51
|
Murlistyarini S, Aninda LP, Widyarti S, Endharti AT, Sardjono TW. Exosomes of Adipose-derived Stem Cells Conditioned Media Promotes Retinoblastoma and Forkhead-Box M1 Protein Expression. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: In the senescence process, the retinoblastoma (Rb) protein binds to E2F in hypophosphorylated conditions, preventing the cell to enter the S-phase in the cell cycle. Human Forkhead Box M1 (FOXM1) protein, key regulator G1/S and G2/M phases, decreases in the senescence process. Many studies have been carried out to reverse this system, one of which used exosomes of adipose-derived stem c ells conditioned media (ADSC-CM). These exosomes contain a variety of specific proteins which have pro-proliferation properties, however, little is known on the role of these exosomes toward the change of phosphorylated Rb and FOXM1.
AIM: This study aims to find out the involvement of exosomes of ADSC-CM on these two proteins on senescence human dermal fibroblasts (HDFs).
METHODS: In vitro experiment was undergone randomization sample and non-blinded pre-/post-test control group. The primary culture of senescent HDFs was transfected with exosomes of ADSC-CM; then, its effect on migration and senescence reversal was observed through analyzing Sa-β-gal, Rb, and FOXM1 protein expression.
RESULTS: The expression of Sa-β-gal was higher in the control group. Our result demonstrated the exosome of ADSC-CM significantly induced the expression of Rb and FOXM1 protein in senescent HDFs (p < 0.05).
CONCLUSION: It proved that exosomes of ADSC-CM could shift the senescent fibroblast into metabolically active cells.
Collapse
|
52
|
Zhang W, Zhang X, Huang S, Chen J, Ding P, Wang Q, Li L, Lv X, Li L, Zhang P, Zhou D, Wen W, Wang Y, Lei Q, Wu J, Hu W. FOXM1D potentiates PKM2-mediated tumor glycolysis and angiogenesis. Mol Oncol 2021; 15:1466-1485. [PMID: 33314660 PMCID: PMC8096781 DOI: 10.1002/1878-0261.12879] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/16/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor growth, especially in the late stage, requires adequate nutrients and rich vasculature, in which PKM2 plays a convergent role. It has been reported that PKM2, together with FOXM1D, is upregulated in late-stage colorectal cancer and associated with metastasis; however, their underlying mechanism for promoting tumor progression remains elusive. Herein, we revealed that FOXM1D potentiates PKM2-mediated glycolysis and angiogenesis through multiple protein-protein interactions. In the presence of FBP, FOXM1D binds to tetrameric PKM2 and assembles a heterooctamer, restraining PKM2 metabolic activity by about a half and thereby promoting aerobic glycolysis. Furthermore, FOXM1D interacts with PKM2 and NF-κB and induces their nuclear translocation with the assistance of the nuclear transporter importin 4. Once in the nucleus, PKM2 and NF-κB complexes subsequently augment VEGFA transcription. The increased VEGFA is secreted extracellularly via exosomes, an event potentiated by the interaction of FOXM1 with VPS11, eventually promoting tumor angiogenesis. Based on these findings, our study provides another insight into the role of PKM2 in the regulation of glycolysis and angiogenesis.
Collapse
Affiliation(s)
- Wei Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xin Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Sheng Huang
- Department of Breast SurgeryBreast Cancer InstituteFudan University Shanghai Cancer CenterShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jianfeng Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qi Wang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Luying Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xinyue Lv
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Ling Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Pingzhao Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Danlei Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wenyu Wen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yiping Wang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qun‐Ying Lei
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jiong Wu
- Department of Breast SurgeryBreast Cancer InstituteFudan University Shanghai Cancer CenterShanghai Medical CollegeFudan UniversityShanghaiChina
- Key Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Key Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
| |
Collapse
|
53
|
Barger CJ, Chee L, Albahrani M, Munoz-Trujillo C, Boghean L, Branick C, Odunsi K, Drapkin R, Zou L, Karpf AR. Co-regulation and function of FOXM1/ RHNO1 bidirectional genes in cancer. eLife 2021; 10:e55070. [PMID: 33890574 PMCID: PMC8104967 DOI: 10.7554/elife.55070] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
The FOXM1 transcription factor is an oncoprotein and a top biomarker of poor prognosis in human cancer. Overexpression and activation of FOXM1 is frequent in high-grade serous carcinoma (HGSC), the most common and lethal form of human ovarian cancer, and is linked to copy number gains at chromosome 12p13.33. We show that FOXM1 is co-amplified and co-expressed with RHNO1, a gene involved in the ATR-Chk1 signaling pathway that functions in the DNA replication stress response. We demonstrate that FOXM1 and RHNO1 are head-to-head (i.e., bidirectional) genes (BDG) regulated by a bidirectional promoter (BDP) (named F/R-BDP). FOXM1 and RHNO1 each promote oncogenic phenotypes in HGSC cells, including clonogenic growth, DNA homologous recombination repair, and poly-ADP ribosylase inhibitor resistance. FOXM1 and RHNO1 are one of the first examples of oncogenic BDG, and therapeutic targeting of FOXM1/RHNO1 BDG is a potential therapeutic approach for ovarian and other cancers.
Collapse
MESH Headings
- Ataxia Telangiectasia Mutated Proteins/genetics
- Ataxia Telangiectasia Mutated Proteins/metabolism
- Carboplatin/pharmacology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Checkpoint Kinase 1/genetics
- Checkpoint Kinase 1/metabolism
- Databases, Genetic
- Drug Resistance, Neoplasm
- Female
- Forkhead Box Protein M1/genetics
- Forkhead Box Protein M1/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Neoplasms, Cystic, Mucinous, and Serous/drug therapy
- Neoplasms, Cystic, Mucinous, and Serous/genetics
- Neoplasms, Cystic, Mucinous, and Serous/metabolism
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Promoter Regions, Genetic
- Recombinational DNA Repair
- Signal Transduction
Collapse
Affiliation(s)
- Carter J Barger
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Linda Chee
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Mustafa Albahrani
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Catalina Munoz-Trujillo
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Lidia Boghean
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Connor Branick
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Kunle Odunsi
- Departments of Gynecologic Oncology, Immunology, and Center for Immunotherapy, Roswell Park Comprehensive Cancer CenterBuffaloUnited States
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Lee Zou
- Massachusetts General Hospital Cancer Center, Harvard Medical SchoolCharlestownUnited States
| | - Adam R Karpf
- Eppley Institute for Cancer Research and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| |
Collapse
|
54
|
Sun G, Wu L, Sun G, Shi X, Cao H, Tang W. WNT5a in Colorectal Cancer: Research Progress and Challenges. Cancer Manag Res 2021; 13:2483-2498. [PMID: 33758546 PMCID: PMC7981155 DOI: 10.2147/cmar.s289819] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/17/2021] [Indexed: 12/31/2022] Open
Abstract
Despite the clinical development of new adjuvant and neoadjuvant chemotherapy drugs, colorectal cancer is still one of the leading causes of cancer-related death in human beings. WNT5a, an autocrine and paracrine β-catenin independent ligand, has been shown to induce tumor inhibition and carcinogenic signals, depending on the type of cancer. In patients with colorectal cancer, WNT5a triggers a variety of downstream signaling pathways, which mainly affect the migration and invasion of tumor cells. This article reviews the mechanism and therapeutic potential of WNT5a in colorectal cancer. In short, an in-depth understanding of the role of WNT5a in colorectal cancer is very helpful to better deal with this disease.
Collapse
Affiliation(s)
- Guangshun Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Liangliang Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Guoqiang Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xuesong Shi
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Hongyong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Weiwei Tang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
55
|
Identification of Redox-Sensitive Transcription Factors as Markers of Malignant Pleural Mesothelioma. Cancers (Basel) 2021; 13:cancers13051138. [PMID: 33799965 PMCID: PMC7961847 DOI: 10.3390/cancers13051138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Malignant pleural mesothelioma is a lung tumor associated with asbestos exposure, with a poor prognosis, and a difficult pharmacological approach. Asbestos exposure is very toxic for the lungs, which counteract this toxic effect by activating some antioxidant defense proteins. When these proteins are more active that in normal conditions, as in several cancers, these tumors become able to survive and resist to stress or chemotherapy. In our laboratory, we collected cellular samples of mesothelioma and non-transformed mesothelium from Hospital’s Biobank and we evaluated these proteins. Our results demonstrated these proteins are upregulated in mesothelioma cells and not in non-transformed mesothelium. This event could be associated to toxic effects evoked by asbestos exposure, highlighting the need in the future to monitor asbestos-exposed people by measuring biomarkers identified, in the attempt to identify them as possible predictive markers and potential pharmacological targets addressed to improve mesothelioma prognosis. Abstract Although asbestos has been banned in most countries around the world, malignant pleural mesothelioma (MPM) is a current problem. MPM is an aggressive tumor with a poor prognosis, so it is crucial to identify new markers in the preventive field. Asbestos exposure induces oxidative stress and its carcinogenesis has been linked to a strong oxidative damage, event counteracted by antioxidant systems at the pulmonary level. The present study has been focused on some redox-sensitive transcription factors that regulate cellular antioxidant defense and are overexpressed in many tumors, such as Nrf2 (Nuclear factor erythroid 2-related factor 2), Ref-1 (Redox effector factor 1), and FOXM1 (Forkhead box protein M1). The research was performed in human mesothelial and MPM cells. Our results have clearly demonstrated an overexpression of Nrf2, Ref-1, and FOXM1 in mesothelioma towards mesothelium, and a consequent activation of downstream genes controlled by these factors, which in turn regulates antioxidant defense. This event is mediated by oxidative free radicals produced when mesothelial cells are exposed to asbestos fibers. We observed an increased expression of Nrf2, Ref-1, and FOXM1 towards untreated cells, confirming asbestos as the mediator of oxidative stress evoked at the mesothelium level. These factors can therefore be considered predictive biomarkers of MPM and potential pharmacological targets in the treatment of this aggressive cancer.
Collapse
|
56
|
FOXM1 Inhibition in Ovarian Cancer Tissue Cultures Affects Individual Treatment Susceptibility Ex Vivo. Cancers (Basel) 2021; 13:cancers13050956. [PMID: 33668819 PMCID: PMC7956612 DOI: 10.3390/cancers13050956] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/03/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Late diagnosis of ovarian cancer is a major reason for the high mortality rate of this tumor entity. The time to determine tumor susceptibility to treatment is scarce and resistance to therapy occurs very frequently. Here, we aim for a model system that can determine tumor response to (I) study novel drugs and (II) enhance patient stratification. Tissue specimens (n = 10) were acquired from fresh surgical samples. Tissue cultures were cultivated and treated with clinically relevant therapeutics and an FOXM1 inhibitor for 3–6 days. The transcription factor FOXM1 is a key regulator of tumor survival affecting multiple cancerogenic target genes. Gene expression of FOXM1 and its targets BRCA1/2 and RAD51 were investigated together with tumor susceptibility. Tissue cultures successfully demonstrated the individual benefit of FOXM1 inhibition and revealed the potency of the complex model system for oncological research. Abstract Diagnosis in an advanced state is a major hallmark of ovarian cancer and recurrence after first line treatment is common. With upcoming novel therapies, tumor markers that support patient stratification are urgently needed to prevent ineffective therapy. Therefore, the transcription factor FOXM1 is a promising target in ovarian cancer as it is frequently overexpressed and associated with poor prognosis. In this study, fresh tissue specimens of 10 ovarian cancers were collected to investigate tissue cultures in their ability to predict individual treatment susceptibility and to identify the benefit of FOXM1 inhibition. FOXM1 inhibition was induced by thiostrepton (3 µM). Carboplatin (0.2, 2 and 20 µM) and olaparib (10 µM) were applied and tumor susceptibility was analyzed by tumor cell proliferation and apoptosis in immunofluorescence microscopy. Resistance mechanisms were investigated by determining the gene expression of FOXM1 and its targets BRCA1/2 and RAD51. Ovarian cancer tissue was successfully maintained for up to 14 days ex vivo, preserving morphological characteristics of the native specimen. Thiostrepton downregulated FOXM1 expression in tissue culture. Individual responses were observed after combined treatment with carboplatin or olaparib. Thus, we successfully implemented a complex tissue culture model to ovarian cancer and showed potential benefit of combined FOXM1 inhibition.
Collapse
|
57
|
Kalathil D, John S, Nair AS. FOXM1 and Cancer: Faulty Cellular Signaling Derails Homeostasis. Front Oncol 2021; 10:626836. [PMID: 33680951 PMCID: PMC7927600 DOI: 10.3389/fonc.2020.626836] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Forkhead box transcription factor, FOXM1 is implicated in several cellular processes such as proliferation, cell cycle progression, cell differentiation, DNA damage repair, tissue homeostasis, angiogenesis, apoptosis, and redox signaling. In addition to being a boon for the normal functioning of a cell, FOXM1 turns out to be a bane by manifesting in several disease scenarios including cancer. It has been given an oncogenic status based on several evidences indicating its role in tumor development and progression. FOXM1 is highly expressed in several cancers and has also been implicated in poor prognosis. A comprehensive understanding of various aspects of this molecule has revealed its role in angiogenesis, invasion, migration, self- renewal and drug resistance. In this review, we attempt to understand various mechanisms underlying FOXM1 gene and protein regulation in cancer including the different signaling pathways, post-transcriptional and post-translational modifications. Identifying crucial molecules associated with these processes can aid in the development of potential pharmacological approaches to curb FOXM1 mediated tumorigenesis.
Collapse
Affiliation(s)
- Dhanya Kalathil
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Samu John
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Research Centre, University of Kerala, Thiruvananthapuram, India
| | - Asha S Nair
- Cancer Research Program-4, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Research Centre, University of Kerala, Thiruvananthapuram, India
| |
Collapse
|
58
|
Gao J, Ding C, Zhou J, Wu G, Han Z, Li J, Hei F. Propofol suppresses lung cancer tumorigenesis by modulating the circ-ERBB2/miR-7-5p/FOXM1 axis. Thorac Cancer 2021; 12:824-834. [PMID: 33506582 PMCID: PMC7952809 DOI: 10.1111/1759-7714.13856] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Propofol is a commonly used anesthetic for cancer surgery. Previous studies have shown that propofol has an anticancer role in various cancers, including lung cancer. This study aimed to investigate the role of propofol in lung cancer and its underlying mechanism. METHODS Cell proliferation was determined by cell counting kit-8 (CCK-8) and colony formation assays. Flow cytometry and transwell assays were used to detect cell apoptosis and invasion, respectively. Glycolysis was evaluated by detecting glucose consumption, lactate production and ATP/ADP ratios. The levels of circular RNA erb-b2 receptor tyrosine kinase 2 (circ-ERBB2), microRNA-7-5p (miR-7-5p) and forkhead box M1 (FOXM1) were tested by quantitative real-time PCR and Western blot. The binding relationship between miR-7-5p and circ-ERBB2/FOXM1 was verified by dual-luciferase reporter assay. Moreover, in vivo experiments were performed by establishing a mouse xenograft model. RESULTS Propofol suppressed cell proliferation, invasion and glycolysis and expedited apoptosis in lung cancer cells. Circ-ERBB2 and FOXM1 were upregulated, while miR-7-5p was decreased in lung cancer tissues and cells. Propofol suppressed lung cancer cell progression by regulating circ-ERBB2. Additionally, miR-7-5p directly interacted with circ-ERBB2 and FOXM1. Also, propofol played an antitumor role in lung cancer via modulating miR-7-5p or FOXM1. Moreover, circ-ERBB2 knockdown enhanced the suppressive effect of propofol on tumor growth in vivo. CONCLUSIONS Propofol inhibited lung cancer progression via mediating circ-ERBB2/miR-7-5p/FOXM1 axis, which might provide an effective therapeutic target for lung cancer therapy.
Collapse
Affiliation(s)
- Jie Gao
- Department of Anesthesiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Chengzhi Ding
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Junhui Zhou
- Department of Anesthesiology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Gang Wu
- Department of Cardiovascular Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Zongmao Han
- Department of Cardiology, People's Hospital of Zhengzhou University, People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jianchao Li
- Department of Extracorporeal Circulation, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Feilong Hei
- Department of Extracorporeal Circulation, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
59
|
Detarya M, Thaenkaew S, Seubwai W, Indramanee S, Phoomak C, Saengboonmee C, Wongkham S, Wongkham C. High glucose upregulates FOXM1 expression via EGFR/STAT3 dependent activation to promote progression of cholangiocarcinoma. Life Sci 2021; 271:119114. [PMID: 33513399 DOI: 10.1016/j.lfs.2021.119114] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/21/2022]
Abstract
AIMS Epidemiological studies indicate diabetes mellitus and hyperglycemia as risk factors of cancers including cholangiocarcinoma (CCA). How high glucose promotes cancer development and progression, however, is still unrevealed. In this study, insight into the molecular pathway of high glucose promoting progression of CCA cells was investigated. MAIN METHODS Human CCA cell lines, KKU-213A and KKU-213B were cultured in normal glucose (NG; 5.56 mM) or high glucose (HG; 25 mM) and used as NG and HG cells. Forkhead box M1 (FOXM1) expression was transiently suppressed using siFOXM1. Western blotting and image analysis were employed to semi-quantitatively determine the expression levels of the specified proteins. The migration and invasion of CCA cells were revealed using Boyden chamber assays. KEY FINDINGS All HG cells exhibited higher expression of FOXM1 than the corresponding NG cells in a dose dependent manner. Suppression of FOXM1 expression by siFOXM1 significantly reduced migration and invasion abilities of CCA cells by suppression of Slug and MMP2 expression. Inhibition of STAT3 activation using Stattic, significantly suppressed expression of FOXM1 and Slug and decreased migration and invasion abilities of HG cells. In addition, EGFR expression was significantly higher in HG cells than NG cells and increased dependently with glucose concentration. Inhibition of EGFR activation by cetuximab significantly suppressed STAT3 activation and FOXM1 expression. SIGNIFICANCE The mechanism of high glucose promoting progression of CCA cells was revealed to be via in part by upregulation of FOXM1 expression under EGF/EGFR and STAT3 dependent activation.
Collapse
Affiliation(s)
- Marutpong Detarya
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Salak Thaenkaew
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Basic-Related subject Department, Khon Kaen Vocational College, Khon Kaen 40000, Thailand
| | - Wunchana Seubwai
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Department of Forensic Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Somsiri Indramanee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chatchai Phoomak
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chaisiri Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
60
|
Xu J, Wang K, Zhang Z, Xue D, Li W, Pan Z. The Role of Forkhead Box Family in Bone Metabolism and Diseases. Front Pharmacol 2021; 12:772237. [PMID: 35153742 PMCID: PMC8832510 DOI: 10.3389/fphar.2021.772237] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022] Open
Abstract
Forkhead box (Fox) family, an evolutionarily conserved family of transcription factors carrying the "Forkhead" motif, plays an indispensable role in human health and disease. Fox family genes are involved in cell differentiation, proliferation and apoptosis, embryonic development, aging, glucose and lipid metabolism, and immune regulation. The regulatory role of the Fox family in the context of bone metabolism and orthopedic diseases is an emerging research hotspot. In this review, we highlight the major molecular mechanisms underlying the regulatory role of Fox factors in bone metabolism, bone development, bone homeostasis, and bone diseases associated with inhibition or upregulation of Fox factors. In addition, we discuss the emerging evidence in the realm of Fox factor-based therapeutics.
Collapse
Affiliation(s)
- Jianxiang Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Kanbin Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Zengjie Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Deting Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- *Correspondence: Deting Xue, ; Weixu Li, ; Zhijun Pan,
| | - Weixu Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- *Correspondence: Deting Xue, ; Weixu Li, ; Zhijun Pan,
| | - Zhijun Pan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- *Correspondence: Deting Xue, ; Weixu Li, ; Zhijun Pan,
| |
Collapse
|
61
|
Transcription factors in colorectal cancer: molecular mechanism and therapeutic implications. Oncogene 2020; 40:1555-1569. [PMID: 33323976 DOI: 10.1038/s41388-020-01587-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is a major cause of cancer mortality worldwide, however, the molecular mechanisms underlying the pathogenesis of CRC remain largely unclear. Recent studies have revealed crucial roles of transcription factors in CRC development. Transcription factors essential for the regulation of gene expression by interacting with transcription corepressor/enhancer complexes and they orchestrate downstream signal transduction. Deregulation of transcription factors is a frequent occurrence in CRC, and the accompanying drastic changes in gene expression profiles play fundamental roles in multistep process of tumorigenesis, from cellular transformation, disease progression to metastatic disease. Herein, we summarized current and emerging key transcription factors that participate in CRC tumorigenesis, and highlighted their oncogenic or tumor suppressive functions. Moreover, we presented critical transcription factors of CRC, emphasized the major molecular mechanisms underlying their effect on signal cascades associated with tumorigenesis, and summarized of their potential as molecular biomarkers for CRC prognosis therapeutic response, as well as drug targets for CRC treatment. A better understanding of transcription factors involved in the development of CRC will provide new insights into the pathological mechanisms and reveal novel prognostic biomarkers and therapeutic strategies for CRC.
Collapse
|
62
|
Yang Y, Jiang H, Li W, Chen L, Zhu W, Xian Y, Han Z, Yin L, Liu Y, Wang Y, Pan K, Zhang K. FOXM1/DVL2/Snail axis drives metastasis and chemoresistance of colorectal cancer. Aging (Albany NY) 2020; 12:24424-24440. [PMID: 33291076 PMCID: PMC7762457 DOI: 10.18632/aging.202300] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022]
Abstract
Colorectal cancer (CRC) is the third most common type of cancer worldwide. Metastasis and chemoresistance are regarded as the two leading causes of treatment failure and high mortality in CRC. Forkhead Box M1 (FOXM1) has been involved in malignant behaviors of cancer. However, the role and mechanism of FOXM1 in simultaneously regulating metastasis and chemoresistance of CRC remain poorly understood. Here, we found that FOXM1 was overexpressed in oxaliplatin- and vincristine-resistant CRC cells (HCT-8/L-OHP and HCT-8/VCR) with enhanced metastatic potential, compared with HCT-8 cells. FOXM1 overexpression increased migration, invasion and drug-resistance to oxaliplatin and vincristine in HCT-8 cells, while FOXM1 knockdown using shFOXM1 impaired metastasis and drug-resistance in HCT-8/L-OHP and HCT-8/VCR cells. Moreover, FOXM1 up-regulated Snail to trigger epithelial-mesenchymal transition-like molecular changes and multidrug-resistance protein P-gp expression, while silencing Snail inhibited FOXM1-induced metastasis and drug-resistance. We further identified that disheveled-2 (DVL2) was crucial for FOXM1-induced Snail expression, metastasis and chemoresistance. Furthermore, FOXM1 bound to DVL2, and enhanced nuclear translocation of DVL2 and DVL2-mediated transcriptional activity of Wnt/β-catenin known to induce Snail expression. In conclusion, FOXM1/DVL2/Snail axis triggered aggressiveness of CRC. Blocking FOXM1/DVL2/Snail pathway simultaneously inhibited metastasis and chemoresistance in CRC cells, providing a new strategy for successful CRC treatment.
Collapse
Affiliation(s)
- Yuhan Yang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Hequn Jiang
- First Afflicted Hospital, Chengdu Medical College, Chengdu, China
| | - Wanxin Li
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Linyi Chen
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Wanglong Zhu
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Yu Xian
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Zhengyu Han
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Lan Yin
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Yao Liu
- School of Medical Laboratory Science, Chengdu Medical College, Chengdu, China
| | - Yi Wang
- First Afflicted Hospital, Chengdu Medical College, Chengdu, China
| | - Kejian Pan
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Kun Zhang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
63
|
Cai X, Xiao W, Shen J, Lian H, Lu Y, Liu X, Gu J. Thiostrepton and miR-216b synergistically promote osteosarcoma cell cytotoxicity and apoptosis by targeting FoxM1. Oncol Lett 2020; 20:391. [PMID: 33193851 PMCID: PMC7656114 DOI: 10.3892/ol.2020.12254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/07/2020] [Indexed: 01/08/2023] Open
Abstract
Osteosarcoma is a common primary bone cancer that there are currently no effective treatment strategies for. Forkhead box M1 (FoxM1) is key in the development of osteosarcoma, and microRNA (miR)-216b serves an antitumor role by targeting FoxM1. Moreover, thiostrepton (TST), a natural thiazole antibiotic, induces antitumor effects and specifically targets FoxM1. Therefore, the present study investigated whether thiostrepton and miR-216b synergistically inhibited osteosarcoma cells by targeting FoxM1. The MTT assay, reverse transcription-quantitative PCR, a dual-luciferase reporter assay and flow cytometry were performed. Compared with the human osteoblast cell line hFOB1.19, miR-216b expression was significantly downregulated in the osteosarcoma cell lines U2OS, MG63 and Saos-2. By contrast, FoxM1 expression was significantly upregulated in osteosarcoma cell lines compared with the hFOB1.19 cell line. The results indicated that miR-216b targeted the 3′-untranslated region of FoxM1. Moreover, the results suggested that miR-216b cooperated with TST to decrease cell cytotoxicity and increase cell apoptosis. In addition, miR-216b cooperated with TST to increase Bax expression and decrease Bcl-2 expression. In conclusion, the combination of TST and miR-216b synergistically promoted osteosarcoma cell cytotoxicity and apoptosis by targeting FoxM1. Therefore, the present study suggested that the combination of TST and miR-216b may serve as a promising therapeutic strategy for osteosarcoma.
Collapse
Affiliation(s)
- Xiaobing Cai
- Department of Orthopedics, Chongming Branch of Tongji Univercity Affiliated the Tenth People's Hospital, Shanghai 202157, P.R. China
| | - Wenyu Xiao
- Department of Orthopedics, Jiangwan Hospital, Shanghai 200434, P.R. China
| | - Juexin Shen
- Department of Orthopedics, Jiangwan Hospital, Shanghai 200434, P.R. China
| | - Hui Lian
- Department of Orthopedics, Jiangwan Hospital, Shanghai 200434, P.R. China
| | - Yi Lu
- Department of Orthopedics, Jiangwan Hospital, Shanghai 200434, P.R. China
| | - Xianmiao Liu
- Department of Orthopedics, Jiangwan Hospital, Shanghai 200434, P.R. China
| | - Jisheng Gu
- Department of Orthopedics, Jiangwan Hospital, Shanghai 200434, P.R. China
| |
Collapse
|
64
|
Long J, Huang S, Bai Y, Mao J, Wang A, Lin Y, Yang X, Wang D, Lin J, Bian J, Yang X, Sang X, Wang X, Zhao H. Transcriptional landscape of cholangiocarcinoma revealed by weighted gene coexpression network analysis. Brief Bioinform 2020; 22:5923107. [PMID: 33051665 DOI: 10.1093/bib/bbaa224] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 12/19/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a type of cancer with limited treatment options and a poor prognosis. Although some important genes and pathways associated with CCA have been identified, the relationship between coexpression and phenotype in CCA at the systems level remains unclear. In this study, the relationships underlying the molecular and clinical characteristics of CCA were investigated by employing weighted gene coexpression network analysis (WGCNA). The gene expression profiles and clinical features of 36 patients with CCA were analyzed to identify differentially expressed genes (DEGs). Subsequently, the coexpression of DEGs was determined by using the WGCNA method to investigate the correlations between pairs of genes. Network modules that were significantly correlated with clinical traits were identified. In total, 1478 mRNAs were found to be aberrantly expressed in CCA. Seven coexpression modules that significantly correlated with clinical characteristics were identified and assigned representative colors. Among the 7 modules, the green and blue modules were significantly related to tumor differentiation. Seventy-eight hub genes that were correlated with tumor differentiation were found in the green and blue modules. Survival analysis showed that 17 hub genes were prognostic biomarkers for CCA patients. In addition, we found five new targets (ISM1, SULT1B1, KIFC1, AURKB and CCNB1) that have not been studied in the context of CCA and verified their differential expression in CCA through experiments. Our results not only promote our understanding of the relationship between the transcriptome and clinical data in CCA but will also guide the development of targeted molecular therapy for CCA.
Collapse
Affiliation(s)
- Junyu Long
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shan Huang
- Department of Immunology, School of Basic Medical Sciences; Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory for Cancer Invasion and Metastasis, Department of Oncology, Capital Medical University, Beijing, China
| | - Yi Bai
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinzhu Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Anqiang Wang
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, China
| | - Yu Lin
- Shenzhen Withsum Technology Limited, Shenzhen, China
| | - Xu Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Dongxu Wang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianzhen Lin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jin Bian
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaobo Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xi Wang
- Department of Immunology, School of Basic Medical Sciences; Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory for Cancer Invasion and Metastasis, Department of Oncology, Capital Medical University, Beijing, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
65
|
Han H, Davidson LA, Fan Y, Goldsby JS, Yoon G, Jin U, Wright GA, Landrock KK, Weeks BR, Wright RC, Allred CD, Jayaraman A, Ivanov I, Roper J, Safe SH, Chapkin RS. Loss of aryl hydrocarbon receptor potentiates FoxM1 signaling to enhance self-renewal of colonic stem and progenitor cells. EMBO J 2020; 39:e104319. [PMID: 32915464 PMCID: PMC7527924 DOI: 10.15252/embj.2019104319] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that senses xenobiotics, diet, and gut microbial-derived metabolites, is increasingly recognized as a key regulator of intestinal biology. However, its effects on the function of colonic stem and progenitor cells remain largely unexplored. Here, we observed that inducible deletion of AhR in Lgr5+ stem cells increases the percentage of colonic stem cells and enhances organoid initiating capacity and growth of sorted stem and progenitor cells, while AhR activation has the opposite effect. Moreover, intestinal-specific AhR knockout increases basal stem cell and crypt injury-induced cell proliferation and promotes colon tumorigenesis in a preclinical colitis-associated tumor model by upregulating FoxM1 signaling. Mechanistically, AhR transcriptionally suppresses FoxM1 expression. Activation of AhR in human organoids recapitulates phenotypes observed in mice, such as reduction in the percentage of colonic stem cells, promotion of stem cell differentiation, and attenuation of FoxM1 signaling. These findings indicate that the AhR-FoxM1 axis, at least in part, mediates colonic stem/progenitor cell behavior.
Collapse
Affiliation(s)
- Huajun Han
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA,Department of Biochemistry & BiophysicsTexas A&M UniversityCollege StationTXUSA
| | - Laurie A Davidson
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA,Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Yang‐Yi Fan
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA,Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Jennifer S Goldsby
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA,Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Grace Yoon
- Department of StatisticsTexas A&M UniversityCollege StationTXUSA
| | - Un‐Ho Jin
- Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Gus A Wright
- Department of Veterinary PathobiologyTexas A&M UniversityCollege StationTXUSA
| | - Kerstin K Landrock
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA,Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Bradley R Weeks
- Department of Veterinary PathobiologyTexas A&M UniversityCollege StationTXUSA
| | - Rachel C Wright
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA,Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | | | - Arul Jayaraman
- Department of Chemical EngineeringTexas A&M UniversityCollege StationTXUSA
| | - Ivan Ivanov
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA,Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Jatin Roper
- Department of MedicineDivision of GastroenterologyDuke University School of MedicineDurhamNCUSA
| | - Stephen H Safe
- Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA,Department of Biochemistry & BiophysicsTexas A&M UniversityCollege StationTXUSA,Department of NutritionTexas A&M UniversityCollege StationTXUSA
| |
Collapse
|
66
|
Momeny M, Sankanian G, Hamzehlou S, Yousefi H, Esmaeili F, Alishahi Z, Karimi B, Zandi Z, Shamsaiegahkani S, Sabourinejad Z, Kashani B, Nasrollahzadeh A, Mousavipak SH, Mousavi SA, Ghaffari SH. Cediranib, an inhibitor of vascular endothelial growth factor receptor kinases, inhibits proliferation and invasion of prostate adenocarcinoma cells. Eur J Pharmacol 2020; 882:173298. [PMID: 32593665 DOI: 10.1016/j.ejphar.2020.173298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/29/2022]
Abstract
Prostate Cancer is the second cause of cancer-related death in men and development of metastatic castration-resistant prostate cancer (mCRPC) is the major reason for its high mortality rate. Despite various treatments, all patients succumb to resistant disease, suggesting that there is a pressing need for novel and more efficacious treatments. Members of the vascular endothelial growth factor (VEGF) family play key roles in the tumorigenesis of mCRPC, indicating that VEGF-targeted therapies may have potential anti-tumor efficacy in this malignancy. However, due to compensatory activation of other family members, clinical trials with single-targeted VEGF inhibitors were discouraging. Here, we determined the anti-neoplastic activity of Cediranib, a pan-VEGF receptor inhibitor, in the mCRPC cell lines. Anti-growth effects of Cediranib were studied by MTT and BrdU cell proliferation assays and crystal violet staining. Annexin V/PI, radiation therapy and cell motility assays were carried out to examine the effects of Cediranib on apoptosis, radio-sensitivity and cell motility. Quantitative reverse transcription-PCR (qRT-PCR) and Western blot analyses were conducted to determine the molecular mechanisms underlying the anti-tumor activity of Cediranib. Cediranib decreased cell viability and induced apoptosis via inhibition of the anti-apoptotic proteins. Combination with Cediranib synergistically increased Docetaxel sensitivity and potentiated the effects of radiation therapy. Furthermore, Cediranib impaired cell motility via decrease in the expression of the epithelial-to-mesenchymal transition markers. These findings suggest that Cediranib may have anti-tumor activity in mCRPC cells and warrant further investigation on the therapeutic activity of this pan-VEGF receptor inhibitor in mCRPC.
Collapse
Affiliation(s)
- Majid Momeny
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| | - Ghazaleh Sankanian
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Hamzehlou
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Fatemeh Esmaeili
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zivar Alishahi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnaz Karimi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Shamsaiegahkani
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Sabourinejad
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Kashani
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Nasrollahzadeh
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyedeh H Mousavipak
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed A Mousavi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
67
|
Vinogradov AA, Suga H. Introduction to Thiopeptides: Biological Activity, Biosynthesis, and Strategies for Functional Reprogramming. Cell Chem Biol 2020; 27:1032-1051. [PMID: 32698017 DOI: 10.1016/j.chembiol.2020.07.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022]
Abstract
Thiopeptides (also known as thiazolyl peptides) are structurally complex natural products with rich biological activities. Known for over 70 years for potent killing of Gram-positive bacteria, thiopeptides are experiencing a resurgence of interest in the last decade, primarily brought about by the genomic revolution of the 21st century. Every area of thiopeptide research-from elucidating their biological function and biosynthesis to expanding their structural diversity through genome mining-has made great strides in recent years. These advances lay the foundation for and inspire novel strategies for thiopeptide engineering. Accordingly, a number of diverse approaches are being actively pursued in the hope of developing the next generation of natural-product-inspired therapeutics. Here, we review the contemporary understanding of thiopeptide biological activities, biosynthetic pathways, and approaches to structural and functional reprogramming, with a special focus on the latter.
Collapse
Affiliation(s)
- Alexander A Vinogradov
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
68
|
Matsushita K, Toyoda T, Yamada T, Morikawa T, Ogawa K. Comprehensive expression analysis of mRNA and microRNA for the investigation of compensatory mechanisms in the rat kidney after unilateral nephrectomy. J Appl Toxicol 2020; 40:1373-1383. [PMID: 32369870 DOI: 10.1002/jat.3990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 01/02/2023]
Abstract
Compensation is a physiological response that occurs during chemical exposure to maintain homeostasis. Because compensatory responses are not usually considered adverse effects, it is important to understand compensatory mechanisms for chemical risk assessment. Although the kidney is a major target organ for toxicity, there is controversy over whether hyperplasia or hypertrophy contributes to the compensatory mechanism, and there is limited information to apply for chemical risk assessment. In the present study, compensatory mechanisms of the kidney were investigated in a unilateral nephrectomy (UNx) model using adult male and female F344 rats. In residual kidneys of male and female rats after UNx, 5-bromo-2'-deoxyuridine-labeling indices and mRNA expression of cell cycle-related genes were increased, although there were no fluctuations in mRNA expression of transforming growth factor-β1, which contributes to hypertrophy in renal tubules. Pathway analysis using mRNA expression data from a complementary DNA (cDNA) microarray revealed that canonical pathways related to cell proliferation were mainly activated and that forkhead box M1 (FOXM1) was an upstream regulator of compensatory cell proliferation in residual kidneys of male and female rats. cDNA microarray for microRNAs (miRNAs) demonstrated that nine miRNAs were downregulated in residual kidneys, and mRNA/miRNA integrated analysis indicated that miRNAs were associated with the expression of factors downstream of FOXM1. Overall, these results suggested that FOXM1-mediated hyperplasia rather than hypertrophy contributed to compensatory mechanisms in the kidney and that miRNAs regulated downstream FOXM1 signaling. These results will be beneficial for evaluating nephrotoxicity in chemical risk assessment and for developing new biomarkers to predict nephrotoxicity.
Collapse
Affiliation(s)
- Kohei Matsushita
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Takeshi Toyoda
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Takanori Yamada
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan.,Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tomomi Morikawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| |
Collapse
|
69
|
Yun SH, Park JI. Recent progress on the role and molecular mechanism of chicken ovalbumin upstream promoter-transcription factor II in cancer. J Int Med Res 2020; 48:300060520919236. [PMID: 32338091 PMCID: PMC7218465 DOI: 10.1177/0300060520919236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) is an orphan receptor that regulates the expression of genes involved in development and homeostasis. COUP-TFII is also dysregulated in cancer, where it plays important roles in oncogenesis and malignant progression. Recent studies have also investigated altered microRNA-mediated regulation of COUP-TFII in cancer. Although many investigators have studied the expression and clinical significance of COUP-TFII in several cancer types, there remain many controversies regarding its role in these diseases. In this review, we will describe the functions and underlying molecular mechanisms of COUP-TFII in several cancers, especially colorectal, gastric, breast, and prostate cancer; additionally, we will briefly summarize what is known about microRNA-mediated regulation of COUP-TFII.
Collapse
Affiliation(s)
- Seong-Hoon Yun
- Department of Biochemistry, Dong-A University College of Medicine, Busan, Republic of Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
| | - Joo-In Park
- Department of Biochemistry, Dong-A University College of Medicine, Busan, Republic of Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
| |
Collapse
|
70
|
Zhang J, Lu J, Chen Y, Li H, Lin L. WHSC1 promotes wnt/β-catenin signaling in a FoxM1-dependent manner facilitating proliferation, invasion and epithelial-mesenchymal transition in breast cancer. J Recept Signal Transduct Res 2020; 40:410-418. [PMID: 32314642 DOI: 10.1080/10799893.2020.1747490] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objectives: Wolf-Hirschhorn syndrome candidate gene-1 (WHSC1) is highly expressed in various malignant tumors. We investigated the correlation and regulatory pathway of WHSC1 in the progression of breast cancer (BC).Methods: The expression and distribution of WHSC1 in the BC tissues and cell lines were determined by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and immunohistochemical staining. Spearman correlation analysis demonstrated the correlation between WHSC1 high expression level and the clinical characteristics of BC patients. The effects of WHSC1 on the proliferation, apoptosis, migration and invasion of BC cells were analyzed by cell transfection, MTT, colony formation, scratch assay, and transwell. Furthermore, the expression of Forkhead box M1 (FoxM1) and the location of β-catenin were detected by qRT-PCR and western blot.Results: Firstly, WHSC1 expression was up-regulated in BC tissues and cell lines. The high expression of WHSC1 in BC is associated with the tumor size (p = 0.027), metastasis (p = 0.018) and pathological stages (p = 0.025) of the BC patients. The knockdown of WHSC1 inhibited the growth, proliferation migration, invasion and EMT of BC cell lines. Furthermore, WHSC1 could promote the expression of FoxM1 in BC cells and tissues. WHSC1 enhanced the expression of FoxM1, and promoted the nuclear localization of β-catenin, and thus activated the downstream genes expression of Wnt/β-catenin signaling pathway to regulate the development of BC.Conclusion: In summary, our study elucidates the correlation and specific regulatory mechanism between WHSC1 and the progression of BC, thus implying that WHSC1 may function as molecular diagnosis, prognosis and molecular targeted therapy of BC.
Collapse
Affiliation(s)
- Jinfan Zhang
- Department of Breast Surgery, The Affiliated Hospital of Putian University, Putian City, Fujian Province, China
| | - Jingyu Lu
- Department of Breast Surgery, The Affiliated Hospital of Putian University, Putian City, Fujian Province, China
| | - Yu Chen
- Department of Breast Surgery, The Affiliated Hospital of Putian University, Putian City, Fujian Province, China
| | - Hang Li
- Department of Breast Surgery, The Affiliated Hospital of Putian University, Putian City, Fujian Province, China
| | - Lisheng Lin
- Department of Breast Surgery, The Affiliated Hospital of Putian University, Putian City, Fujian Province, China
| |
Collapse
|
71
|
Rankin EB. Genomics and molecular mechanisms of high grade serous ovarian cancer: the 12th Biennial Rivkin Center Ovarian Cancer Research Symposium. Int J Gynecol Cancer 2020; 29:s7-s11. [PMID: 31462542 DOI: 10.1136/ijgc-2019-000490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE The aim of this study was to review current research efforts in genomics and molecular mechanisms of high grade serous ovarian cancer, presented at the 12th Biennial Rivkin Center Ovarian Cancer Research Symposium, held at the University of Washington. METHODS The 12th Biennial Rivkin Center Ovarian Cancer Research Symposium brought together leaders in the field to discuss recent advances in ovarian cancer research and therapy. RESULTS The genomics and molecular mechanisms of ovarian cancer session featured invited speaker presentations by Dr Alan D' Andrea on 'Deoxyribonucleic acid (DNA) repair in ovarian cancer' and Dr Kathleen Cho on 'Modeling the genomics of high grade serous carcinoma in the mouse'. Eight additional oral presentations and 46 poster presentations were selected from the submitted abstracts that highlighted current research efforts in p53, DNA repair, genomic instability and modeling disease in mice, and organoids in high grade serous ovarian cancer. CONCLUSIONS New technologies utilizing clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (CAS9) approaches in mice, organoids, and cell based screens continue to advance our knowledge of key molecular drivers of ovarian cancer initiation, progression, and drug resistance. Improved understanding of the mechanisms of poly ADP ribose polymerase inhibitor resistance may lead to new therapeutic strategies to enhance outcomes in women with high grade serous ovarian cancer.
Collapse
Affiliation(s)
- Erinn B Rankin
- Radiation Oncology and Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| |
Collapse
|
72
|
FOXM1 Deubiquitination by USP21 Regulates Cell Cycle Progression and Paclitaxel Sensitivity in Basal-like Breast Cancer. Cell Rep 2020; 26:3076-3086.e6. [PMID: 30865895 PMCID: PMC6425951 DOI: 10.1016/j.celrep.2019.02.054] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/17/2018] [Accepted: 02/13/2019] [Indexed: 12/23/2022] Open
Abstract
The transcription factor FOXM1 contributes to cell cycle progression and is significantly upregulated in basal-like breast cancer (BLBC). Despite its importance in normal and cancer cell cycles, we lack a complete understanding of mechanisms that regulate FOXM1. We identified USP21 in an RNAi-based screen for deubiquitinases that control FOXM1 abundance. USP21 increases the stability of FOXM1, and USP21 binds and deubiquitinates FOXM1 in vivo and in vitro, indicating a direct enzyme-substrate relationship. Depleting USP21 downregulates the FOXM1 transcriptional network and causes a signifi-cant delay in cell cycle progression. Significantly, USP21 depletion sensitized BLBC cell lines and mouse xenograft tumors to paclitaxel, an anti-mitotic, frontline therapy in BLBC treatment. USP21 is the most frequently amplified deubiquitinase in BLBC patient tumors, and its amplification co-occurs with the upregulation of FOXM1 protein. Altogether, these data suggest a role for USP21 in the proliferation and potentially treatment of FOXM1-high, USP21-high BLBC. The cell cycle transcription factor FOXM1 is activated in basal-like breast cancer (BLBC) and associated with therapeutic resistance and poor patient outcomes. Arceci et al. show USP21 antagonizes FOXM1 degradation, thereby promoting proliferation and paclitaxel resistance. USP21 is catalytically active and recurrently overexpressed in BLBC, representing a potential therapeutic target.
Collapse
|
73
|
Forkhead box M1 transcription factor: a novel target for pulmonary arterial hypertension therapy. World J Pediatr 2020; 16:113-119. [PMID: 31190319 DOI: 10.1007/s12519-019-00271-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Forkhead box M1 (FoxM1), a member of forkhead family, plays a key role in carcinogenesis, progression, invasion, metastasis and drug resistance. Based on the similarities between cancer and pulmonary arterial hypertension, studies on the roles and mechanisms of FoxM1 in pulmonary arterial hypertension have been increasing. This article aims to review recent advances in the mechanisms of signal transduction associated with FoxM1 in pulmonary arterial hypertension. DATA SOURCES Articles were retrieved from PubMed and MEDLINE published after 1990, including-but not limited to-FoxM1 and pulmonary arterial hypertension. RESULTS FoxM1 is overexpressed in pulmonary artery smooth muscle cells in both pulmonary arterial hypertension patients and animal models, and promotes pulmonary artery smooth muscle cell proliferation and inhibits cell apoptosis via regulating cell cycle progression. Multiple signaling molecules and pathways, including hypoxia-inducible factors, transforming growth factor-β/Smad, SET domain-containing 3/vascular endothelial growth factor, survivin, cell cycle regulatory genes and DNA damage response network, are reported to cross talk with FoxM1 in pulmonary arterial hypertension. Proteasome inhibitors are effective in the prevention and treatment of pulmonary arterial hypertension by inhibiting the expression and transcriptional activity of FoxM1. CONCLUSIONS FoxM1 has a crucial role in the pathogenesis of pulmonary arterial hypertension and may represent a novel therapeutic target. But more details of interaction between FoxM1 and other signaling pathways need to be clarified in the future.
Collapse
|
74
|
Abstract
Introduction: FOXM1 is one of the most frequently overexpressed proteins in human solid cancers. Here, we discuss novel direct targets of FOXM1 as well as new pathways involving FOXM1, through which this protein exerts its oncogenic activity.Areas covered: We give a detailed review of FOXM1 transcriptional targets involved in 16 different types of human cancer as published in the literature in the last 5 years. We also discuss a novel positive feedback loop between FOXM1 and AKT - both well-established master regulators of cancer.Expert opinion: Despite the discovery of several FOXM1 inhibitors over the years (by our team and others), their therapeutic use is limited by their adverse off-target effects.Newly-discovered proteins regulated by FOXM1 present a promising alternative approach to target its pro-cancer activity. In addition, targeting regulating proteins that take part in the positive feedback loop between FOXM1/AKT has the double advantage of suppressing both, and can lead to developing novel anti-cancer drugs.
Collapse
Affiliation(s)
- Soheila Borhani
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrei L Gartel
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
75
|
Higurashi M, Maruyama T, Nogami Y, Ishikawa F, Yoshida Y, Mori K, Fujita KI, Shibanuma M. High expression of FOXM1 critical for sustaining cell proliferation in mitochondrial DNA-less liver cancer cells. Exp Cell Res 2020; 389:111889. [PMID: 32032602 DOI: 10.1016/j.yexcr.2020.111889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/06/2020] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
Abstract
The copy number of mitochondrial DNA (mtDNA) is decreased in most cancer types, including hepatocellular carcinoma (HCC), compared to normal counterparts. However, a decrease in mtDNA usually leads to defects in cell proliferation, which contradicts the robustness of cancer cell proliferation. In this study, we found that four out of seven HCC cell lines were of the mtDNA-less type. Interestingly, FOXM1, a member of the FOX transcription factor family, was highly expressed in a subset of them with proliferative potential maintained. B-MYB, a partner of FOXM1, was also expressed in the same cell lines. RNAi-mediated experiments demonstrated that when FOXM1/B-MYB was silenced in the cell lines, cell cycle-related genes were downregulated, while p21Cip1 was induced with senescence-associated β-galactosidase, resulting in G1/S cell cycle arrest. These results suggest that high expression of FOXM1/B-MYB is critical for sustaining cell proliferation in mtDNA-less cells. In addition, we found that high expression of FOXM1 was mediated by the deubiquitinating enzyme, OTUB1, in one cell line. Thus, interference with FOXM1/B-MYB expression, such as through OTUB1 inhibition, may induce a dormant state of senescence-like proliferation arrest in mtDNA-less cancer cells. This finding may be utilized for the development of precision medicine for relevant cancers.
Collapse
Affiliation(s)
- Masato Higurashi
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Tsuyoshi Maruyama
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Yusuke Nogami
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Fumihiro Ishikawa
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Yukiko Yoshida
- Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Kazunori Mori
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Ken-Ichi Fujita
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Motoko Shibanuma
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan.
| |
Collapse
|
76
|
Tang JH, Yang L, Chen JX, Li QR, Zhu LR, Xu QF, Huang GH, Zhang ZX, Xiang Y, Du L, Zhou Z, Lv SQ. Bortezomib inhibits growth and sensitizes glioma to temozolomide (TMZ) via down-regulating the FOXM1-Survivin axis. Cancer Commun (Lond) 2019; 39:81. [PMID: 31796105 PMCID: PMC6892143 DOI: 10.1186/s40880-019-0424-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Background High-grade glioma (HGG) is a fatal human cancer. Bortezomib, a proteasome inhibitor, has been approved for the treatment of multiple myeloma but its use in glioma awaits further investigation. This study aimed to explore the chemotherapeutic effect and the underlying mechanism of bortezomib on gliomas. Methods U251 and U87 cell viability and proliferation were detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, tumor cell spheroid growth, and colony formation assay. Cell apoptosis and cell cycle were detected by flow cytometry. Temozolomide (TMZ)-insensitive cell lines were induced by long-term TMZ treatment, and cells with stem cell characteristics were enriched with stem cell culture medium. The mRNA levels of interested genes were measured via reverse transcription-quantitative polymerase chain reaction, and protein levels were determined via Western blotting/immunofluorescent staining in cell lines and immunohistochemical staining in paraffin-embedded sections. Via inoculating U87 cells subcutaneously, glioma xenograft models in nude mice were established for drug experiments. Patient survival data were analyzed using the Kaplan–Meier method. Results Bortezomib inhibited the viability and proliferation of U251 and U87 cells in a dose- and time-dependent manner by inducing apoptosis and cell cycle arrest. Bortezomib also significantly inhibited the spheroid growth, colony formation, and stem-like cell proliferation of U251 and U87 cells. When administrated in combination, bortezomib showed synergistic effect with TMZ in vitro and sensitized glioma to TMZ treatment both in vitro and in vivo. Bortezomib reduced both the mRNA and protein levels of Forkhead Box M1 (FOXM1) and its target gene Survivin. The FOXM1–Survivin axis was markedly up-regulated in established TMZ-insensitive glioma cell lines and HGG patients. Expression levels of FOXM1 and Survivin were positively correlated with each other and both related to poor prognosis in glioma patients. Conclusions Bortezomib was found to inhibit glioma growth and improved TMZ chemotherapy efficacy, probably via down-regulating the FOXM1–Survivin axis. Bortezomib might be a promising agent for treating malignant glioma, alone or in combination with TMZ.
Collapse
Affiliation(s)
- Jun-Hai Tang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Lin Yang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Ju-Xiang Chen
- Department of Neurosurgery, Changzheng Hospital and Shanghai Institute of Neurosurgery, Second Military Medical University, Shanghai, 200003, P. R. China
| | - Qing-Rui Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Li-Rong Zhu
- Department of Ultrasound, Children Hospital, Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Qing-Fu Xu
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Guo-Hao Huang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Zuo-Xin Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Yan Xiang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Lei Du
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Zheng Zhou
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China.
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China.
| |
Collapse
|
77
|
Shi Y, Li Y, Yan C, Su H, Ying K. Identification of key genes and evaluation of clinical outcomes in lung squamous cell carcinoma using integrated bioinformatics analysis. Oncol Lett 2019; 18:5859-5870. [PMID: 31788059 PMCID: PMC6865087 DOI: 10.3892/ol.2019.10933] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/02/2019] [Indexed: 12/26/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Despite progress in the treatment of non-small-cell lung cancer, there are limited treatment options for lung squamous cell carcinoma (LUSC), compared with lung adenocarcinoma. The present study investigated the disease mechanism of LUSC in order to identify key candidate genes for diagnosis and therapy. A total of three gene expression profiles (GSE19188, GSE21933 and GSE74706) were analyzed using GEO2R to identify common differentially expressed genes (DEGs). The DEGs were then investigated using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. A protein-protein interaction (PPI) network was constructed via the Search Tool for the Retrieval of Interacting Genes/Proteins, and visualized using Cytoscape software. The expression levels of the hub genes identified using CytoHubba were validated using the University of California, Santa Cruz (UCSC) database and the Human Protein Atlas. A Kaplan-Meier curve and Gene Expression Profiling Interactive Analysis were then employed to evaluate the associated prognosis and clinical pathological stage of the hub genes. Furthermore, non-coding RNA regulatory networks were constructed using the Gene-Cloud Biotechnology information website. A total of 359 common DEGs (155 upregulated and 204 downregulated) were identified, which were predominantly enriched in 'mitotic nuclear division', 'cell division', 'cell cycle' and 'p53 signaling pathway'. The PPI network consisted of 257 nodes and 2,772 edges, and the most significant module consisted of 66 upregulated genes. A total of 19 hub genes exhibited elevated RNA levels, and 10 hub genes had elevated protein levels compared with normal lung tissues. The upregulation of five hub genes (CCNB1, CEP55, FOXM1, MKI67 and TYMS; defined in Table I) were significantly associated with poor overall survival and unfavorable clinical pathological stages. Various ncRNAs, such as C1orf220, LINC01561 and MGC39584, may also play important roles in hub-gene regulation. In conclusion, the present study provides further understanding of the pathogenesis of LUSC, and reveals CCNB1, CEP55, FOXM1, MKI67 and TYMS as potential biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Yangfeng Shi
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Yeping Li
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Chao Yan
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Hua Su
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Kejing Ying
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
78
|
Huang J, Shen G, Ren H, Zhang Z, Yu X, Zhao W, Shang Q, Cui J, Yu P, Peng J, Liang D, Yang Z, Jiang X. Role of forkhead box gene family in bone metabolism. J Cell Physiol 2019; 235:1986-1994. [DOI: 10.1002/jcp.29178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Jinjing Huang
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Gengyang Shen
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Hui Ren
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
- Department of Spinal Surgery The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Qi Shang
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Jianchao Cui
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Peiyuan Yu
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - Jiancheng Peng
- Guangzhou University of Chinese Medicine Guangzhou China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
| | - De Liang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
- Department of Spinal Surgery The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou China
| | - Zhidong Yang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
- Department of Spinal Surgery The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou China
| | - Xiaobing Jiang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine Guangzhou China
- Department of Spinal Surgery The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou China
| |
Collapse
|
79
|
Dai Z, Zhu MM, Peng Y, Jin H, Machireddy N, Qian Z, Zhang X, Zhao YY. Endothelial and Smooth Muscle Cell Interaction via FoxM1 Signaling Mediates Vascular Remodeling and Pulmonary Hypertension. Am J Respir Crit Care Med 2019; 198:788-802. [PMID: 29664678 DOI: 10.1164/rccm.201709-1835oc] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Angioproliferative vasculopathy is a hallmark of pulmonary arterial hypertension (PAH). However, little is known about how endothelial cell (EC) and smooth muscle cell (SMC) crosstalk regulates the angioproliferative vascular remodeling. OBJECTIVES To investigate the role of EC and SMC interaction and underlying signaling pathways in pulmonary hypertension (PH) development. METHODS SMC-specific Foxm1 (forkhead box M1) or Cxcr4 knockout mice, EC-specific Foxm1 or Egln1 knockout mice, and EC-specific Egln1/Cxcl12 double knockout mice were used to assess the role of FoxM1 on SMC proliferation and PH. Lung tissues and cells from patients with PAH were used to validate clinical relevance. FoxM1 inhibitor thiostrepton was used in Sugen 5416/hypoxia- and monocrotaline-challenged rats. MEASUREMENTS AND MAIN RESULTS FoxM1 expression was markedly upregulated in lungs and pulmonary arterial SMCs of patients with idiopathic PAH and four discrete PH rodent models. Mice with SMC- (but not EC-) specific deletion of Foxm1 were protected from hypoxia- or Sugen 5416/hypoxia-induced PH. The upregulation of FoxM1 in SMCs induced by multiple EC-derived factors (PDGF-B, CXCL12, ET-1, and MIF) mediated SMC proliferation. Genetic deletion of endothelial Cxcl12 in Egln1Tie2Cre mice or loss of its cognate receptor Cxcr4 in SMCs in hypoxia-treated mice inhibited FoxM1 expression, SMC proliferation, and PH. Accordingly, pharmacologic inhibition of FoxM1 inhibited severe PH in both Sugen 5416/hypoxia and monocrotaline-challenged rats. CONCLUSIONS Multiple factors derived from dysfunctional ECs induced FoxM1 expression in SMCs and activated FoxM1-dependent SMC proliferation, which contributes to pulmonary vascular remodeling and PH. Thus, targeting FoxM1 signaling represents a novel strategy for treatment of idiopathic PAH.
Collapse
Affiliation(s)
- Zhiyu Dai
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Maggie M Zhu
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Yi Peng
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Hua Jin
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Narsa Machireddy
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - Zhijian Qian
- 5 Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Xianming Zhang
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| | - You-Yang Zhao
- 1 Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,2 Division of Critical Care, Department of Pediatrics.,6 Department of Pharmacology, and.,7 Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,3 Department of Pharmacology and.,4 The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois; and
| |
Collapse
|
80
|
Stenmark KR, Hu CJ, Pullamsetti SS. How Many FOXs Are There on The Road to Pulmonary Hypertension? Am J Respir Crit Care Med 2019; 198:704-707. [PMID: 29694238 DOI: 10.1164/rccm.201804-0702ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Kurt R Stenmark
- 1 Department of Pediatrics.,2 Department of Medicine.,3 Cardiovascular Research Laboratories University of Colorado Anschutz Medical Campus Aurora, Colorado
| | - Cheng-Jun Hu
- 3 Cardiovascular Research Laboratories University of Colorado Anschutz Medical Campus Aurora, Colorado.,4 Department of Craniofacial Biology
| | - Soni S Pullamsetti
- 5 Department of Lung Development and Remodeling Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research Bad Nauheim, Germany.,6 Department of Internal Medicine Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research Giessen, Germany and.,7 Justus-Liebig University Giessen, Germany
| |
Collapse
|
81
|
Abstract
BACKGROUND The Hippo/YAP signaling pathway is a central regulator of organ growth and cell proliferation. Activation of the transcriptional co-activator and oncogene YAP (yes-associated protein) supports the development of liver cancer. AIMS The aim of this work was to analyze the molecular mechanisms which are responsible for YAP-induced hepatocarcinogenesis. METHODS YAP was silenced using siRNAs in liver cancer cell lines and effects on target gene expression were analyzed via real-time polymerase chain reaction (PCR) and western immunoblotting. Immunoprecipitation and chromatin immunoprecipitation was used to study interacting proteins and binding to target gene promoter regions, respectively. Transgenic mice with liver-specific and inducible YAP expression were used for in vivo analysis. Gene expression data from hepatocellular carcinoma (HCC) patients were used to analyze YAP-dependent gene signatures and to correlate with clinical data. HCC tissue microarrays were analyzed using immunohistochemistry. RESULTS Together with the transcription factors TEAD4 and FOXM1, YAP induces the expression of genes which are responsible for the development of chromosomal instability (CIN). The overexpression of these CIN genes characterizes liver cancer patients with a poor prognosis. Mechanistically, YAP/TEAD4 and FOXM1 bind to the promoter regions of the CIN genes to directly regulate their expression. The treatment of YAP-transgenic mice with a specific FOXM1 inhibitor reduces the YAP-dependent hepatomegaly, CIN gene expression and CIN. The analysis of human HCC tissue samples confirms the statistical correlation between YAP, FOXM1 and CIN. DISCUSSION These results reveal a new oncogenic mechanism of the Hippo/YAP signaling pathway and identify YAP and FOXM1 as potential targets for targeted therapies.
Collapse
Affiliation(s)
- S Weiler
- Pathologisches Institut, Universität Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Deutschland.
| |
Collapse
|
82
|
Yang N, Wang C, Wang J, Wang Z, Huang D, Yan M, Kamran M, Liu Q, Xu B. Aurora kinase A stabilizes FOXM1 to enhance paclitaxel resistance in triple-negative breast cancer. J Cell Mol Med 2019; 23:6442-6453. [PMID: 31359594 PMCID: PMC6714217 DOI: 10.1111/jcmm.14538] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/21/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has a relatively poor outcome. Acquired chemoresistance is a major clinical challenge for TNBC patients. Previously, we reported that kinase-dead Aurora kinase A (Aurora-A) could effectively transactivate the FOXM1 promoter. Here, we demonstrate an additional pathway through which Aurora-A stabilizes FOXM1 by attenuating its ubiquitin in TNBC. Specifically, Aurora-A stabilizes FOXM1 in late M phase and early G1 phase of the cell cycle, which promotes proliferation of TNBC cells. Knock-down of Aurora-A significantly suppresses cell proliferation in TNBC cell lines and can be rescued by FOXM1 overexpression. We observe that paclitaxel-resistant TNBC cells exhibit high expression of Aurora-A and FOXM1. Overexpression of Aurora-A offers TNBC cells an additional growth advantage and protection against paclitaxel. Moreover, Aurora-A and FOXM1 could be simultaneously targeted by thiostrepton. Combination of thiostrepton and paclitaxel treatment reverses paclitaxel resistance and significantly inhibits cell proliferation. In conclusion, our study reveals additional mechanism through which Aurora-A regulates FOXM1 and provides a new therapeutic strategy to treat paclitaxel-resistant triple-negative breast cancer.
Collapse
Affiliation(s)
- Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chang Wang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jian Wang
- Department of Pathology, GanZhou Municipal People's Hospital, NanChang University, GanZhou, China
| | - Zifeng Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Di Huang
- Department of Breast Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Min Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Muhammad Kamran
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Quentin Liu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - BangLao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
83
|
Shahoumi LA, Yeudall WA. Targeted therapies for non-HPV-related head and neck cancer: challenges and opportunities in the context of predictive, preventive, and personalized medicine. EPMA J 2019; 10:291-305. [PMID: 31462945 DOI: 10.1007/s13167-019-00177-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/04/2019] [Indexed: 12/19/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) develops in the mucosal lining of the upper aerodigestive tract, principally as a result of exposure to carcinogens present in tobacco products and alcohol, with oncogenic papillomaviruses also being recognized as etiological agents in a limited proportion of cases. As such, there is considerable scope for prevention of disease development and progression. However, despite multimodal approaches to treatment, tumor recurrence and metastatic disease are common problems, and clinical outcome is unsatisfactory. As our understanding of the genetics and biochemical aberrations in HNSCC has improved, so the development and use of molecularly targeted drugs to combat the disease have come to the fore. In this article, we review molecular mechanisms that alter signal transduction downstream of the epidermal growth factor receptor (EGFR) as well as those that perturb orderly cell cycle progression, such as p53 mutation, cyclin overexpression, and loss of cyclin-dependent kinase inhibitor function. We outline some of the tactics that have been employed to combat the altered biochemistry. These include blockade of the EGFR using humanized monoclonal antibodies such as cetuximab and small molecule tyrosine kinase inhibitors (TKIs) such as erlotinib/gefitinib and subsequent generations of TKIs, restoration of p53 function using MIRA compounds, and inhibition of cyclin-dependent kinase and aurora kinase activity using drugs such as palbociclib and alisertib. Knowledge of the underlying molecular mechanisms may be utilizable in order to predict disease behavior and tailor therapeutic interventions in a more personalized approach to improve clinical response. Use of liquid biopsy, omics platforms, and salivary diagnostics hold promise in this regard.
Collapse
Affiliation(s)
- Linah A Shahoumi
- 1Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912 USA.,2The Graduate School, Augusta University, Augusta, GA USA
| | - W Andrew Yeudall
- 1Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912 USA.,2The Graduate School, Augusta University, Augusta, GA USA.,3Georgia Cancer Center, Augusta University, Augusta, GA USA
| |
Collapse
|
84
|
Zanin R, Pegoraro S, Ros G, Ciani Y, Piazza S, Bossi F, Bulla R, Zennaro C, Tonon F, Lazarevic D, Stupka E, Sgarra R, Manfioletti G. HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:313. [PMID: 31311575 PMCID: PMC6636010 DOI: 10.1186/s13046-019-1307-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
Background Breast cancer is the most common malignancy in women worldwide. Among the breast cancer subtypes, triple-negative breast cancer (TNBC) is the most aggressive and the most difficult to treat. One of the master regulators in TNBC progression is the architectural transcription factor HMGA1. This study aimed to further explore the HMGA1 molecular network to identify molecular mechanisms involved in TNBC progression. Methods RNA from the MDA-MB-231 cell line, silenced for HMGA1 expression, was sequenced and, with a bioinformatic analysis, molecular partners HMGA1 could cooperate with in regulating common downstream gene networks were identified. Among the putative partners, the FOXM1 transcription factor was selected. The relationship occurring between HMGA1 and FOXM1 was explored by qRT-PCR, co-immunoprecipitation and protein stability assays. Subsequently, the transcriptional activity of HMGA1 and FOXM1 was analysed by luciferase assay on the VEGFA promoter. The impact on angiogenesis was assessed in vitro, evaluating the tube formation ability of endothelial cells exposed to the conditioned medium of MDA-MB-231 cells silenced for HMGA1 and FOXM1 and in vivo injecting MDA-MB-231 cells, silenced for the two factors, in zebrafish larvae. Results Here, we discover FOXM1 as a novel molecular partner of HMGA1 in regulating a gene network implicated in several breast cancer hallmarks. HMGA1 forms a complex with FOXM1 and stabilizes it in the nucleus, increasing its transcriptional activity on common target genes, among them, VEGFA, the main inducer of angiogenesis. Furthermore, we demonstrate that HMGA1 and FOXM1 synergistically drive breast cancer cells to promote tumor angiogenesis both in vitro in endothelial cells and in vivo in a zebrafish xenograft model. Moreover, using a dataset of breast cancer patients we show that the co-expression of HMGA1, FOXM1 and VEGFA is a negative prognostic factor of distant metastasis-free survival and relapse-free survival. Conclusions This study reveals FOXM1 as a crucial interactor of HMGA1 and proves that their cooperative action supports breast cancer aggressiveness, by promoting tumor angiogenesis. Therefore, the possibility to target HMGA1/FOXM1 in combination should represent an attractive therapeutic option to counteract breast cancer angiogenesis. Electronic supplementary material The online version of this article (10.1186/s13046-019-1307-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rossella Zanin
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Silvia Pegoraro
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy.
| | - Gloria Ros
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Yari Ciani
- Laboratorio Nazionale CIB, Area Science Park, Padriciano 99, Trieste, Italy.,Present address: Department of Cellular, Computational and Integrative Biology - (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Silvano Piazza
- Department of Cellular, Computational and Integrative Biology - (CIBIO), University of Trento, Via Sommarive 9, 38123, Povo, Trento, Italy
| | - Fleur Bossi
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) "Burlo Garofolo", via dell'Istria 65/1, 34134, Trieste, Italy
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Cristina Zennaro
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149, Trieste, Italy
| | - Federica Tonon
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149, Trieste, Italy
| | - Dejan Lazarevic
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elia Stupka
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Present address: Life Sciences Business Health Catalyst, Cambridge, Via Sommarive 9, 38123, USA
| | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | | |
Collapse
|
85
|
Mittermair E, Krenn L, Marian B. Prenylated xanthones from Metaxya rostrata suppress FoxM1 and induce active cell death by distinct mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152912. [PMID: 30979690 DOI: 10.1016/j.phymed.2019.152912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Metaxya rostrata C.Presl (Metaxyaceae) is a tree fern widespread in Central and South America and the dried rhizome is used in ethnic medicine against intestinal ulcers or tumors. An activity-guided isolation resulted in two structurally related xanthones: 2-deprenyl-rheediaxanthone B (XB) and 2-deprenyl-7-hydroxy-rheediaxanthone B (OH-XB). HYPOTHESIS/PURPOSE This study analyzed the cytotoxic activity and underlying cellular mechanisms of OH-XB for the first time in comparison to XB. METHODS We exposed the colorectal cancer cell line SW480 and F331 fibroblasts to XB and OH-XB and determined cell viability by neutral red uptake and nuclear morphology by staining with Hoechst dye. Cell cycle distribution and the mechanism of cell death were analyzed by FACS and western blot. Knockdown of FoxM1 expression was performed with siRNA. RESULTS OH-XB was at least as cytotoxic as XB in the induction of cell cycle arrest and active cell death. While both compounds strongly inhibited the transcription factor FoxM1, the cellular mechanisms of growth arrest and cell death induction differed widely: OH-XB induced S-phase cell cycle arrest in contrast to a G2-M-phase arrest by XB. It caused morphological modifications typical for classical apoptosis with increased caspase 7 activity and enhanced cleavage of PARP, while XB caused caspase 2 activation and mitotic catastrophe. After knockdown of FoxM1 expression no induction of caspase activity could be observed. CONCLUSION In summary, our data clearly showed that XB and OH-XB are promising new lead compounds for cancer therapy with distinct cellular mechanisms. Both compounds are candidates for further pre-clinical and clinical investigations.
Collapse
Affiliation(s)
- Eva Mittermair
- Medical University Vienna, Department of Medicine I, Institute of Cancer Research, Borschkegasse 8a, 1090 Vienna, Austria; University of Vienna, Department of Pharmacognosy, Althanstraße 14, 1090 Vienna, Austria
| | - Liselotte Krenn
- University of Vienna, Department of Pharmacognosy, Althanstraße 14, 1090 Vienna, Austria
| | - Brigitte Marian
- Medical University Vienna, Department of Medicine I, Institute of Cancer Research, Borschkegasse 8a, 1090 Vienna, Austria.
| |
Collapse
|
86
|
Hu G, Yan Z, Zhang C, Cheng M, Yan Y, Wang Y, Deng L, Lu Q, Luo S. FOXM1 promotes hepatocellular carcinoma progression by regulating KIF4A expression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:188. [PMID: 31072351 PMCID: PMC6507024 DOI: 10.1186/s13046-019-1202-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/30/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Forkhead box M1 (FOXM1) is a proliferation-associated transcription factor of the forkhead box proteins superfamily, which includes four isoforms FOXM1a, b, c, and d. FOXM1 has been implicated in hepatocellular carcinoma (HCC) progression, but the underlying molecular mechanism remains elusive. In this study, we aim to clarify the molecular basis for FOXM1-mediated HCC progression. METHODS Bioinformatic analysis was used to explore the differentially expressed genes predicting HCC proliferation. The expression of FOXM1 and kinesin family member (KIF)4A was confirmed by western blotting and immunohistochemistry in HCC tissues. Kaplan-Meier survival analysis was conducted to analyze the clinical impact of FOXM1 and KIF4A on HCC. The effect of FOXM1 on the regulation of KIF4A expression was studied in cell biology experiments. The interaction between KIF4A and FOXM1 was analyzed by chromatin immunoprecipitation and luciferase experiments. A series of experiments was performed to explore the functions of FOXM1/KIF4A in HCC progression, such as cell proliferation, cell growth, cell viability, and cell cycle. A xenograft mouse model was used to explore the regulatory effect of FOXM1-KIF4A axis on HCC tumor growth. RESULTS FOXM1 and KIF4A were overexpressed in human primary HCC tissues compared to that in matched adjacent normal liver tissue and are significant risk factors for HCC recurrence and shorter survival. We found that KIF4A was dominantly regulated by FOXM1c among the four isoforms, and further identified KIF4A as a direct downstream target of FOXM1c. Inhibiting FOXM1 decreased KIF4A expression in HCC cells, whereas its overexpression had the opposite effect. FOXM1-induced HCC cell proliferation was dependent on elevated KIF4A expression as KIF4A knockdown abolished FOXM1-induced proliferation of HCC cells both in vitro and in vivo. CONCLUSION The FOXM1-KIF4A axis mediates human HCC progression and is a potential therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Guohui Hu
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, 17 Yongwai Street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Zhengwei Yan
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, 17 Yongwai Street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Cheng Zhang
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, 17 Yongwai Street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Minzhang Cheng
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, 17 Yongwai Street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Yehong Yan
- Department of General Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yiting Wang
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, 17 Yongwai Street, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Libin Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Quqin Lu
- Department of Epidemiology & Biostatistics, School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Shiwen Luo
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China. .,Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, 17 Yongwai Street, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
87
|
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: 7.0] [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
|
88
|
Barger CJ, Branick C, Chee L, Karpf AR. Pan-Cancer Analyses Reveal Genomic Features of FOXM1 Overexpression in Cancer. Cancers (Basel) 2019; 11:cancers11020251. [PMID: 30795624 PMCID: PMC6406812 DOI: 10.3390/cancers11020251] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 01/22/2023] Open
Abstract
FOXM1 is frequently overexpressed in cancer, but this has not been studied in a comprehensive manner. We utilized genotype-tissue expression (GTEx) normal and The Cancer Genome Atlas (TCGA) tumor data to define FOXM1 expression, including its isoforms, and to determine the genetic alterations that promote FOXM1 expression in cancer. Additionally, we used human fallopian tube epithelial (FTE) cells to dissect the role of Retinoblastoma (Rb)-E2F and Cyclin E1 in FOXM1 regulation, and a novel human embryonic kidney cell (HEK293T) CRISPR FOXM1 knockout model to define isoform-specific transcriptional programs. FOXM1 expression, at the mRNA and protein level, was significantly elevated in tumors with FOXM1 amplification, p53 inactivation, and Rb-E2F deregulation. FOXM1 expression was remarkably high in testicular germ cell tumors (TGCT), high-grade serous ovarian cancer (HGSC), and basal breast cancer (BBC). FOXM1 expression in cancer was associated with genomic instability, as measured using aneuploidy signatures. FTE models confirmed a role for Rb-E2F signaling in FOXM1 regulation and in particular identified Cyclin E1 as a novel inducer of FOXM1 expression. Among the three FOXM1 isoforms, FOXM1c showed the highest expression in normal and tumor tissues and cancer cell lines. The CRISPR knockout model demonstrated that FOXM1b and FOXM1c are transcriptionally active, while FOXM1a is not. Finally, we were unable to confirm the existence of a FOXM1 auto-regulatory loop. This study provides significant and novel information regarding the frequency, causes, and consequences of elevated FOXM1 expression in human cancer.
Collapse
Affiliation(s)
- Carter J Barger
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Connor Branick
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Linda Chee
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Adam R Karpf
- Eppley Institute for Cancer Research, University of Nebraska Medical Center, Omaha, NE 68198, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
89
|
Werwein E, Cibis H, Hess D, Klempnauer KH. Activation of the oncogenic transcription factor B-Myb via multisite phosphorylation and prolyl cis/trans isomerization. Nucleic Acids Res 2019; 47:103-121. [PMID: 30321399 PMCID: PMC6326806 DOI: 10.1093/nar/gky935] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/01/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022] Open
Abstract
The oncogenic transcription factor B-Myb is an essential regulator of late cell cycle genes whose activation by phosphorylation is still poorly understood. We describe a stepwise phosphorylation mechanism of B-Myb, which involves sequential phosphorylations mediated by cyclin-dependent kinase (Cdk) and Polo-like kinase 1 (Plk1) and Pin1-facilitated peptidyl-prolyl cis/trans isomerization. Our data suggest a model in which initial Cdk-dependent phosphorylation of B-Myb enables subsequent Pin1 binding and Pin1-induced conformational changes of B-Myb. This, in turn, initiates further phosphorylation of Cdk-phosphosites, enabling Plk1 docking and subsequent Plk1-mediated phosphorylation of B-Myb to finally allow B-Myb to stimulate transcription of late cell cycle genes. Our observations reveal novel mechanistic hierarchies of B-Myb phosphorylation and activation and uncover regulatory principles that might also apply to other Myb family members. Strikingly, overexpression of B-Myb and of factors mediating its activation strongly correlates with adverse prognoses for tumor patients, emphasizing B-Myb's role in tumorigenesis.
Collapse
Affiliation(s)
- Eugen Werwein
- Institute for Biochemistry Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Hannah Cibis
- Institute for Biochemistry Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Daniel Hess
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstr. 66, CH-4058 Basel, Switzerland
| | - Karl-Heinz Klempnauer
- Institute for Biochemistry Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| |
Collapse
|
90
|
Laissue P. The forkhead-box family of transcription factors: key molecular players in colorectal cancer pathogenesis. Mol Cancer 2019; 18:5. [PMID: 30621735 PMCID: PMC6325735 DOI: 10.1186/s12943-019-0938-x] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/01/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly occurring cancer worldwide and the fourth most frequent cause of death having an oncological origin. It has been found that transcription factors (TF) dysregulation, leading to the significant expression modifications of genes, is a widely distributed phenomenon regarding human malignant neoplasias. These changes are key determinants regarding tumour’s behaviour as they contribute to cell differentiation/proliferation, migration and metastasis, as well as resistance to chemotherapeutic agents. The forkhead box (FOX) transcription factor family consists of an evolutionarily conserved group of transcriptional regulators engaged in numerous functions during development and adult life. Their dysfunction has been associated with human diseases. Several FOX gene subgroup transcriptional disturbances, affecting numerous complex molecular cascades, have been linked to a wide range of cancer types highlighting their potential usefulness as molecular biomarkers. At least 14 FOX subgroups have been related to CRC pathogenesis, thereby underlining their role for diagnosis, prognosis and treatment purposes. This manuscript aims to provide, for the first time, a comprehensive review of FOX genes’ roles during CRC pathogenesis. The molecular and functional characteristics of most relevant FOX molecules (FOXO, FOXM1, FOXP3) have been described within the context of CRC biology, including their usefulness regarding diagnosis and prognosis. Potential CRC therapeutics (including genome-editing approaches) involving FOX regulation have also been included. Taken together, the information provided here should enable a better understanding of FOX genes’ function in CRC pathogenesis for basic science researchers and clinicians.
Collapse
Affiliation(s)
- Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 N° 63C-69, Bogotá, Colombia.
| |
Collapse
|
91
|
Zhou Y, Wang Q, Chu L, Dai W, Zhang X, Chen J, Zhang L, Ding P, Zhang X, Gu H, Zhang P, Li L, Zhang W, Li L, Lv X, Zhou D, Cai G, Chen L, Zhao K, Hu W. FOXM1c promotes oesophageal cancer metastasis by transcriptionally regulating IRF1 expression. Cell Prolif 2018; 52:e12553. [PMID: 30485581 PMCID: PMC6496730 DOI: 10.1111/cpr.12553] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 09/14/2018] [Accepted: 10/21/2018] [Indexed: 12/15/2022] Open
Abstract
Objectives We aimed to elucidate the role and molecular mechanisms of FOXM1 in regulating metastasis in oesophageal squamous cell carcinoma (ESCC) as well as its clinical implications. Materials and methods The expression levels of four isoforms of FOXM1 were analysed by real‐time PCR. Next, genetically modification using overexpression and RNAi systems and transwell were employed to examine FOXM1c function in invasion and migration. Dual luciferase and ChIP assays were performed to decipher the underlying mechanism for transcriptional regulation. The expression levels of FOXM1 and IRF1 were determined by immunohistochemistry staining in ESCC specimens. Results The FOXM1c was predominantly overexpressed in ESCC cell lines compared to the other FOXM1 isoforms. Ectopic expression of FOXM1c promoted invasion and migration of ESCC cells lines, whereas downregulation of FOXM1c inhibited these processes. Moreover, FOXM1c expression was positively correlated with IRF1 expression in ESCC cell lines and tumour specimens. IRF1 is, at least in part, responsible for FOXM1c‐mediated invasion and migration. Mechanistically, we identified IRF1 as a transcriptional target of FOXM1c and found a FOXM1c‐binding site in the IRF1 promoter region. Furthermore, high expression levels of both FOXM1c and IRF1 were positively associated with low survival rate and predicted a poor prognosis of oesophageal cancer patients. Conclusion FOXM1c promotes the metastasis by transcriptionally targeting IRF1 and may serve as a potential prognostic predictor for oesophageal cancer.
Collapse
Affiliation(s)
- Yuzhen Zhou
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Wang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weixing Dai
- Department of Colorectal Surgery, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaozhou Zhang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianfeng Chen
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Long Zhang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Zhang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyu Gu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Pingzhao Zhang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Li
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Luying Li
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyue Lv
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Danlei Zhou
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Chen
- Key Laboratory of Functional Protein Research of Guangdong Higher Education, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
92
|
Bach DH, Long NP, Luu TTT, Anh NH, Kwon SW, Lee SK. The Dominant Role of Forkhead Box Proteins in Cancer. Int J Mol Sci 2018; 19:E3279. [PMID: 30360388 PMCID: PMC6213973 DOI: 10.3390/ijms19103279] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/16/2022] Open
Abstract
Forkhead box (FOX) proteins are multifaceted transcription factors that are significantly implicated in cancer, with various critical roles in biological processes. Herein, we provide an overview of several key members of the FOXA, FOXC, FOXM1, FOXO and FOXP subfamilies. Important pathophysiological processes of FOX transcription factors at multiple levels in a context-dependent manner are discussed. We also specifically summarize some major aspects of FOX transcription factors in association with cancer research such as drug resistance, tumor growth, genomic alterations or drivers of initiation. Finally, we suggest that targeting FOX proteins may be a potential therapeutic strategy to combat cancer.
Collapse
Affiliation(s)
- Duc-Hiep Bach
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | | | | | - Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Sang Kook Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
93
|
Qiu J, Zhao J, Zuo A, Liu L, Liu Q, Pan H, Yuan X. Lentiviral RNA interference-mediated downregulation of Forkhead box M1 expression suppresses growth of oral squamous cell carcinoma in vitro. Oncol Lett 2018; 17:525-531. [PMID: 30655797 DOI: 10.3892/ol.2018.9536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/10/2018] [Indexed: 11/06/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most fatal types of oral cancer worldwide. Forkhead box M1 (FOXM1) is associated with the occurrence and development of a number of types of human cancer, but its function in OSCC remains unclear. The present study aimed to explore the effect of FOXM1 downregulation using lentivirus-mediated short hairpin (sh)RNA against FOXM1 (LV-shFOXM1) in the cell line Tca8113 in vitro. Infection of Tca8113 cells with LV-shFOXM1 inhibited the mRNA and protein expression level of FOXM1. The downregulation of FOXM1 resulted in cell cycle arrest of Tca8113 cells, and the inhibition of proliferation, migration and invasion. The protein expression level of cyclins B1 and D1 were downregulated, whereas those of p27 and p21 were upregulated following infection with LV-shFOXM1, compared with the blank control and LV-shCON groups. In addition, FOXM1 downregulation decreased the expression of matrix metalloproteinase-2 and LV-shFOXM1 significantly suppressed OSCC cell viability. Therefore, FOXM1 may be a target for the treatment of OSCC.
Collapse
Affiliation(s)
- Jing Qiu
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| | - Juan Zhao
- Department of Pediatrics, Jiaozhou People's Hospital, Qingdao, Shandong 266300, P.R. China
| | - Anjun Zuo
- Medical Services, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lan Liu
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| | - Qiaoqiao Liu
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| | - Huazheng Pan
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiao Yuan
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
94
|
Khan I, Halasi M, Patel A, Schultz R, Kalakota N, Chen YH, Aardsma N, Liu L, Crispino JD, Mahmud N, Frankfurt O, Gartel AL. FOXM1 contributes to treatment failure in acute myeloid leukemia. JCI Insight 2018; 3:121583. [PMID: 30089730 PMCID: PMC6129129 DOI: 10.1172/jci.insight.121583] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/21/2018] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) patients with NPM1 mutations demonstrate a superior response to standard chemotherapy treatment. Our previous work has shown that these favorable outcomes are linked to the cytoplasmic relocalization and inactivation of FOXM1 driven by mutated NPM1. Here, we went on to confirm the important role of FOXM1 in increased chemoresistance in AML. A multiinstitution retrospective study was conducted to link FOXM1 expression to clinical outcomes in AML. We establish nuclear FOXM1 as an independent clinical predictor of chemotherapeutic resistance in intermediate-risk AML in a multivariate analysis incorporating standard clinicopathologic risk factors. Using colony assays, we show a dramatic decrease in colony size and numbers in AML cell lines with knockdown of FOXM1, suggesting an important role for FOXM1 in the clonogenic activity of AML cells. In order to further prove a potential role for FOXM1 in AML chemoresistance, we induced an FLT3-ITD-driven myeloid neoplasm in a FOXM1-overexpressing transgenic mouse model and demonstrated significantly higher residual disease after standard chemotherapy. This suggests that constitutive overexpression of FOXM1 in this model induces chemoresistance. Finally, we performed proof-of-principle experiments using a currently approved proteasome inhibitor, ixazomib, to target FOXM1 and demonstrated a therapeutic response in AML patient samples and animal models of AML that correlates with the suppression of FOXM1 and its transcriptional targets. Addition of low doses of ixazomib increases sensitization of AML cells to chemotherapy backbone drugs cytarabine and the hypomethylator 5-azacitidine. Our results underscore the importance of FOXM1 in AML progression and treatment, and they suggest that targeting it may have therapeutic benefit in combination with standard AML therapies.
Collapse
Affiliation(s)
- Irum Khan
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Marianna Halasi
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | | | - Rachael Schultz
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Nandini Kalakota
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Yi-Hua Chen
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| | - Nathan Aardsma
- Department of Pathology, University of Illinois, Chicago, Illinois, USA
| | - Li Liu
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois, Chicago, Illinois, USA
| | | | - Nadim Mahmud
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | | | - Andrei L Gartel
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
95
|
Gouazé-Andersson V, Ghérardi MJ, Lemarié A, Gilhodes J, Lubrano V, Arnauduc F, Cohen-Jonathan Moyal E, Toulas C. FGFR1/FOXM1 pathway: a key regulator of glioblastoma stem cells radioresistance and a prognosis biomarker. Oncotarget 2018; 9:31637-31649. [PMID: 30167084 PMCID: PMC6114977 DOI: 10.18632/oncotarget.25827] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma are known to be aggressive and therapy-resistant tumors, due to the presence of glioblastoma stem cells inside this heterogeneous tumor. We investigate here the involvement of FGFR1 in glioblastoma stem-like cells (GSLC) radioresistance mechanisms. We first demonstrated that the survival after irradiation was significantly diminished in FGFR1-silenced (FGFR1-) GSLC compared to control GSLC. The transcriptome analysis of GSLCs FGFR1(-) showed that FOX family members are differentially regulated by FGFR1 inhibition, particularly with an upregulation of FOXN3 and a downregulation of FOXM1. GSLC survival after irradiation was significantly increased after FOXN3 silencing and decreased after FOXM1 inhibition, showing opposite effects of FGFR1/FOX family members on cell response to ionizing radiation. Silencing FGFR1 or FOXM1 downregulated genes involved in mesenchymal transition such as GLI2, TWIST1, and ZEB1 in glioblastoma stem-like cells. It also dramatically reduced GSLC migration. Databases analysis confirmed that the combined expression of FGFR1/FOXM1/MELK/GLI2/ZEB1/TWIST1 is significantly associated with patients overall survival after chemo-radiotherapy treatment. All these results, associated with our previous conduced ones with differentiated cells, clearly established that FGFR1-FOXM1 dependent glioblastoma stem-like cells radioresistance pathway is a central actor of GBM treatment resistance and a key target to inhibit in the aim to increase the sensitivity of GBM to the radiotherapy.
Collapse
Affiliation(s)
- Valérie Gouazé-Andersson
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Marie-Julie Ghérardi
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Anthony Lemarié
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Julia Gilhodes
- Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| | - Vincent Lubrano
- CHU PURPAN-Pavillon Baudot, Place du Dr Baylac, Toulouse-Cedex 3, 31024, France
| | - Florent Arnauduc
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France
| | - Elizabeth Cohen-Jonathan Moyal
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France.,Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| | - Christine Toulas
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1037/Université Toulouse III Paul Sabatier, Cancer Research Center of Toulouse (CRCT), Toulouse, F-31000, France.,Institut Claudius Regaud, IUCT-O, Toulouse, F-31059, France
| |
Collapse
|
96
|
Amair-Pinedo F, Matos I, Saurí T, Hernando J, Capdevila J. The Treatment Landscape and New Opportunities of Molecular Targeted Therapies in Gastroenteropancreatic Neuroendocrine Tumors. Target Oncol 2018; 12:757-774. [PMID: 29143176 DOI: 10.1007/s11523-017-0532-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neuroendocrine neoplasms (NENs) are a heterogeneous group of neoplasms that originate from neuroendocrine stem cells and express both neural and endocrine markers. They are found in almost every organ, and while NENs are mostly associated with slow growth, complications due to the uncontrolled secretion of active peptides, and metastatic disease, may significantly impair the quality of life and can ultimately lead to the death of affected individuals. Expanding knowledge of the genetic, epigenetic, and proteomic landscapes of NENs has led to a better understanding of their molecular pathology and consequently increased treatment options for patients. Here, we review the principal breakthroughs in NEN treatment management, owing largely to omics technologies over the last few years, current recommendations of systemic treatment, and ongoing research into the identification of predictive and response biomarkers based on molecular targeted therapies.
Collapse
Affiliation(s)
| | - Ignacio Matos
- Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Tamara Saurí
- Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jorge Hernando
- Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jaume Capdevila
- Vall d'Hebron University Hospital, Barcelona, Spain. .,Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
97
|
Liu L, Zhai C, Pan Y, Zhu Y, Shi W, Wang J, Yan X, Su X, Song Y, Gao L, Li M. Sphingosine-1-phosphate induces airway smooth muscle cell proliferation, migration, and contraction by modulating Hippo signaling effector YAP. Am J Physiol Lung Cell Mol Physiol 2018; 315:L609-L621. [PMID: 29999407 DOI: 10.1152/ajplung.00554.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sphingosine-1-phosphate (S1P), a bioactive lipid, has been shown to be elevated in the airways of individuals with asthma and modulates the airway smooth muscle cell (ASMC) functions, yet its underlying molecular mechanisms are not completely understood. The aim of the present study is to address this issue. S1P induced yes-associated protein (YAP) dephosphorylation and nuclear localization via the S1PR2/3/Rho-associated protein kinase (ROCK) pathway, and this in turn increased forkhead box M1 (FOXM1) and cyclin D1 expression leading to ASMC proliferation, migration, and contraction. Pretreatment of cells with S1PR2 antagonist JTE013, S1PR3 antagonist CAY10444, or ROCK inhibitor Y27632 blocked S1P-induced alterations of YAP, FOXM1, cyclin D1, and ASMC proliferation, migration, and contraction. In addition, prior silencing of YAP or FOXM1 with siRNA reversed the effect of S1P on ASMC functions. Taken together, our study indicates that S1P stimulates ASMC proliferation, migration, and contraction by binding to S1PR2/3 and modulating ROCK/YAP/FOXM1 axis and suggests that targeting this pathway might have potential value in the management of asthma.
Collapse
Affiliation(s)
- Lu Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Cui Zhai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Yilin Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Yanting Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Xiaofan Su
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Yang Song
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| | - Li Gao
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi , People's Republic of China
| |
Collapse
|
98
|
Inhibition of maternal embryonic leucine zipper kinase with OTSSP167 displays potent anti-leukemic effects in chronic lymphocytic leukemia. Oncogene 2018; 37:5520-5533. [PMID: 29895969 DOI: 10.1038/s41388-018-0333-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 11/08/2022]
Abstract
TP53 pathway defects contributed to therapy resistance and adverse clinical outcome in chronic lymphocytic leukemia (CLL), which represents an unmet clinical need with few therapeutic options. Maternal embryonic leucine zipper kinase (MELK) is a novel oncogene, which plays crucial roles in mitotic progression and stem cell maintenance. OTSSP167, an orally administrated inhibitor targeting MELK, is currently in a phase I/II clinical trial in patients with advanced breast cancer and acute myeloid leukemia. Yet, no investigation has been elucidated to date regarding the oncogenic role of MELK and effects of OTSSP167 in chronic lymphocytic leukemia (CLL). Previous studies confirmed MELK inhibition abrogated cancer cell survival via p53 signaling pathway. Thus, we aimed to determine the biological function of MELK and therapeutic potential of OTSSP167 in CLL. Herein, MELK over-expression was observed in CLL cells, and correlated with higher WBC count, advanced stage, elevated LDH, increased β2-MG level, unmutated IGHV, positive ZAP-70, deletion of 17p13 and inferior prognosis of CLL patients. In accordance with functional enrichment analyses in gene expression profiling, CLL cells with depletion or inhibition of MELK exhibited impaired cell proliferation, enhanced fast-onset apoptosis, induced G2/M arrest, attenuated cell chemotaxis and promoted sensitivity to fludarabine and ibrutinib. However, gain-of-function assay showed increased cell proliferation and cell chemotaxis. In addition, OTSSP167 treatment reduced phosphorylation of AKT and ERK1/2. It decreased FoxM1 phosphorylation, expression of FoxM1, cyclin B1 and CDK1, while up-regulating p53 and p21 expression. Taken together, MELK served as a candidate of therapeutic target in CLL. OTSSP167 exhibits potent anti-tumor activities in CLL cells, highlighting a novel molecule-based strategy for leukemic interventions.
Collapse
|
99
|
Zhang Y, Qiao WB, Shan L. Expression and functional characterization of FOXM1 in non-small cell lung cancer. Onco Targets Ther 2018; 11:3385-3393. [PMID: 29928129 PMCID: PMC6001838 DOI: 10.2147/ott.s162523] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objectives FOXM1 is a key member of the FOX transcription factor family, which plays a vital role in a series of physiological processes. In the present study, non-small cell lung cancer (NSCLC) patients and cell lines were studied to explore the correlation between FOXM1 expression and this malignancy. Materials and methods The expression status of FOXM1 was detected in 128 cases of NSCLC tissues and NSCLC cell lines. The relationship of FOXM1 expression and clinicopathological features of NSCLC patients was evaluated by us. In addition, we also explored the biological functions of FOXM1 in NSCLC cell lines. Results The FOXM1 is highly expressed in NSCLC tissues and cell lines. FOXM1 expression was closely correlated with lymph node status and TNM stage. Cox regression analysis were performed to demonstrate the prognosis role of FOXM1. Conclusion FOXM1 conferred a proliferation and invasion advantage to NSCLC cell. The FOXM1 can be regarded as an important molecular marker in NSCLC prognosis.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Thoracic Oncology, Tumor Hospital Affiliated to Xinjiang Medical University, Xinjiang, People's Republic of China
| | - Wen-Bin Qiao
- Department of Thoracic Oncology, Tumor Hospital Affiliated to Xinjiang Medical University, Xinjiang, People's Republic of China
| | - Li Shan
- Department of Thoracic Oncology, Tumor Hospital Affiliated to Xinjiang Medical University, Xinjiang, People's Republic of China
| |
Collapse
|
100
|
Xiao Z, Jia Y, Jiang W, Wang Z, Zhang Z, Gao Y. FOXM1: A potential indicator to predict lymphatic metastatic recurrence in stage IIA esophageal squamous cell carcinoma. Thorac Cancer 2018; 9:997-1004. [PMID: 29877046 PMCID: PMC6068428 DOI: 10.1111/1759-7714.12776] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/03/2018] [Accepted: 05/05/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Previous studies have elucidated that FOXM1 may predict poor prognosis in patients with multiple solid malignant tumors. In this study we explored the differential expression of FOXM1 in stage IIA esophageal squamous cell carcinoma (ESCC) and investigated its prognostic value. METHODS Immunohistochemistry (IHC) and Western blot were used to detect FOXM1 expression in ESCC. Correlations between FOXM1 expression and clinicopathological variables, and five-year lymphatic metastatic recurrence (LMR) and overall survival (OS) of patients were analyzed. RESULTS FOXM1 was aberrantly expressed in ESCC. Statistical analysis revealed a close relationship between FOXM1 expression and tumor size (P = 0.024), depth of invasion (P = 0.048), and degree of differentiation (P = 0.043). The five-year LMR of patients in the FOXM1 overexpression group was significantly increased compared to the low expression group (P = 0.001). The five-year OS of patients in the FOXM1 overexpression group was significantly reduced compared to the low expression group (P = 0.007). Log-rank tests demonstrated that large tumor size (P = 0.044), poor differentiation degree (P = 0.005), deep invasion (P = 0.000), and FOXM1 overexpression (P = 0.007) may indicate poor prognosis in stage IIA ESCC. Cox multivariate regression analysis revealed that all of these variables were independent predictors of unfavorable outcome (P < 0.05). CONCLUSION FOXM1 could be a predictor of lymphatic metastatic recurrence in stage IIA ESCC after Ivor Lewis esophagectomy.
Collapse
Affiliation(s)
- Zhaohua Xiao
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yang Jia
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Wenpeng Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhou Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhiping Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Department of Thoracic Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Yanyun Gao
- Department of Gynaecology and Obstetrics, Jining Traditional Chinese Medicine Hospital, Jining, China
| |
Collapse
|