression profile of oxidative stress and antioxidant defense in lung tissue of patients exposed to sulfur mustard. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 800-801:12-21. [PMID: 27085470 DOI: 10.1016/j.mrgentox.2016.03.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/07/2016] [Accepted: 03/10/2016] [Indexed: 01/13/2023]
Abstract
Sulfur mustard (SM) is a potent alkylating agent that targets several organs, especially lung tissue. Although pathological effects of SM on mustard lung have been widely considered, molecular and cellular mechanisms for these pathologies are poorly understood. We investigated changes in expression of genes related to oxidative stress (OS) and antioxidant defense caused by SM in lung tissue of patients. We performed gene expression profiling of OS and antioxidant defense in lung tissue samples from healthy controls (n=5) and SM-exposed patients (n=6). Changes in gene expression were measured using a 96-well RT(2) Profiler ™PCR Array: Human Oxidative Stress and Antioxidant Defense, which arrayed 84 genes functionally involved in cellular OS response. 47 (55.95%) genes were found to be significantly upregulated in patients with mustard lung compared with controls (p<0.05), whereas 7 (8.33%) genes were significantly downregulated (p<0.05). Among the most upregulated genes were OS responsive-1 (OXSR1), forkhead box M1 (FOXM1), and glutathione peroxidase-2 (GPX2), while metallothionein-3 (MT3) and glutathione reductase (GSR) were the most downregulated genes. Expression of hypoxia-induced genes (CYGB and MB), antioxidants and reactive oxygen species (ROS)-producing genes were significantly altered, suggesting an increased oxidative damage in mustard lungs. Mustard lungs were characterized by hypoxia, massive production of ROS, OS, disruption of epithelial cells, surfactant dysfunction, as well as increased risk of lung cancer and pulmonary fibrosis. Oxidative stress induced by ROS is the major mechanism for direct effect of SM exposure on respiratory system. Antioxidant treatment may improve the main features of mustard lungs.
Collapse
Affiliation(s)
- Eisa Tahmasbpour
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran
| | - Ali Qazvini
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran; Department of Pulmonology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ensieh Vahedi
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran; Department of Pulmonology, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Yunes Panahi
- Chemical Injuries Research Center, Baqiatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Maekawa A, Kohashi K, Setsu N, Kuda M, Iura K, Ishii T, Matsunobu T, Nakatsura T, Iwamoto Y, Oda Y. Expression of Forkhead box M1 in soft tissue leiomyosarcoma: Clinicopathologic and in vitro study using a newly established cell line. Cancer Sci 2016; 107:95-102. [PMID: 26560505 PMCID: PMC4724818 DOI: 10.1111/cas.12846] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 02/07/2023] Open
Abstract
Leiomyosarcoma (LMS) of soft tissue is a sarcoma with smooth‐muscle differentiation, and conventional chemotherapy does not improve its outcome. The application of novel antitumor agents and precise prognostication has been demanded. The expression of the protein Forkhead box M1 (FOXM1), a member of the FOX family, is considered an independent predictor of poor survival in many cancers and sarcomas. However, the expression status of FOXM1 in LMS is poorly understood. The purposes of this study were to examine the correlation between the expression of FOXM1 and clinicopathologic or prognostic factors and to clarify the efficacy of FOXM1 target therapy in LMS. We evaluated the immunohistochemical expressions of FOXM1 using 123 LMS tumor specimens. Univariate and multivariate survival analyses revealed that FOXM1 expression was associated with poor prognosis in LMS. An in vitro study was then carried out to examine the antitumor effect of a FOXM1 inhibitor (thiostrepton) and siRNA on a novel LMS cell line, TC616. We also assessed the efficacy of the combined use of doxorubicin and thiostrepton. Thiostrepton showed dose‐dependent antitumor activity and TC616 cells treated with the combination of thiostrepton and doxorubicin showed lower proliferation compared to those treated with either drug individually. FOXM1 interruption by siRNA decreased cell proliferation and increased chemosensitivity. In conclusion, FOXM1 has potential to be a therapeutic target for LMS.
Collapse
Affiliation(s)
- Akira Maekawa
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nokitaka Setsu
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaaki Kuda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kunio Iura
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeaki Ishii
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoya Matsunobu
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yukihide Iwamoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
25
|
Ito T, Kohashi K, Yamada Y, Maekawa A, Kuda M, Furue M, Oda Y. Prognostic significance of forkhead box M1 (FoxM1) expression and antitumour effect of FoxM1 inhibition in melanoma. Histopathology 2016; 69:63-71. [PMID: 26619071 DOI: 10.1111/his.12909] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/25/2015] [Indexed: 01/01/2023]
Abstract
AIMS Forkhead box M1 (FoxM1) is a transcription factor that regulates cell-cycle progression and tumour progression, but limited information is available regarding its clinical significance in melanoma. The aim of this study was to investigate the potency of FoxM1 as a therapeutic target in melanoma. METHODS AND RESULTS We investigated 60 melanoma clinical samples and a melanoma WM266-4 cell line using immunohistochemical staining and molecular biological approaches. Patients with a FoxM1-overexpressing melanoma had significantly shorter survival [both for melanoma-specific survival (MSS) and disease-free survival (DFS)] than the other patients (P < 0.001, respectively). The FoxM1 overexpression was also an adverse prognostic factor for both MSS and DFS on the Cox multivariate analyses [hazard ratio (HR): 3.96, 95% confidence interval (CI): 1.12-14.27, P = 0.032; HR: 3.21, 95% CI: 1.08-9.67, P = 0.037, respectively). FoxM1 inhibition using siRNA and an inhibitor (thiostrepton) each suppressed the cell proliferation of the melanoma cell line. Furthermore, FoxM1 inhibition improved chemosensitivity to dacarbazine, whereas it reduced cell migration and invasion. These results suggest that FoxM1 plays important roles in tumour progression and the chemoresistance of melanoma. CONCLUSION We have shown the prognostic impact of FoxM1 on melanoma patients. FoxM1 inhibition may be a potential therapeutic option for advanced melanoma.
Collapse
Affiliation(s)
- Takamichi Ito
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Maekawa
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaaki Kuda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
26
|
Prognostic Value of FOXM1 in Patients with Malignant Solid Tumor: A Meta-Analysis and System Review. DISEASE MARKERS 2015; 2015:352478. [PMID: 26451068 PMCID: PMC4584221 DOI: 10.1155/2015/352478] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/23/2015] [Indexed: 01/18/2023]
Abstract
Forkhead box M1 (FOXM1), a member of the Fox transcription factors family, was closely related with cell cycle. FOXM1 played an important role in MST and prompted a poor prognosis for MST patients. However, there were also some studies revealing no significant association between the FOXM1 expression and prognosis of patients. Therefore, we conducted meta-analysis to investigate whether the expression of FOXM1 was associated with MST prognosis. We collected 36 relevant studies through PubMed database and obtained research data of 4946 patients. Stata 12.0 was used to express the results as hazard ratio (HR) for time-to-event outcomes with 95% confidence intervals (95% CI). It was shown that overexpression of FOXM1 was relevant to worse survival of MST patients (HR = 1.99, 95% CI = 1.79–2.21, P < 0.001; I2 = 26.4%, Ph = 0.076). Subgroup analysis suggested that overexpression of FOXM1 in breast cancer (BC), gastric cancer (GC), hepatocellular carcinoma (HCC), pancreatic ductal adenocarcinoma (PDA), and non-small-cell lung cancer (NSCLC) all predicted a worse survival (P < 0.05), in addition to ovarian cancer (OC) (P = 0.084). In conclusion, our research indicated that overexpression of FOXM1 was to the disadvantage of the prognosis for majority of MST and therefore can be used as an evaluation index of prognosis.
Collapse
|
27
|
Ahn H, Sim J, Abdul R, Chung MS, Paik SS, Oh YH, Park CK, Jang K. Increased expression of forkhead box M1 is associated with aggressive phenotype and poor prognosis in estrogen receptor-positive breast cancer. J Korean Med Sci 2015; 30:390-7. [PMID: 25829806 PMCID: PMC4366959 DOI: 10.3346/jkms.2015.30.4.390] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 11/12/2014] [Indexed: 11/20/2022] Open
Abstract
Fox transcription factors play a critical role in the regulation of a variety of biological processes. While FoxM1 behaves like the oncogenic transcription factor, FoxO3a is known as a tumor suppressor by inhibiting FoxM1. This study aimed to investigate the clinicopathological significance of FoxM1 and FoxO3a expression in breast cancer. Expression of FoxM1 and FoxO3a were analyzed by immunohistochemical staining on tissue microarray sections from 236 breast cancer patients, and correlated with various clinicopathological characteristics. Overexpression of FoxM1 correlated with adverse clinicopathological features, such as larger tumor size, lymph node metastasis, advanced tumor stage, and lymphovascular invasion. The Kaplan-Meier survival curves revealed no prognostic significance of FoxM1 expression. However, in subgroup analyses with patients of estrogen receptor (ER) positive breast cancers, FoxM1 overexpression associated with poor disease free and overall survival. No association was found between FoxO3a and FoxM1 expression. Regarding clinicopathological variables, the only association between histologic grade and FoxO3a was observed. In conclusion, FoxM1 overexpression was significantly associated with aggressive phenotypes and poor prognosis of ER-positive breast cancer. These findings suggest the possible role of FoxM1 as a prognostic biomarker and putative target of anti-cancer therapy.
Collapse
Affiliation(s)
- Hyein Ahn
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Jongmin Sim
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Rehman Abdul
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Min Sung Chung
- Department of Surgery, College of Medicine, Hanyang University, Seoul, Korea
| | - Seung Sam Paik
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Young-Ha Oh
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Chan Kum Park
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Kiseok Jang
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| |
Collapse
|
28
|
Xu XS, Miao RC, Wan Y, Zhang LQ, Qu K, Liu C. FoxM1 as a Novel Therapeutic Target for Cancer Drug Therapy. Asian Pac J Cancer Prev 2015; 16:23-9. [DOI: 10.7314/apjcp.2015.16.1.23] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
29
|
Lindskog C, Edlund K, Mattsson JSM, Micke P. Immunohistochemistry-based prognostic biomarkers in NSCLC: novel findings on the road to clinical use? Expert Rev Mol Diagn 2015; 15:471-90. [DOI: 10.1586/14737159.2015.1002772] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Wei P, Zhang N, Wang Y, Li D, Wang L, Sun X, Shen C, Yang Y, Zhou X, Du X. FOXM1 promotes lung adenocarcinoma invasion and metastasis by upregulating SNAIL. Int J Biol Sci 2015; 11:186-98. [PMID: 25561901 PMCID: PMC4279094 DOI: 10.7150/ijbs.10634] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/17/2014] [Indexed: 01/31/2023] Open
Abstract
The forkhead box M1 (FOXM1) transcription factor is one of the key genes inducing tumor invasion and metastasis by an unknown mechanism. In this study, we set out to investigate the effects of FOXM1 overexpression on metastatic human lung adenocarcinoma and the underlying mechanism. FOXM1 expression was analyzed in 78 frozen lung adenocarcinoma tissue samples using an Affymetrix microarray and a 155-paraffin-embedded lung adenocarcinoma tissue microarray with immunohistochemical detection. FOXM1 was found to be overexpressed in lung adenocarcinoma, particularly in metastatic patients, compared to non-metastatic patients. Knockdown of FOXM1 by a specific siRNA significantly suppressed EMT progression, migration and invasion of lung adenocarcinoma cells in vitro, and tumor growth and metastasis in vivo, whereas restored expression of FOXM1 had the opposite effect. FOXM1 binds directly to the SNAIL promoter through two specific binding sites and constitutively transactivates it. Collectively, our findings indicate that FOXM1 may play an important role in advancing lung adenocarcinoma progression. Aberrant FOXM1 expression directly and constitutively activates SNAIL, thereby promoting lung adenocarcinoma metastasis. Inhibition of FOXM1-SNAIL signaling may present an ideal target for future treatment.
Collapse
Affiliation(s)
- Ping Wei
- 1. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 2. Institute of Pathology, Fudan University, Shanghai 200032, China ; 3. Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Nu Zhang
- 7. Department of neurosurgery, 1st affiliated hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yiqin Wang
- 1. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 2. Institute of Pathology, Fudan University, Shanghai 200032, China ; 4. Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China. ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Dawei Li
- 5. Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lisha Wang
- 1. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 2. Institute of Pathology, Fudan University, Shanghai 200032, China ; 4. Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China. ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiangjie Sun
- 1. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 2. Institute of Pathology, Fudan University, Shanghai 200032, China ; 4. Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China. ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chen Shen
- 1. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 2. Institute of Pathology, Fudan University, Shanghai 200032, China ; 4. Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China. ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yusi Yang
- 1. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 2. Institute of Pathology, Fudan University, Shanghai 200032, China ; 4. Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China. ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoyan Zhou
- 1. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 2. Institute of Pathology, Fudan University, Shanghai 200032, China ; 4. Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China. ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiang Du
- 1. Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China ; 2. Institute of Pathology, Fudan University, Shanghai 200032, China ; 4. Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China. ; 6. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
31
|
Chen PM, Cheng YW, Wang YC, Wu TC, Chen CY, Lee H. Up-regulation of FOXM1 by E6 oncoprotein through the MZF1/NKX2-1 axis is required for human papillomavirus-associated tumorigenesis. Neoplasia 2014; 16:961-71. [PMID: 25425970 PMCID: PMC4240922 DOI: 10.1016/j.neo.2014.09.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/20/2014] [Accepted: 09/22/2014] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Foxhead box M1 (FOXM1) expression has been shown to be linked with human papillomavirus (HPV) 16/18-infected cervical cancer. However, the mechanism underlying the induction of FOXM1 in HPV 16/18-infected cancers remains elusive. EXPERIMENTAL DESIGN The mechanistic actions of FOXM1 induced by the E6/NKX2-1 axis in tumor aggressiveness were elucidated in cellular and animal models. The prognostic value of FOXM1 for overall survival (OS) and relapse-free survival (RFS) in HPV-positive oral and lung cancers was assessed using Kaplan-Meier and Cox regression models. RESULTS Herein, FOXM1 expression is upregulated by E6-mediated NKX2-1 in HPV-positive cervical, oral, and lung cancer cells. Induction of FOXM1 by E6 through the MZF1/NKX2-1 axis is responsible for HPV-mediated soft agar growth, invasiveness, and stemness through activating Wnt/β-catenin signaling pathway. In a nude mice model, metastatic lung tumor nodules in HPV 18 E6-positive GNM or HPV 16 E6-positive TL-1-injected nude mice were markedly decreased in both cell types with E6 knockdown, FOXM1 knockdown, or treatment with FOXM1 inhibitor (thiostrepton). Among the four subgroup patients, the worst FOXM1 prognostic value for OS and RFS was observed in HPV 16/18-positive patients with tumors with high-expressing FOXM1. CONCLUSIONS Induction of FOXM1 by E6 oncoprotein through the MZF1/NKX2-1 axis may be responsible for HPV 16/18-mediated tumor progression and poor outcomes in HPV-positive patients.
Collapse
Affiliation(s)
- Po-Ming Chen
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Ya-Wen Cheng
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chen Wang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan ; Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Tzu-Chin Wu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan ; Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Yi Chen
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
32
|
Abstract
BACKGROUND The forkhead box M1 (FOXM1) transcription factor plays an important role in the metastases of many cancers. Down-regulation of FOXM1 by its inhibitor, thiostrepton, can inhibit the metastatic potential of some cancers; however, there are few studies regarding the functional significance of FOXM1 and thiostrepton in the metastases of nasopharyngeal carcinoma (NPC) and the underlying mechanism. METHODS Expression of FOXM1 in NPC, normal nasopharyngeal tissues, a NPC cell line (C666-1), and a nasopharyngeal epithelial cell line (NP69) was investigated by immunohistochemical staining, qRT-PCR, and Western blot. The correlation between FOXM1 expression and the clinical characteristics of patients was analyzed. Moreover, the effects of thiostrepton on expression of FOXM1 in C666-1 and NP69 cells, and the invasion and migration ability of C666-1 cells were examined. The expressions of MMP-2, MMP-9, fascin-1, ezrin, and paxillin were determined after treatment with thiostrepton. RESULTS FOXM1 was overexpressed in NPC and C666-1 cells compared with normal nasopharyngeal tissues and NP69 cells. Overexpression of FOXM1 was associated with lymph node metastasis and advanced tumor stage. Moreover, thiostrepton inhibited expression of FOXM1 in C666-1 cells in a dose-dependent manner, but had a minimal effect on NP69 cells. Thiostrepton inhibited the migration and invasion ability of C666-1 cells by down-regulating the expression of MMP-2, MMP-9, fascin-1, and paxillin. CONCLUSIONS Overexpression of FOXM1 is associated with metastases of NPC patients. Thiostrepton inhibits the metastatic ability of NPC cells by down-regulating the expression of FOXM1, MMP-2, MMP-9, fascin-1, and paxillin.
Collapse
Affiliation(s)
- Lizhu Jiang
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, 400016 Chongqing, P.R. China
| | - Peng Wang
- Department of Orthopedics, Dazu District People’s Hospital, 402360 Chongqing, P.R. China
| | - Hongyan Chen
- Department of Otolaryngology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, 400016 Chongqing, P.R. China
| |
Collapse
|
33
|
Kong FF, Qu ZQ, Yuan HH, Wang JY, Zhao M, Guo YH, Shi J, Gong XD, Zhu YL, Liu F, Zhang WY, Jiang B. Overexpression of FOXM1 is associated with EMT and is a predictor of poor prognosis in non-small cell lung cancer. Oncol Rep 2014; 31:2660-8. [PMID: 24715097 DOI: 10.3892/or.2014.3129] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/17/2014] [Indexed: 01/06/2023] Open
Abstract
Forkhead box M1 (FOXM1), a member of the Fox family of transcriptional factors, is considered to be an independent predictor of poor survival in many solid cancers. However, the underlying mechanism is not yet clear. The aim of the present study was to investigate the clinical significance of the correlation between FOXM1 and epithelial-mesenchymal transition (EMT) in non-small cell lung carcinoma and the possible mechanism responsible for FOXM1-induced EMT and metastasis. In the present study, expression levels of FOXM1 and EMT indicator proteins were determined by tissue microarray (TMA) and immunohistochemical staining, western blotting and reverse transcription-PCR (RT-PCR). Other cellular and molecular approaches including gene transfection, small interfering RNA (siRNA), and migration and invasion assays were utilized. Our results demonstrated that FOXM1 overexpression was statistically significantly associated with a higher TNM stage (p=0.036), lymph node metastasis (p=0.009) and a positive smoking history of the patients (p=0.044). Additionally, high expression of FOXM1 correlated with loss of E-cadherin expression (p<0.001) and anomalous immunopositivity of Vimentin (p=0.002). Moreover, patient survival analysis demonstrated that high expression of FOXM1 (p=0.043) and the presence of lymph node metastasis (p=0.042) were independent prognostic factors for non-small cell lung cancer (NSCLC). Furthermore, various in vitro experiments indicated that overexpression or knockdown of FOXM1 expression altered EMT through activation or inhibition of the AKT/p70S6K signaling pathway. Collectively, the results suggest that FOXM1 may be used as a prognostic indicator for patients with NSCLC and promotes metastasis by inducing EMT of lung cancer cells through activation of the AKT/p70S6K pathway. Therefore, we suggest that FOXM1 may be a potential target for lung cancer therapy.
Collapse
Affiliation(s)
- Fei-Fei Kong
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Zeng-Qiang Qu
- Department of Interventional Radiology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Hai-Hua Yuan
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Jiong-Yi Wang
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Mei Zhao
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Yue-Hui Guo
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Jing Shi
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Xiao-Di Gong
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - You-Long Zhu
- Department of General Surgery, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Feng Liu
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Wen-Ying Zhang
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| | - Bin Jiang
- Department of Oncology, No. 3 People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine (SJTU-SM), Shanghai 201900, P.R. China
| |
Collapse
|
34
|
WANG YIRU, YAO BINWEI, WANG YU, ZHANG MINGBO, FU SHUAI, GAO HANJING, PENG RUIYUN, ZHANG LINGQIANG, TANG JIE. Increased FoxM1 expression is a target for metformin in the suppression of EMT in prostate cancer. Int J Mol Med 2014; 33:1514-22. [DOI: 10.3892/ijmm.2014.1707] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/07/2014] [Indexed: 11/05/2022] Open
|
35
|
Huang C, Du J, Xie K. FOXM1 and its oncogenic signaling in pancreatic cancer pathogenesis. Biochim Biophys Acta Rev Cancer 2014; 1845:104-16. [PMID: 24418574 DOI: 10.1016/j.bbcan.2014.01.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 12/30/2013] [Accepted: 01/03/2014] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer is a devastating disease with an overall 5-year survival rate less than 5%. Multiple signaling pathways are implicated in the pathogenesis of pancreatic cancer, such as Wnt/β-catenin, Notch, Hedgehog, hypoxia-inducible factor, signal transducer and activator of transcription, specificity proteins/Krüppel-like factors, and Forkhead box (FOX). Recently, increasing evidence has demonstrated that the transcription factor FOXM1 plays important roles in the initiation, progression, and metastasis of a variety of human tumors, including pancreatic cancer. In this review, we focus on the current understanding of the molecular pathogenesis of pancreatic cancer with a special focus on the function and regulation of FOXM1 and rationale for FOXM1 as a novel molecular target for pancreatic cancer prevention and treatment.
Collapse
Affiliation(s)
- Chen Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, People's Republic of China; Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Jiawei Du
- Department of Laboratory Medicine, Zhenjiang Second People's Hospital, Jiangsu University College of Medicine, Zhenjiang, People's Republic of China
| | - Keping Xie
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
36
|
Abstract
BACKGROUND An understanding of the activated protein signaling architecture in non-small-cell lung cancer (NSCLC) is of critical importance to the development of new therapeutic approaches and identification of predictive and prognostic biomarkers for patient stratification. METHODS We used reverse-phase protein microarrays to map the activated protein signaling networks of 47 NSCLC tumors, 28 of which were node negative, which were subjected to tumor cellular enrichment using laser capture microdissection. The phosphorylation/cleavage levels of 111 key signaling proteins and total levels of 17 proteins were measured for broadscale signaling analysis. RESULTS Pathway activation mapping of NSCLC revealed distinct subgroups composed of epidermal growth factor receptor (ERBB1), v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 (ERBB2), v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (ERBB3), v-erb-a erythroblastic leukemia viral oncogene homolog 4 (ERBB4), v-akt murine thymoma viral oncogene homolog 1- mammalian target of rapamycin (AKT-mTOR), protein kinase, AMP-activated, alpha 2 catalytic subunit (AMPK), and autophagy-related signaling, along with transforming growth factor-beta-signaling protein 1 (SMAD), insulin-line growth factor receptor (IGFR), rearranged during transfection proto-oncogene (RET), and activated CDC42-associated kinase (ACK) activation. Investigation of epidermal growth factor receptor (EGFR)-driven signaling identified a unique cohort of tumors with low EGFR protein expression yet high relative levels of phosphorylated EGFR and high EGFR total protein with low relative levels of phosphorylation. Last, mapping analysis of patients with NSCLC with N0 disease revealed a pilot pathway activation signature composed of linked epidermal growth factor receptor family (HER)-AMPK-AKT-mTOR signaling network along with focal adhesion kinase- LIM domain kinase-1 (FAK-LIMK) and janus kinase (JAK)-signal transducers and activators of transcription (STAT) pathways that correlated with short-term survival and aggressive disease. CONCLUSIONS Functional protein pathway activation mapping of NSCLC reveals distinct activation subgroups that are underpinned by important therapeutic targets and that patients with early-stage node negative disease and poor prognosis may be identified by activation of defined, biochemically linked protein signaling events. Such findings, if confirmed in larger study sets, could help select and stratify patients for personalized targeted therapies.
Collapse
|
37
|
Feng Y, Wang L, Zeng J, Shen L, Liang X, Yu H, Liu S, Liu Z, Sun Y, Li W, Chen C, Jia J. FoxM1 is overexpressed in Helicobacter pylori-induced gastric carcinogenesis and is negatively regulated by miR-370. Mol Cancer Res 2013; 11:834-44. [PMID: 23576572 DOI: 10.1158/1541-7786.mcr-13-0007] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Helicobacter pylori (H. pylori) infections are strongly implicated in human gastric mucosa-associated diseases. Forkhead box M1 (FoxM1), a key positive regulator of cell proliferation, is overexpressed in gastric cancer. MicroRNAs are important post-transcriptional regulators of gene expression. In this study, the effects of H. pylori infection on FoxM1 expression and possible mechanisms of carcinogenesis were explored. The expression of FoxM1 was gradually increased in human gastric specimens from inflammation to cancer. FoxM1 upregulation was time- and concentration-dependent in gastric epithelial-derived cell lines infected with H. pylori. CagA, a key virulence factor of H. pylori, was associated with increased FoxM1 expression. H. pylori and CagA inhibited the expression of p27(Kip1) (CDKN1B) and promoted cell proliferation by upregulating FoxM1. The expression of miR-370 was decreased in human gastritis and gastric cancer. FoxM1 was directly downregulated by miR-370 in gastric cell lines. H. pylori and CagA inhibited miR-370 expression, which led to overexpression of FoxM1 and cell proliferation. Furthermore, the overexpression of FoxM1 and reduced expression of miR-370 was confirmed in H. pylori-infected C57BL/6J mice. H. pylori infection and CagA upregulated FoxM1 expression, dependent on miR-370, altered the expression of p27(Kip1), and promoted proliferation in gastric cells. IMPLICATIONS These findings delineate the mechanisms governing FoxM1 regulation and the role of H. pylori in the process of gastric carcinogenesis.
Collapse
Affiliation(s)
- Yimin Feng
- Department of Microbiology/Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan 250012, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Xu N, Jia D, Chen W, Wang H, Liu F, Ge H, Zhu X, Song Y, Zhang X, Zhang D, Ge D, Bai C. FoxM1 is associated with poor prognosis of non-small cell lung cancer patients through promoting tumor metastasis. PLoS One 2013; 8:e59412. [PMID: 23536876 PMCID: PMC3607616 DOI: 10.1371/journal.pone.0059412] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 02/14/2013] [Indexed: 02/06/2023] Open
Abstract
Background FoxM1 has been reported to be important in initiation and progression of various tumors. However, whether FoxM1 has any indication for prognosis in non-small cell lung cancer patients remains unclear. Methodology/Principal Findings In this study, FoxM1 expression in tumor cells was examined first by immunohistochemistry in 175 NSCLC specimens, the result of which showed that FoxM1 overexpression was significantly associated with positive smoking status (P = 0.001), poorer tissue differentiation (P = 0.0052), higher TNM stage (P<0.0001), lymph node metastasis (P<0.0001), advanced tumor stage (P<0.0001), and poorer prognosis (P<0.0001). Multivariable analysis showed that FoxM1 expression increased the hazard of death (hazard ratio, 1.899; 95% CI, 1.016–3.551). Furthermore, by various in vitro and in vivo experiments, we showed that targeted knockdown of FoxM1 expression could inhibit the migratory and invasive abilities of NSCLC cells, whereas enforced expression of FoxM1 could increased the invasion and migration of NSCLC cells. Finally, we found that one of the cellular mechanisms by which FoxM1 promotes tumor metastasis is through inducing epithelial-mesenchymal transition (EMT) program. Conclusions These results suggested that FoxM1 overexpression in tumor tissues is significantly associated with the poor prognosis of NSCLC patients through promoting tumor metastasis.
Collapse
Affiliation(s)
- Nuo Xu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Deshui Jia
- Shanghai Medical School, Fudan University, Shanghai, China
| | - Wenfeng Chen
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Hao Wang
- Department of thoracology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fanglei Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haiyan Ge
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaodan Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - David Zhang
- Molecular Pathology Division, Department of Pathology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Di Ge
- Department of thoracology, Zhongshan Hospital, Fudan University, Shanghai, China
- * E-mail: (CXB); (DG)
| | - Chunxue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- * E-mail: (CXB); (DG)
| |
Collapse
|
39
|
Wierstra I. FOXM1 (Forkhead box M1) in tumorigenesis: overexpression in human cancer, implication in tumorigenesis, oncogenic functions, tumor-suppressive properties, and target of anticancer therapy. Adv Cancer Res 2013; 119:191-419. [PMID: 23870513 DOI: 10.1016/b978-0-12-407190-2.00016-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor and is also intimately involved in tumorigenesis. FOXM1 stimulates cell proliferation and cell cycle progression by promoting the entry into S-phase and M-phase. Additionally, FOXM1 is required for proper execution of mitosis. In accordance with its role in stimulation of cell proliferation, FOXM1 exhibits a proliferation-specific expression pattern and its expression is regulated by proliferation and anti-proliferation signals as well as by proto-oncoproteins and tumor suppressors. Since these factors are often mutated, overexpressed, or lost in human cancer, the normal control of the foxm1 expression by them provides the basis for deregulated FOXM1 expression in tumors. Accordingly, FOXM1 is overexpressed in many types of human cancer. FOXM1 is intimately involved in tumorigenesis, because it contributes to oncogenic transformation and participates in tumor initiation, growth, and progression, including positive effects on angiogenesis, migration, invasion, epithelial-mesenchymal transition, metastasis, recruitment of tumor-associated macrophages, tumor-associated lung inflammation, self-renewal capacity of cancer cells, prevention of premature cellular senescence, and chemotherapeutic drug resistance. However, in the context of urethane-induced lung tumorigenesis, FOXM1 has an unexpected tumor suppressor role in endothelial cells because it limits pulmonary inflammation and canonical Wnt signaling in epithelial lung cells, thereby restricting carcinogenesis. Accordingly, FOXM1 plays a role in homologous recombination repair of DNA double-strand breaks and maintenance of genomic stability, that is, prevention of polyploidy and aneuploidy. The implication of FOXM1 in tumorigenesis makes it an attractive target for anticancer therapy, and several antitumor drugs have been reported to decrease FOXM1 expression.
Collapse
|
40
|
Increased expression of forkhead box M1 protein is associated with poor prognosis in clear cell renal cell carcinoma. Med Oncol 2012; 30:346. [PMID: 23263830 DOI: 10.1007/s12032-012-0346-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 08/20/2012] [Indexed: 10/27/2022]
Abstract
Forkhead box protein M1 (FoxM1) is crucial in the regulation of various biological processes, including cell proliferation, organogenesis, and angiogenesis. Overexpression of FoxM1 is associated with carcinogenesis. In this study, immunohistochemistry was carried out to examine FoxM1 expression in clear cell renal cell carcinoma (ccRCC), and these data were examined for correlation with clinicopathological parameters and prognosis. FoxM1 protein had high expression in 37 of 87 cases of ccRCC (42.5 %), which was significantly higher than in normal tissues, and FoxM1 overexpression was significantly associated with tumor stage (P = 0.005) and recurrence (P = 0.027). The Kaplan-Meier survival analysis demonstrated that FoxM1 expression was significantly associated with shorter recurrence-free survival and overall survival (P = 0.007 and P = 0.008, respectively). Multivariate analysis further demonstrated that FoxM1 was an independent prognostic factor for patients with ccRCC. So FoxM1 might be a potential molecular marker to predict the prognosis of patients with ccRCC.
Collapse
|
41
|
FOXM1 promotes tumor cell invasion and correlates with poor prognosis in early-stage cervical cancer. Gynecol Oncol 2012; 127:601-10. [DOI: 10.1016/j.ygyno.2012.08.036] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 08/24/2012] [Accepted: 08/25/2012] [Indexed: 12/12/2022]
|
42
|
Forkhead box M1 (FoxM1) gene is a new STAT3 transcriptional factor target and is essential for proliferation, survival and DNA repair of K562 cell line. PLoS One 2012; 7:e48160. [PMID: 23110199 PMCID: PMC3480485 DOI: 10.1371/journal.pone.0048160] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 09/21/2012] [Indexed: 01/16/2023] Open
Abstract
The forkhead box (Fox) M1 gene belongs to a superfamily of evolutionarily conserved transcriptional regulators that are involved in a wide range of biological processes, and its deregulation has been implicated in cancer survival, proliferation and chemotherapy resistance. However, the role of FoxM1, the signaling involved in its activation and its role in leukemia are poorly known. Here, we demonstrate by gene promoter analysis, Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assays that FoxM1 is a new target of the STAT3 transcriptional activator. Additionally, FoxM1 is transcriptionally dependent on STAT3 signaling activation. Furthermore, we verified that FoxM1 is crucial for K562 cell proliferation, cell cycle checkpoints and viability and could be related to chemotherapeutic resistance. By microarray analysis, we determined the signaling pathways related to FoxM1 expression and its role in DNA repair using K562 cells. Our results revealed new signaling involved in FoxM1 expression and its role in leukemic cells that elucidate cellular mechanisms associated with the development of leukemia and disease progression.
Collapse
|
43
|
Halasi M, Gartel AL. Targeting FOXM1 in cancer. Biochem Pharmacol 2012; 85:644-652. [PMID: 23103567 DOI: 10.1016/j.bcp.2012.10.013] [Citation(s) in RCA: 358] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/15/2012] [Accepted: 10/17/2012] [Indexed: 11/28/2022]
Abstract
Oncogenic transcription factor FOXM1 is overexpressed in the majority of human cancers. In addition, FOXM1 has been implicated in cell migration, invasion, angiogenesis and metastasis. The important role of FOXM1 in cancer affirms its significance for therapeutic intervention. Current data suggest that targeting FOXM1 in mono- or combination therapy may have promising therapeutic benefits for the treatment of cancer. However, challenges with the delivery of anti-FOXM1 siRNA to tumors and the absence of small molecules, which specifically inhibit FOXM1, are delaying the development of FOXM1 inhibitors as feasible anticancer drugs. In this review, we describe and summarize the efforts that have been made to target FOXM1 in cancer and the consequences of FOXM1 suppression in human cancer cells.
Collapse
Affiliation(s)
- Marianna Halasi
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Andrei L Gartel
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America.
| |
Collapse
|
44
|
Teh MT. FOXM1 coming of age: time for translation into clinical benefits? Front Oncol 2012; 2:146. [PMID: 23087907 PMCID: PMC3471356 DOI: 10.3389/fonc.2012.00146] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/01/2012] [Indexed: 01/01/2023] Open
Abstract
A decade since the first evidence implicating the cell cycle transcription factor Forkhead Box M1 (FOXM1) in human tumorigenesis, a slew of subsequent studies revealed an oncogenic role of FOXM1 in the majority of human cancers including oral, nasopharynx, oropharynx, esophagus, breast, ovary, prostate, lung, liver, pancreas, kidney, colon, brain, cervix, thyroid, bladder, uterus, testis, stomach, skin, and blood. Its aberrant upregulation in almost all different cancer types suggests a fundamental role for FOXM1 in tumorigenesis. Its dose-dependent expression pattern correlated well with tumor progression starting from cancer predisposition and initiation, early premalignancy and progression, to metastatic invasion. In addition, emerging studies have demonstrated a causal link between FOXM1 and chemotherapeutic drug resistance. Despite the well-established multifaceted roles for FOXM1 in all stages of oncogenesis, its translation into clinical benefit is yet to materialize. In this contribution, I reviewed and discussed how our current knowledge on the oncogenic mechanisms of FOXM1 could be exploited for clinical use as biomarker for risk prediction, early cancer screening, molecular diagnostics/prognostics, and/or companion diagnostics for personalized cancer therapy.
Collapse
Affiliation(s)
- Muy-Teck Teh
- Centre for Clinical and Diagnostic Oral Sciences, Barts and The London School of Medicine and Dentistry, Queen Mary University of London London, UK
| |
Collapse
|
45
|
Cancer genetics and genomics of human FOX family genes. Cancer Lett 2012; 328:198-206. [PMID: 23022474 DOI: 10.1016/j.canlet.2012.09.017] [Citation(s) in RCA: 302] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 09/20/2012] [Accepted: 09/21/2012] [Indexed: 12/16/2022]
Abstract
Forkhead-box (FOX) family proteins, involved in cell growth and differentiation as well as embryogenesis and longevity, are DNA-binding proteins regulating transcription and DNA repair. The focus of this review is on the mechanisms of FOX-related human carcinogenesis. FOXA1 is overexpressed as a result of gene amplification in lung cancer, esophageal cancer, ER-positive breast cancer and anaplastic thyroid cancer and is point-mutated in prostate cancer. FOXA1 overexpression in breast cancer and prostate cancer is associated with good or poor prognosis, respectively. Single nucleotide polymorphism (SNP) within the 5'-UTR of the FOXE1 (TTF2) gene is associated with thyroid cancer risk. FOXF1 overexpression in breast cancer is associated with epithelial-to-mesenchymal transition (EMT). FOXM1 is overexpressed owing to gene amplification in basal-type breast cancer and diffuse large B-cell lymphoma (DLBCL), and it is transcriptionally upregulated owing to Hedgehog-GLI, hypoxia-HIF1α or YAP-TEAD signaling activation. FOXM1 overexpression leads to malignant phenotypes by directly upregulating CCNB1, AURKB, MYC and SKP2 and indirectly upregulating ZEB1 and ZEB2 via miR-200b downregulation. Tumor suppressor functions of FOXO transcription factors are lost in cancer cells as a result of chromosomal translocation, deletion, miRNA-mediated repression, AKT-mediated cytoplasmic sequestration or ubiquitination-mediated proteasomal degradation. FOXP1 is upregulated as a result of gene fusion or amplification in DLBCL and MALT lymphoma and also repression of miRNAs, such as miR-1, miR-34a and miR-504. FOXP1 overexpression is associated with poor prognosis in DLBCL, gastric MALT lymphoma and hepatocellular carcinoma but with good prognosis in breast cancer. In neuroblastoma, the entire coding region of the FOXR1 (FOXN5) gene is fused to the MLL or the PAFAH1B gene owing to interstitial deletions. FOXR1 fusion genes function as oncogenes that repress transcription of FOXO target genes. Whole-genome sequencing data from tens of thousands of human cancers will uncover the mutational landscape of FOX family genes themselves as well as FOX-binding sites, which will be ultimately applied for cancer diagnostics, prognostics, and therapeutics.
Collapse
|
46
|
Xue YJ, Xiao RH, Long DZ, Zou XF, Wang XN, Zhang GX, Yuan YH, Wu GQ, Yang J, Wu YT, Xu H, Liu FL, Liu M. Overexpression of FoxM1 is associated with tumor progression in patients with clear cell renal cell carcinoma. J Transl Med 2012; 10:200. [PMID: 23006512 PMCID: PMC3492118 DOI: 10.1186/1479-5876-10-200] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 09/19/2012] [Indexed: 11/14/2022] Open
Abstract
Background Fork head box M1 (FoxM1) is a proliferation-associated transcription factor essential for cell cycle progression. Numerous studies have documented that FoxM1 has multiple functions in tumorigenesis and its elevated levels are frequently associated with cancer progression. The present study was conducted to investigate the expression of FoxM1 and its prognostic significance in clear cell renal cell carcinoma (ccRCC). Meanwhile, the function of FoxM1 in human ccRCC was further investigated in cell culture models. Methods Real-time quantitative PCR, western blot and immunohistochemistry were used to explore FoxM1 expression in ccRCC cell lines and primary ccRCC clinical specimens. FoxM1 expression was knocked down by small interfering RNA (siRNA) in Caki-1 and 786-O cells; proliferation, colony formation, cell cycle, migration, invasion, and angiogenesis were assayed. Results FoxM1 expression was up-regulated in the majority of the ccRCC clinical tissue specimens at both mRNA and protein levels. Clinic pathological analysis showed that FoxM1 expression was significantly correlated with primary tumor stage (P <0.001), lymph node metastasis (P = 0.01), distant metastasis (P = 0.01), TNM stage (P < 0.001) and histological grade (P = 0.003). The Kaplan–Meier survival curves revealed that high FoxM1 expression was associated with poor prognosis in ccRCC patients (P < 0.001). FoxM1 expression was an independent prognostic marker of overall ccRCC patient survival in a multivariate analysis (P = 0.008). Experimentally, we found that down-regulation of FoxM1 inhibited cell proliferation and induced cell cycle arrest with reduced expression of cyclin B1, cyclin D1, and Cdk2, and increased expression of p21 and p27. Also, down-regulation of FoxM1 reduced expression and activity of matrix metalloproteinase-2 (MMP-2), MMP-9 and vascular endothelial growth factor (VEGF), resulting in the inhibition of migration, invasion, and angiogenesis. Conclusions These results suggest that FoxM1 expression is likely to play important roles in ccRCC development and progression, and that FoxM1 is a prognostic biomarker and a promising therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Yi-Jun Xue
- Department of Urology, First Affiliated Hospital of Gannan Medical University, No 23, Qing Nian Road, Ganzhou 341000, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Down CF, Millour J, Lam EWF, Watson RJ. Binding of FoxM1 to G2/M gene promoters is dependent upon B-Myb. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1819:855-62. [PMID: 22513242 DOI: 10.1016/j.bbagrm.2012.03.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 01/17/2023]
Abstract
The promoters of genes which regulate entry into and progress through mitosis are typically induced maximally in G2 by transcription factors that include B-Myb and FoxM1. As FoxM1 gene transcription is a target of B-Myb, we investigated in this study how these transcription factors functionally interact to regulate these G2/M genes. Using a 3T3 cell line containing floxed B-myb alleles (B-myb(F/F)) that could be conditionally deleted by Cre recombinase, we confirmed that B-myb knockout caused both decreased mRNA expression of several G2/M genes, including FoxM1, and delayed entry into mitosis. Although FoxM1 protein expression was actually unaffected by B-myb knockout when quiescent B-myb(F/F) 3T3 cells re-entered the cell cycle upon serum-stimulation, chromatin immunoprecipitation revealed that FoxM1 binding to G2/M promoters was substantially reduced. FoxM1 transcriptional activity requires sequential phosphorylation by Cyclin-dependent kinases and Plk1, which are B-Myb target genes, and we found that phosphorylation at Plk1-specific sites was somewhat reduced upon B-myb knockout. Neither this effect nor nuclear accumulation of FoxM1, which was unaffected by B-myb knockout, was sufficient to account for the dependence on B-Myb for FoxM1 promoter binding, however. More significantly, assays using paired Birc5 (survivin) promoter-luciferase reporters with either wild-type or mutated Myb binding sites showed that FoxM1 was unable to bind and activate the promoter in the absence of B-Myb binding. Our data suggest that B-Myb is required as a pioneer factor to enable FoxM1 binding to G2/M gene promoters and explains how these transcription factors may collaborate to induce mitosis.
Collapse
|
48
|
Ha SY, Lee CH, Chang HK, Chang S, Kwon KY, Lee EH, Roh MS, Seo B. Differential expression of forkhead box M1 and its downstream cyclin-dependent kinase inhibitors p27(kip1) and p21(waf1/cip1) in the diagnosis of pulmonary neuroendocrine tumours. Histopathology 2012; 60:731-9. [PMID: 22296117 DOI: 10.1111/j.1365-2559.2011.04137.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIMS Pulmonary neuroendocrine (NE) tumours represent a spectrum of phenotypically distinct entities with different biological behaviours. Difficulties in classifying these tumours are frequently encountered in clinical practice. Forkhead box M1 (FoxM1) is essential for the development of various cancers and is a proliferation-specific transcription factor that regulates transcription of cell cycle genes, including cyclin-dependent kinase inhibitors p27(kip1) and p21(waf1/cip1) . This study was performed to determine the utility of FoxM1, p27(kip1) and p21(waf1/cip1) as immunomarkers for subtyping pulmonary NE tumours. METHODS AND RESULTS FoxM1, p27(kip1) and p21(waf1/cip1) expression was evaluated by immunohistochemistry in 60 pulmonary NE tumours [19 typical carcinoids (TCs), six atypical carcinoids (ACs), 17 large cell neuroendocrine carcinomas (LCNECs) and 18 small cell lung cancers (SCLCs)]. The frequencies of FoxM1 and p21(waf1/cip1) expression were significantly different between TCs and ACs (each P = 0.009), and those of FoxM1 and p27(kip1) expression were significantly different between LCNECs and SCLCs (P = 0.012 and P = 0.002, respectively). The combined FoxM1((-)) /p21(waf1/cip1(-)) and FoxM1((+)) /p27(kip1(high)) phenotypes had the best diagnostic accuracy for distinguishing TCs from ACs, and SCLCs from LCNECs, respectively. CONCLUSIONS FoxM1, p27(kip1) and p21(waf1/cip1) showed distinct immunoreactivity according to histological subtype, which may be of value as an ancillary test in the differential diagnosis of pulmonary NE tumours.
Collapse
Affiliation(s)
- Seung Yeon Ha
- Department of Pathology, Gachon University of Medicine and Science, Incheon, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Lynch TP, Ferrer CM, Jackson SR, Shahriari KS, Vosseller K, Reginato MJ. Critical role of O-Linked β-N-acetylglucosamine transferase in prostate cancer invasion, angiogenesis, and metastasis. J Biol Chem 2012; 287:11070-81. [PMID: 22275356 DOI: 10.1074/jbc.m111.302547] [Citation(s) in RCA: 245] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Cancer cells universally increase glucose and glutamine consumption, leading to the altered metabolic state known as the Warburg effect; one metabolic pathway, highly dependent on glucose and glutamine, is the hexosamine biosynthetic pathway. Increased flux through the hexosamine biosynthetic pathway leads to increases in the post-translational addition of O-linked β-N-acetylglucosamine (O-GlcNAc) to various nuclear and cytosolic proteins. A number of these target proteins are implicated in cancer, and recently, O-GlcNAcylation was shown to play a role in breast cancer; however, O-GlcNAcylation in other cancers remains poorly defined. Here, we show that O-GlcNAc transferase (OGT) is overexpressed in prostate cancer compared with normal prostate epithelium and that OGT protein and O-GlcNAc levels are elevated in prostate carcinoma cell lines. Reducing O-GlcNAcylation in PC3-ML cells was associated with reduced expression of matrix metalloproteinase (MMP)-2, MMP-9, and VEGF, resulting in inhibition of invasion and angiogenesis. OGT-mediated regulation of invasion and angiogenesis was dependent upon regulation of the oncogenic transcription factor FoxM1, a key regulator of invasion and angiogenesis, as reducing OGT expression led to increased FoxM1 protein degradation. Conversely, overexpression of a degradation-resistant FoxM1 mutant abrogated OGT RNAi-mediated effects on invasion, MMP levels, angiogenesis, and VEGF expression. Using a mouse model of metastasis, we found that reduction of OGT expression blocked bone metastasis. Altogether, these data suggest that as prostate cancer cells alter glucose and glutamine levels, O-GlcNAc modifications and OGT levels become elevated and are required for regulation of malignant properties, implicating OGT as a novel therapeutic target in the treatment of cancer.
Collapse
Affiliation(s)
- Thomas P Lynch
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, USA
| | | | | | | | | | | |
Collapse
|
50
|
Priller M, Pöschl J, Abrão L, von Bueren AO, Cho YJ, Rutkowski S, Kretzschmar HA, Schüller U. Expression of FoxM1 is required for the proliferation of medulloblastoma cells and indicates worse survival of patients. Clin Cancer Res 2011; 17:6791-801. [PMID: 21918172 DOI: 10.1158/1078-0432.ccr-11-1214] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The transcription factor Forkhead box M1 (FoxM1) is a key regulator of cell-cycle progression. It is involved in the development of multiple organs, and we have previously reported on its important role for the mitotic entry of cerebellar granule neuron precursors. Constitutive expression of FoxM1 is required for the growth of multiple cancer types. This study aimed to determine its role in medulloblastoma, the most frequent malignant brain tumor in childhood that can derive from cerebellar granule neuron precursors. EXPERIMENTAL DESIGN We evaluated the expression of FoxM1 together with its prognostic value in two independent series of human medulloblastoma samples using immunohistochemistry (n = 43) and gene expression arrays (n = 193). The functional impact of FoxM1 expression was characterized by knockdown experiments in four human medulloblastoma cell lines, and the thiazole antibiotic siomycin A was tested to downregulate FoxM1 and inhibit tumor cell growth. RESULTS FoxM1 was highly expressed in all subtypes of medulloblastoma. Importantly, expression levels of FoxM1 significantly correlated with unfavorable clinical outcome in univariate analysis (P = 0.0005), and FoxM1 was identified as an independent prognostic marker by multivariate analysis (P = 0.037). Knockdown of FoxM1 in medulloblastoma cell lines resulted in a significant decrease of cell viability which was caused by a failure in mitotic spindle formation and caspase-dependent mitotic catastrophe. Siomycin A significantly inhibited the expression of FoxM1 and the growth of medulloblastoma cells. CONCLUSIONS FoxM1 may be used as an additional prognostic marker and may represent a potential novel target to treat patients suffering from medulloblastoma.
Collapse
Affiliation(s)
- Markus Priller
- Center for Neuropathology, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|