1
|
Xu R, Bai M, Fan Y, Zhu Y, Wang Z, Hui T, Zhang Q, Liu X, Zhang J, Shen J, Bai W. Knockdown of miR-361-5p promotes the induced activation of SHF-stem cells through FOXM1 mediated Wnt/β-catenin pathway in cashmere goats. Anim Biotechnol 2024; 35:2356110. [PMID: 38804592 DOI: 10.1080/10495398.2024.2356110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The inducing activation event of secondary hair follicle (SHF)-stem cells is considered a key biological process in the SHF regeneration, and the morphogenesis of cashmere fiber in cashmere goats. The miR-361-5p was essentially implicated in the induced activation of SHF-stem cells of cashmere goats, but its functional mechanisms are unclear. Here, we confirmed miR-361-5p was significantly downregulated in anagen SHF bugle of cashmere goats compared with that at telogen, and miR-361-5p expression was significantly lower in SHF-stem cells after activation than its counterpart before activation. Further, we found that miR-361-5p could negatively regulate the induced activation event of SHF-stem cells in cashmere goats. Mechanistically, through dual-luciferase reporter assays, miR-361-5p specifically bound with FOXM1 mRNA in SHF-stem cells of cashmere goats and negatively regulated the expression of FOXM1 gene. Also, through overexpression/knockdown analysis of FOXM1 gene, our results indicated that FOXM1 upregulated the expression of Wnt/β-catenin pathway related genes in SHF-stem cells. Moreover, based on TOP/FOP-flash Wnt report assays, the knockdown of miR-361-5p promotes the Wnt/β-catenin pathway activation through upregulating the FOXM1 expression in SHF-stem cells. Finally, we demonstrated that miR-361-5p negatively regulated the induced activation of SHF-stem cells through FOXM1 mediated Wnt/β-catenin pathway in cashmere goats.
Collapse
Affiliation(s)
- Ruqing Xu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Man Bai
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Yixing Fan
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Yubo Zhu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Zeying Wang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Taiyu Hui
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Qi Zhang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Xingwang Liu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Jialiang Zhang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Jincheng Shen
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| | - Wenlin Bai
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, P. R. China
| |
Collapse
|
2
|
Abusharkh KAN, Comert Onder F, Çınar V, Onder A, Sıkık M, Hamurcu Z, Ozpolat B, Ay M. Novel benzothiazole/benzothiazole thiazolidine-2,4-dione derivatives as potential FOXM1 inhibitors: In silico, synthesis, and in vitro studies. Arch Pharm (Weinheim) 2024:e2400504. [PMID: 39318080 DOI: 10.1002/ardp.202400504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024]
Abstract
The oncogenic transcription factor FOXM1 overexpressed in breast and other solid cancers, is a key driver of tumor growth and progression through complex interactions, making it an attractive molecular target for the development of targeted therapies. Despite the availability of small-molecule inhibitors, their limited specificity, potency, and efficacy hinder clinical translation. To identify effective FOXM1 inhibitors, we synthesized novel benzothiazole derivatives (KC10-KC13) and benzothiazole hybrids with thiazolidine-2,4-dione (KC21-KC36). These compounds were evaluated for FOXM1 inhibition. Molecular docking and molecular dynamics simulation analysis revealed their binding patterns and affinities for the FOXM1-DNA binding domain. The interactions with key amino acids such as Asn283, His287, and Arg286, crucial for FOXM1 inhibition, have been determined with the synthesized compounds. Additionally, the molecular modeling study indicated that KC12, KC21, and KC30 aligned structurally and interacted similarly to the reference compound FDI-6. In vitro studies with the MDA-MB-231 breast cancer cell line demonstrated that KC12, KC21, and KC30 significantly inhibited FOXM1, showing greater potency than FDI-6, with IC50 values of 6.13, 10.77, and 12.86 µM, respectively, versus 20.79 µM for FDI-6. Our findings suggest that KC12, KC21, and KC30 exhibit strong activity as FOXM1 inhibitors and may be suitable for in vivo animal studies.
Collapse
Affiliation(s)
- Khaled A N Abusharkh
- Department of Chemistry, School of Graduate Studies, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
- Department of Chemistry, Natural Products and Drug Research Laboratory, Faculty of Science, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
- Department of Chemistry and Chemical Technology, Faculty of Science and Technology, Al-Quds University, East Jerusalem, Palestine
| | - Ferah Comert Onder
- Department of Medical Biology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Venhar Çınar
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Alper Onder
- Department of Chemistry, Natural Products and Drug Research Laboratory, Faculty of Science, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Merve Sıkık
- Department of Medical System Biology, School of Graduate Studies, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Zuhal Hamurcu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Mehmet Ay
- Department of Chemistry, Natural Products and Drug Research Laboratory, Faculty of Science, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| |
Collapse
|
3
|
Liu Y, Ji J, Zheng S, Wei A, Li D, Shi B, Han X, Chen X. Senescent lung-resident mesenchymal stem cells drive pulmonary fibrogenesis through FGF-4/FOXM1 axis. Stem Cell Res Ther 2024; 15:309. [PMID: 39289765 PMCID: PMC11409797 DOI: 10.1186/s13287-024-03866-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/27/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is an age-related disease featured with abnormal fibrotic response and compromised lung function. Cellular senescence is now considered as an essential driving mechanism for IPF. Given the poor knowledge of the mechanisms underpinning IPF progression, understanding the cellular processes and molecular pathways is critical for developing effective therapies of IPF. METHODS Lung fibrosis was induced using bleomycin in C57BL/6 mice. Cellular senescence was measured by immunofluorescence. The effects of FGF-4 on fibroblast activation markers and signaling molecules were assessed with western blot and qPCR. RESULTS We demonstrated elevated abundance of senescent mesenchymal stem cells (MSCs) in IPF lung tissues, which was tightly correlated with the severity of pulmonary fibrosis in vivo. In addition, senescent MSCs could effectively induce the phenotype of pulmonary fibrosis both in vitro and in vivo. To further confirm how senescent MSCs regulate IPF progression, we demonstrate that FGF-4 is significantly elevated in senescent MSCs, which can induce the activation of pulmonary fibroblasts. In vitro, FGF-4 can activate Wnt signaling in a FOXM1-dependent manner. Inhibition of FOXM1 via thiostrepton effectively impairs FGF-4-induced activation of pulmonary fibroblast and dramatically suppresses the development of pulmonary fibrosis. CONCLUSION These findings reveal that FGF-4 plays a crucial role in senescent MSCs-mediated pulmonary fibrogenesis, and suggests that strategies aimed at deletion of senescent MSCs or blocking the FGF-4/FOXM1 axis could be effective in the therapy of IPF.
Collapse
Affiliation(s)
- Yuxin Liu
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Jie Ji
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Shudan Zheng
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Ai Wei
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Dongmei Li
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Bin Shi
- Pulmonary and Critical Care Medicine, Suqian People's Hospital of Nanjing Gulou Hospital Group, Suqian Scientific Research Institute of Nanjing University Medical School, Nanjing University, Suqian, Jiangsu, 223800, China.
| | - Xiaodong Han
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China.
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Xiang Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China.
- Department of Basic Medical Science, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224008, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Sun Y, Mu G, Zhang X, Wu Y, Wang S, Wang X, Xue Z, Wang C, Liu J, Li W, Zhang L, Guo Y, Zhao F, Liu X, Xue Z, Zhang Y, Ni S, Wang J, Li X, Han M, Huang B. Metabolic modulation of histone acetylation mediated by HMGCL activates the FOXM1/β-catenin pathway in glioblastoma. Neuro Oncol 2024; 26:653-669. [PMID: 38069906 PMCID: PMC10995515 DOI: 10.1093/neuonc/noad232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Altered branched-chain amino acid (BCAA) metabolism modulates epigenetic modification, such as H3K27ac in cancer, thus providing a link between metabolic reprogramming and epigenetic change, which are prominent hallmarks of glioblastoma multiforme (GBM). Here, we identified mitochondrial 3-hydroxymethyl-3-methylglutaryl-CoA lyase (HMGCL), an enzyme involved in leucine degradation, promoting GBM progression and glioma stem cell (GSC) maintenance. METHODS In silico analysis was performed to identify specific molecules involved in multiple processes. Glioblastoma multiforme cells were infected with knockdown/overexpression lentiviral constructs of HMGCL to assess malignant performance in vitro and in an orthotopic xenograft model. RNA sequencing was used to identify potential downstream molecular targets. RESULTS HMGCL, as a gene, increased in GBM and was associated with poor survival in patients. Knockdown of HMGCL suppressed proliferation and invasion in vitro and in vivo. Acetyl-CoA was decreased with HMGCL knockdown, which led to reduced NFAT1 nuclear accumulation and H3K27ac level. RNA sequencing-based transcriptomic profiling revealed FOXM1 as a candidate downstream target, and HMGCL-mediated H3K27ac modification in the FOXM1 promoter induced transcription of the gene. Loss of FOXM1 protein with HMGCL knockdown led to decreased nuclear translocation and thus activity of β-catenin, a known oncogene. Finally, JIB-04, a small molecule confirmed to bind to HMGCL, suppressed GBM tumorigenesis in vitro and in vivo. CONCLUSIONS Changes in acetyl-CoA levels induced by HMGCL altered H3K27ac modification, which triggers transcription of FOXM1 and β-catenin nuclear translocation. Targeting HMGCL by JIB-04 inhibited tumor growth, indicating that mediators of BCAA metabolism may serve as molecular targets for effective GBM treatment.
Collapse
Affiliation(s)
- Yanfei Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guangjing Mu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuehai Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yibo Wu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Shuai Wang
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, New York, USA
| | - Xu Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhiwei Xue
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuanwei Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jilong Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Wenbo Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Lin Zhang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, China
| | - Yunyun Guo
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Feihu Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xuemeng Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Zhiyi Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| |
Collapse
|
5
|
Merjaneh N, Hajjar M, Lan YW, Kalinichenko VV, Kalin TV. The Promise of Combination Therapies with FOXM1 Inhibitors for Cancer Treatment. Cancers (Basel) 2024; 16:756. [PMID: 38398147 PMCID: PMC10886945 DOI: 10.3390/cancers16040756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/21/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Forkhead box M1 (FOXM1) is a transcription factor in the forkhead (FOX) family, which is required for cellular proliferation in normal and neoplastic cells. FOXM1 is highly expressed in many different cancers, and its expression is associated with a higher tumor stage and worse patient-related outcomes. Abnormally high expression of FOXM1 in cancers compared to normal tissue makes FOXM1 an attractive target for pharmacological inhibition. FOXM1-inhibiting agents and specific FOXM1-targeted small-molecule inhibitors have been developed in the lab and some of them have shown promising efficacy and safety profiles in mouse models. While the future goal is to translate FOXM1 inhibitors to clinical trials, potential synergistic drug combinations can maximize anti-tumor efficacy while minimizing off-target side effects. Hence, we discuss the rationale and efficacy of all previously studied drug combinations with FOXM1 inhibitors for cancer therapies.
Collapse
Affiliation(s)
- Nawal Merjaneh
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, Division of Hematology and Oncology, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Mona Hajjar
- The Columbian College of Arts and Sciences, George Washington University, Washington, DC 20052, USA;
| | - Ying-Wei Lan
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
| | - Vladimir V. Kalinichenko
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
- Division of Neonatology, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
| | - Tanya V. Kalin
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, Division of Hematology and Oncology, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
| |
Collapse
|
6
|
Lee DY, Chun JN, So I, Jeon JH. Oncogenic role of FOXM1 in human prostate cancer (Review). Oncol Rep 2024; 51:15. [PMID: 38038123 PMCID: PMC10739992 DOI: 10.3892/or.2023.8674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023] Open
Abstract
Prostate cancer is the leading cause of cancer‑related mortality among men worldwide. In particular, castration‑resistant prostate cancer presents a formidable clinical challenge and emphasizes the need to develop novel therapeutic strategies. Forkhead box M1 (FOXM1) is a multifaceted transcription factor that is implicated in the acquisition of the multiple cancer hallmark capabilities in prostate cancer cells, including sustaining proliferative signaling, resisting cell death and the activation of invasion and metastasis. Elevated FOXM1 expression is frequently observed in prostate cancer, and in particular, FOXM1 overexpression is closely associated with poor clinical outcomes in patients with prostate cancer. In the present review, recent advances in the understanding of the oncogenic role of deregulated FOXM1 expression in prostate cancer were highlighted. In addition, the molecular mechanisms by which FOXM1 regulates prostate cancer development and progression were described, thereby providing knowledge and a conceptual framework for FOXM1. The present review also provided valuable insight into the inherent challenges associated with translating biomedical knowledge into effective therapeutic strategies for prostate cancer.
Collapse
Affiliation(s)
- Da Young Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
7
|
Li P, Ma G, Cui Z, Zhang S, Su Q, Cai Z. FOXM1 and CENPF are associated with a poor prognosis through promoting proliferation and migration in lung adenocarcinoma. Oncol Lett 2023; 26:518. [PMID: 37920441 PMCID: PMC10618931 DOI: 10.3892/ol.2023.14105] [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] [Received: 06/20/2023] [Accepted: 09/12/2023] [Indexed: 11/04/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is a clinically challenging disease due to its poor prognosis and limited therapeutic methods. The aim of the present study was to identify prognosis-related genes and therapeutic targets for LUAD. Raw data from the GSE32863, GSE41271 and GSE42127 datasets were downloaded from the Gene Expression Omnibus database. Following normalization, the data were merged into a matrix, which was first used to identify differentially expressed genes (DEGs). Weighted gene co-expression network analysis (WGCNA) and survival analysis were performed to screen potential prognosis-related genes. Gene overlaps among DEGs, survival-related genes and WGCNA genes were finally constructed to obtain candidate genes. An analysis with the STRING database was performed to construct a protein-protein interaction network and hub genes were selected using Cytoscape. The candidate genes were finally identified by univariate and multivariate Cox regression analysis. Furthermore, in vivo and in vitro experiments, including immunohistochemistry, immunofluorescence, Cell Counting Kit-8, colony-formation and migration assays, were performed to validate the potential mechanism of these genes in LUAD. Two genes, namely forkhead box M1 (FOXM1) and centromere protein F (CENPF), were identified as unfavorable indicators of prognosis in patients with LUAD. High expression of FOXM1 and CENPF were associated with poor survival. Furthermore, LUAD cells with FOXM1 and CENPF knockdown showed a significant reduction in proliferation and migration (P<0.05). FOXM1 and CENPF may have an essential role in the prognosis of patients with LUAD by influencing cell proliferation and migration, and they provide potential molecular targets for LUAD therapy.
Collapse
Affiliation(s)
- Peipei Li
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
- Hebei Key Laboratory of Respiratory Critical Care Medicine, The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
- Department of Pulmonary and Critical Care Medicine, Hengshui People's Hospital, Hengshui, Hebei 053000, P.R. China
| | - Geng Ma
- Department of Gastroenterology, Hengshui People's Hospital, Hengshui, Hebei 053000, P.R. China
| | - Zhaobo Cui
- Department of Pulmonary and Critical Care Medicine, Hengshui People's Hospital, Hengshui, Hebei 053000, P.R. China
| | - Shusen Zhang
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
- Hebei Key Laboratory of Respiratory Critical Care Medicine, The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
- Department of Pulmonary and Critical Care Medicine, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, Hebei 054001, P.R. China
| | - Qiao Su
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
- Hebei Key Laboratory of Respiratory Critical Care Medicine, The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zhigang Cai
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
- Hebei Key Laboratory of Respiratory Critical Care Medicine, The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
8
|
Rather TB, Parveiz I, Bhat GA, Rashid G, Wani RA, Khan IY, Mudassar S. Evaluation of Forkhead BOX M1 (FOXM1) gene expression in colorectal cancer. Clin Exp Med 2023; 23:2385-2405. [PMID: 36318377 DOI: 10.1007/s10238-022-00929-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
Abstract
Forkhead Box M1 (FOXM1)-a key cell cycle regulator is a member of the Forkhead transcription factor family. It plays a key role in embryogenesis and cell proliferation and has been strongly linked to various solid tumors. We sought to understand the regulation of FOXM1 in colorectal cancer (CRC), as well as if and to what extent other clinicopathological characteristics are associated with FOXM1. The investigation comprised 98 CRC samples and normal tissues (controls). All colon cancer patients had a colonoscopy and targeted biopsy. All rectal cancer patients had a CT and MRI. Real-time PCR, Immunohistochemistry, and Western blotting were used to evaluate FOXM1 expression, and the findings were analyzed using SPSS (v.26). FOXM1 mRNA and protein expression were substantially upregulated in tumor tissues, with the majority of these proteins localized in nucleo-cytoplasm. Elevated protein levels of FOXM1 were strongly correlated with lower education level, larger tumor size, lymph node status, lymphovascular invasion (LVI), perineural invasion (PNI), lymph node metastasis (LNM), tumor invasion depth (subserosal and serosal invasion), late stage (III and IV), localization (nucleo-cytoplasmic), intensity (strong) and recurrence. Based on survival analysis, FOXM1 overexpression and nucleo-cytoplasmic localization were associated with shorter disease-free survival while stage and PNI were linked to poorer overall and disease-free survival. According to the results of the Cox regression analysis, stage and PNI were significant predictors of prognosis in CRC patients. FOXM1 expression was elevated in CRC and was linked to reduced disease-free survival. These findings support prior reports and hence FOXM1 can be an important prognostic marker for CRC and a promising therapeutic target. Additionally, we found a link between poor disease-free survival and FOXM1's nucleo-cytoplasmic localization. However, since the sample size of this study was small, further research is needed to validate our findings.
Collapse
Affiliation(s)
- Tahseen Bilal Rather
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Kashmir, 190011, India
| | - Ishrat Parveiz
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Kashmir, 190011, India
| | - Gulzar A Bhat
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Kashmir, 190011, India
| | - Gowhar Rashid
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Kashmir, 190011, India
- Department of Medical Lab Technology, Amity Medical School, Amity university, Haryana, India
| | - Rauf A Wani
- Department of General Surgery, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, 190011, India
| | - Ishrat Younas Khan
- Department of Pathology, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, 190011, India
| | - Syed Mudassar
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Kashmir, 190011, India.
| |
Collapse
|
9
|
Castillo-Casas JM, Caño-Carrillo S, Sánchez-Fernández C, Franco D, Lozano-Velasco E. Comparative Analysis of Heart Regeneration: Searching for the Key to Heal the Heart-Part II: Molecular Mechanisms of Cardiac Regeneration. J Cardiovasc Dev Dis 2023; 10:357. [PMID: 37754786 PMCID: PMC10531542 DOI: 10.3390/jcdd10090357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, among which ischemic heart disease is the most representative. Myocardial infarction results from occlusion of a coronary artery, which leads to an insufficient blood supply to the myocardium. As it is well known, the massive loss of cardiomyocytes cannot be solved due the limited regenerative ability of the adult mammalian hearts. In contrast, some lower vertebrate species can regenerate the heart after an injury; their study has disclosed some of the involved cell types, molecular mechanisms and signaling pathways during the regenerative process. In this 'two parts' review, we discuss the current state-of-the-art of the main response to achieve heart regeneration, where several processes are involved and essential for cardiac regeneration.
Collapse
Affiliation(s)
- Juan Manuel Castillo-Casas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Cristina Sánchez-Fernández
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Estefanía Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| |
Collapse
|
10
|
Bu H, Lan X, Cheng H, Pei C, Ouyang M, Chen Y, Huang X, Yu L, Tan Y. Development of an interfering peptide M1-20 with potent anti-cancer effects by targeting FOXM1. Cell Death Dis 2023; 14:533. [PMID: 37598210 PMCID: PMC10439915 DOI: 10.1038/s41419-023-06056-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/21/2023]
Abstract
Disrupting protein-protein interactions (PPIs) has emerged as a promising strategy for cancer drug development. Interfering peptides disrupting PPIs can be rationally designed based on the structures of natural sequences mediating these interactions. Transcription factor FOXM1 overexpresses in multiple cancers and is considered an effective target for cancer therapeutic drug development. Using a rational design approach, we have generated a peptide library from the FOXM1 C-terminal sequence and screened FOXM1-binding peptides. Combining FOXM1 binding and cell inhibitory results, we have obtained a FOXM1-targeting interfering peptide M1-20 that is optimized from the natural parent peptide to the D-retro-inverso peptide. With improved stability characteristics, M1-20 inhibits proliferation and migration, and induces apoptosis of cancer cells. Mechanistically, M1-20 inhibits FOXM1 transcriptional activities by disrupting its interaction between the MuvB complex and the transcriptional co-activator CBP. These are consistent with the results that M1-20 suppresses cancer progression and metastasis without noticeable toxic and side effects in wild-type mice. These findings reveal that M1-20 has the potential to be developed as an anti-cancer drug candidate targeting FOXM1.
Collapse
Affiliation(s)
- Huitong Bu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China
| | - Xianling Lan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China
| | - Haojie Cheng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China
| | - Chaozhu Pei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China
| | - Min Ouyang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China
| | - Yan Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China
| | - Xiaoqin Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China
| | - Li Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China
| | - Yongjun Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, 410082, China.
| |
Collapse
|
11
|
Singh N, Siebzehnrubl FA, Martinez-Garay I. Transcriptional control of embryonic and adult neural progenitor activity. Front Neurosci 2023; 17:1217596. [PMID: 37588515 PMCID: PMC10426504 DOI: 10.3389/fnins.2023.1217596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/10/2023] [Indexed: 08/18/2023] Open
Abstract
Neural precursors generate neurons in the embryonic brain and in restricted niches of the adult brain in a process called neurogenesis. The precise control of cell proliferation and differentiation in time and space required for neurogenesis depends on sophisticated orchestration of gene transcription in neural precursor cells. Much progress has been made in understanding the transcriptional regulation of neurogenesis, which relies on dose- and context-dependent expression of specific transcription factors that regulate the maintenance and proliferation of neural progenitors, followed by their differentiation into lineage-specified cells. Here, we review some of the most widely studied neurogenic transcription factors in the embryonic cortex and neurogenic niches in the adult brain. We compare functions of these transcription factors in embryonic and adult neurogenesis, highlighting biochemical, developmental, and cell biological properties. Our goal is to present an overview of transcriptional regulation underlying neurogenesis in the developing cerebral cortex and in the adult brain.
Collapse
Affiliation(s)
- Niharika Singh
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, United Kingdom
| | - Florian A. Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff, United Kingdom
| | - Isabel Martinez-Garay
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
12
|
Yi J, Li H, Chu B, Kon N, Hu X, Hu J, Xiong Y, Kaniskan HU, Jin J, Gu W. Inhibition of USP7 induces p53-independent tumor growth suppression in triple-negative breast cancers by destabilizing FOXM1. Cell Death Differ 2023; 30:1799-1810. [PMID: 37291217 PMCID: PMC10307817 DOI: 10.1038/s41418-023-01180-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/28/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023] Open
Abstract
Although numerous studies indicate that inhibition of USP7 suppresses tumor growth by activating p53, the precise mechanism by which USP7 contributes to tumor growth through the p53-independent manner is not well understood. p53 is frequently mutated in most triple-negative breast cancers (TNBC), characterized as the very aggressive form of breast cancers with limited treatment options and poor patient outcomes. Here, we found that the oncoprotein Forkhead Box M1 (FOXM1) acts as a potential driver for tumor growth in TNBC and, surprisingly, through a proteomic screen, we identified USP7 as a major regulator of FOXM1 in TNBC cells. USP7 interacts with FOXM1 both in vitro and in vivo. USP7 stabilizes FOXM1 through deubiquitination. Conversely, RNAi-mediated USP7 knockdown in TNBC cells, dramatically reduced the levels of FOXM1. Moreover, based upon the proteolysis targeting chimera (PROTAC) technology, we generated PU7-1 (protein degrader for USP7-1), as a USP7 specific degrader. PU7-1 induces rapid USP7 degradation at low nanomolar concentrations in cells but shows no obvious effect on other USP family proteins. Strikingly, the treatment of TNBC cells with PU7-1 significantly abrogates FOXM1 functions and effectively suppresses cell growth in vitro. By using xenograft mouse models, we found that PU7-1 markedly represses tumor growth in vivo. Notably, ectopic overexpression of FOXM1 can reverse the tumor growth suppressive effects induced by PU7-1, underscored the specific effect on FOXM1 induced by USP7 inactivation. Together, our findings indicate that FOXM1 is a major target of USP7 in modulating tumor growth in a p53-independent manner and reveals the USP7 degrader as a potential therapeutic tool for the treatment of triple-negative breast cancers.
Collapse
Affiliation(s)
- Jingjie Yi
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Huan Li
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Bo Chu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ning Kon
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Xiaoping Hu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jianping Hu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - H Umit Kaniskan
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
| |
Collapse
|
13
|
Cheng H, Yuan J, Pei C, Ouyang M, Bu H, Chen Y, Huang X, Zhang Z, Yu L, Tan Y. The development of an anti-cancer peptide M1-21 targeting transcription factor FOXM1. Cell Biosci 2023; 13:114. [PMID: 37344857 DOI: 10.1186/s13578-023-01059-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Transcription factor FOXM1 is a potential target for anti-cancer drug development. An interfering peptide M1-21, targeting FOXM1 and FOXM1-interacting proteins, is developed and its anti-cancer efficacy is evaluated. METHODS FOXM1 C-terminus-binding peptides are screened by in silico protocols from the peptide library of FOXM1 (1-138aa) and confirmed by cellular experiments. The selected peptide is synthesized into its D-retro-inverso (DRI) form by fusing a TAT cell-penetrating sequence. Anti-cancer activities are evaluated in vitro and in vivo with tumor-grafted nude mice, spontaneous breast cancer mice, and wild-type metastasis-tracing mice. Anti-cancer mechanisms are analyzed. Distribution and safety profiles in mice are evaluated. RESULTS With improved stability and cell inhibitory activity compared to the parent peptide, M1-21 binds to multiple regions of FOXM1 and interferes with protein-protein interactions between FOXM1 and its various known partner proteins, including PLK1, LIN9 and B-MYB of the MuvB complex, and β-catenin. Consequently, M1-21 inhibits FOXM1-related transcriptional activities and FOXM1-mediated nuclear importation of β-catenin and β-catenin transcriptional activities. M1-21 inhibits multiple types of cancer (20 µM in vitro or 30 mg/kg in vivo) by preventing proliferation, migration, and WNT signaling. Distribution and safety profiles of M1-21 are favorable (broad distribution and > 15 h stability in mice) and the tested non-severely toxic dose reaches 200 mg/kg in mice. M1-21 also has low hemolytic toxicity and immunogenicity in mice. CONCLUSIONS M1-21 is a promising interfering peptide targeting FOXM1 for the development of anti-cancer drugs.
Collapse
Affiliation(s)
- Haojie Cheng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China
| | - Jie Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China
| | - Chaozhu Pei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China
| | - Min Ouyang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China
| | - Huitong Bu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China
| | - Yan Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China
| | - Xiaoqin Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China
| | - Zhenwang Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China.
- Medicine Research Institute, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, 437000, Xianning, Hubei, China.
| | - Li Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China.
| | - Yongjun Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, 410082, Changsha, Hunan, China.
| |
Collapse
|
14
|
Chen Y, Wang X, Ye D, Yang Z, Shen Q, Liu X, Chen C, Chen X. Research progress of sophoridine's pharmacological activities and its molecular mechanism: an updated review. Front Pharmacol 2023; 14:1126636. [PMID: 37397472 PMCID: PMC10311568 DOI: 10.3389/fphar.2023.1126636] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 06/07/2023] [Indexed: 07/04/2023] Open
Abstract
Background: Sophoridine, the major active constituent of Sophora alopecuroides and its roots, is a bioactive alkaloid with a wide range of pharmacological effects, including antitumor, anti-inflammatory, antiviral, antibacterial, analgesic, cardioprotective, and immunoprotective activities. Sophora flavescens Aiton is a traditional Chinese medicine that is bitter and cold. Additionally, it also exhibits the effects of clearing heat, eliminating dampness, and expelling insects. Aims of the study: To summarize the pharmacological research and associated mechanisms of sophoridine, we compiled this review by combining a huge body of relevant literature. Materials and methods: The information related to this article was systematically collected from the scientific literature databases including PubMed, Google Scholar, Web of Science, Science Direct, Springer, China National Knowledge Infrastructure, published books, PhD and MS dissertations. Results: Its antitumor activity is particularly remarkable, as it can inhibit cancer cell proliferation, invasion, and metastasis while inducing cell cycle arrest and apoptosis. Additionally, sophoridine also holds therapeutic potential for myocardial ischemia, osteoporosis, arrhythmias, and neurological disorders, primarily through the suppression of related inflammatory factors and cell apoptosis. However, sophoridine has also exhibited adverse effects such as hepatotoxicity and neurotoxicity. The antidisease effect and mechanism of sophoridine are diverse, so it has high research value. Conclusion: As an important traditional Chinese medicine alkaloid, modern pharmacological studies have demonstrated that sophoridine has prominent bioactivities, especially on anti-tumor anti-inflammation activities, and cardiovascular system protection. These activities provide prospects for novel drug development for cancer and some chronic diseases. Nevertheless, the understanding of the multitarget network pharmacology, long-term in vivo toxicity, and clinical efficacy of sophoridine require further detailed research.
Collapse
Affiliation(s)
- Yiwei Chen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Xiang Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dongmei Ye
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Zhousheng Yang
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Qingrong Shen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Xiaoxia Liu
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Chunxia Chen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| | - Xiaoyu Chen
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous, Nanning, China
| |
Collapse
|
15
|
Zhao R, Wang X, Zhou X, Jiang S, Zhang L, Yu Z. Metabolites and gene expression in the myocardium of fasting rats in an acute hypoxic environment. BMC Genomics 2023; 24:251. [PMID: 37165337 PMCID: PMC10173486 DOI: 10.1186/s12864-023-09309-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/12/2023] [Indexed: 05/12/2023] Open
Abstract
With the rising demand for entry to extremely high altitudes (HAs), rapid adaptability to extremely hypoxic environments is a challenge that we need to explore. Fasting was used to evaluate acute hypoxia tolerance at HA and was proven to be an effective method for improving the survival rate at extreme HA. Our experiments also showed that fasting pretreatment for 72 h significantly increased the 24 h survival rate of rats at 7620 m from 10 to 85% and protected the myocardium cells of rats. Here, we compared the metabolites and gene expression in the myocardium of SD rats pretreated with fasting and nonfasting at normal altitude and extreme HA. Our findings demonstrated that the dynamic contents of detected differential metabolites (DMs) between different rat groups were consistent with the expression of differentially expressed genes (DEGs), and DM clusters also showed strong correlations with DEG clusters. DM clusters related to amino acids and lipids were significantly lower in the fasting groups, and the correlated DEG clusters were enriched in mitotic pathways, including CDK1, CDC7, NUF2, and MCM6, suggesting that fasting can attenuate mitotic processes in cardiac tissues and reduce the synthesis of amino acids and lipids. L-Glutamine-related metabolites were particularly low at extreme HA without pretreatment but were normal in the fasting groups. The DEGs in the cluster related to L-glutamine-related metabolites were enriched for T-cell receptor V(D)J recombination, the Hippo signaling pathway, the Wnt signaling pathway, the cGMP-PKG signaling pathway, and the mTOR signaling pathway and were significantly downregulated, indicating that the content of L-glutamine decreased at extreme HA, while fasting increased it to adapt to the environment. Moreover, abundant fatty acids were detected when rats were exposed to extreme HA without pretreatment. Our study revealed the fasting and hypoxic environment-related factors in SD rats and provided new insights into the genetic and molecular characteristics in the myocardium, which is critical to developing more potential rapid adaptation methods to extreme HA.
Collapse
Affiliation(s)
- Ruzhou Zhao
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xiaobo Wang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xiang Zhou
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
- Department of Nuclear Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Shuai Jiang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Lin Zhang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Zhibin Yu
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China.
| |
Collapse
|
16
|
Moore XTR, Gheghiani L, Fu Z. The Role of Polo-Like Kinase 1 in Regulating the Forkhead Box Family Transcription Factors. Cells 2023; 12:cells12091344. [PMID: 37174744 PMCID: PMC10177174 DOI: 10.3390/cells12091344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase with more than 600 phosphorylation substrates through which it regulates many biological processes, including mitosis, apoptosis, metabolism, RNA processing, vesicle transport, and G2 DNA-damage checkpoint recovery, among others. Among the many PLK1 targets are members of the FOX family of transcription factors (FOX TFs), including FOXM1, FOXO1, FOXO3, and FOXK1. FOXM1 and FOXK1 have critical oncogenic roles in cancer through their antagonism of apoptotic signals and their promotion of cell proliferation, metastasis, angiogenesis, and therapeutic resistance. In contrast, FOXO1 and FOXO3 have been identified to have broad functions in maintaining cellular homeostasis. In this review, we discuss PLK1-mediated regulation of FOX TFs, highlighting the effects of PLK1 on the activity and stability of these proteins. In addition, we review the prognostic and clinical significance of these proteins in human cancers and, more importantly, the different approaches that have been used to disrupt PLK1 and FOX TF-mediated signaling networks. Furthermore, we discuss the therapeutic potential of targeting PLK1-regulated FOX TFs in human cancers.
Collapse
Affiliation(s)
- Xavier T R Moore
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Lilia Gheghiani
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Zheng Fu
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| |
Collapse
|
17
|
Alimardan Z, Abbasi M, Hasanzadeh F, Aghaei M, Khodarahmi G, Kashfi K. Heat shock proteins and cancer: The FoxM1 connection. Biochem Pharmacol 2023; 211:115505. [PMID: 36931349 PMCID: PMC10134075 DOI: 10.1016/j.bcp.2023.115505] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Heat shock proteins (Hsp) and FoxM1 have significant roles in carcinogenesis. According to their relative molecular weight, Hsps are divided into Hsp110, Hsp90, Hsp70, Hsp60, Hsp40, and small Hsps. Hsp70 can play essential functions in cancer initiation and is overexpressed in several human cancers. Hsp70, in combination with cochaperones HIP and HOP, refolds partially denatured proteins and acts as a cochaperone for Hsp90. Also, Hsp70, in combination with BAG3, regulates the FoxM1 signaling pathway. FoxM1 protein is a transcription factor of the Forkhead family that is overexpressed in most human cancers and is involved in many cancers' development features, including proliferation, migration, invasion, angiogenesis, metastasis, and resistance to apoptosis. This review discusses the Hsp70, Hsp90, and FoxM1 structure and function, the known Hsp70 cochaperones, and Hsp70, Hsp90, and FoxM1 inhibitors.
Collapse
Affiliation(s)
- Zahra Alimardan
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Pharmacology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Maryam Abbasi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Farshid Hasanzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmud Aghaei
- Department of Biochemistry, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghadamali Khodarahmi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, NY, USA.
| |
Collapse
|
18
|
Donovan J, Deng Z, Bian F, Shukla S, Gomez-Arroyo J, Shi D, Kalinichenko VV, Kalin TV. Improving anti-tumor efficacy of low-dose Vincristine in rhabdomyosarcoma via the combination therapy with FOXM1 inhibitor RCM1. Front Oncol 2023; 13:1112859. [PMID: 36816948 PMCID: PMC9933126 DOI: 10.3389/fonc.2023.1112859] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a highly metastatic soft-tissue sarcoma that often develops resistance to current therapies, including vincristine. Since the existing treatments have not significantly improved survival, there is a critical need for new therapeutic approaches for RMS patients. FOXM1, a known oncogene, is highly expressed in RMS, and is associated with the worst prognosis in RMS patients. In the present study, we found that the combination treatment with specific FOXM1 inhibitor RCM1 and low doses of vincristine is more effective in increasing apoptosis and decreasing RMS cell proliferation in vitro compared to single drugs alone. Since RCM1 is highly hydrophobic, we developed innovative nanoparticle delivery system containing poly-beta-amino-esters and folic acid (NPFA), which efficiently delivers RCM1 to mouse RMS tumors in vivo. The combination of low doses of vincristine together with intravenous administration of NPFA nanoparticles containing RCM1 effectively reduced RMS tumor volumes, increased tumor cell death and decreased tumor cell proliferation in RMS tumors compared to RCM1 or vincristine alone. The combination therapy was non-toxic as demonstrated by liver metabolic panels using peripheral blood serum. Using RNA-seq of dissected RMS tumors, we identified Chac1 as a uniquely downregulated gene after the combination treatment. Knockdown of Chac1 in RMS cells in vitro recapitulated the effects of the combination therapy. Altogether, combination treatment with low doses of vincristine and nanoparticle delivery of FOXM1 inhibitor RCM1 in a pre-clinical model of RMS has superior anti-tumor effects and decreases CHAC1 while reducing vincristine toxicity.
Collapse
Affiliation(s)
- Johnny Donovan
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Zicheng Deng
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States,Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Fenghua Bian
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Samriddhi Shukla
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jose Gomez-Arroyo
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Division of Pulmonary and Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Donglu Shi
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States
| | - Vladimir V. Kalinichenko
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Tanya V. Kalin
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,*Correspondence: Tanya V. Kalin,
| |
Collapse
|
19
|
Conte A, Valente V, Paladino S, Pierantoni GM. HIPK2 in cancer biology and therapy: Recent findings and future perspectives. Cell Signal 2023; 101:110491. [PMID: 36241057 DOI: 10.1016/j.cellsig.2022.110491] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
Homeodomain-interacting protein kinase 2 (HIPK2) is a serine-threonine kinase that phosphorylates and regulates a plethora of transcriptional regulators and chromatin modifiers. The heterogeneity of its interactome allows HIPK2 to modulate several cellular processes and signaling pathways, ultimately regulating cell fate and proliferation. Because of its p53-dependent pro-apoptotic activity and its downregulation in many tumor types, HIPK2 is traditionally considered a bone fide tumor suppressor gene. However, recent findings revealed that the role of HIPK2 in the pathogenesis of cancer is much more complex, ranging from tumor suppressive to oncogenic, strongly depending on the cellular context. Here, we review the very recent data emerged in the last years about the involvement of HIPK2 in cancer biology and therapy, highlighting the various alterations of this kinase (downregulation, upregulation, mutations and/or delocalization) in dependence on the cancer types. In addition, we discuss the recent advancement in the understanding the tumor suppressive and oncogenic functions of HIPK2, its role in establishing the response to cancer therapies, and its regulation by cancer-associated microRNAs. All these data strengthen the idea that HIPK2 is a key player in many types of cancer; therefore, it could represent an important prognostic marker, a factor to predict therapy response, and even a therapeutic target itself.
Collapse
Affiliation(s)
- Andrea Conte
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.
| | - Valeria Valente
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Giovanna Maria Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
20
|
Magee N, Ahamed F, Eppler N, Jones E, Ghosh P, He L, Zhang Y. Hepatic transcriptome profiling reveals early signatures associated with disease transition from non-alcoholic steatosis to steatohepatitis. LIVER RESEARCH 2022; 6:238-250. [PMID: 36864891 PMCID: PMC9977163 DOI: 10.1016/j.livres.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background and aim Non-alcoholic fatty liver disease (NAFLD) is becoming a leading cause of chronic liver disease worldwide. The molecular events that influence disease progression from non-alcoholic fatty liver (NAFL) to aggressive non-alcoholic steatohepatitis (NASH) remain incompletely understood, leading to lack of mechanism-based targeted treatment options for NASH. This study aims to identify early signatures associated with disease progression from NAFL to NASH in mice and humans. Materials and methods Male C57BL/6J mice were fed a high-fat, -cholesterol, and - fructose (HFCF) diet for up to 9 months. The extent of steatosis, inflammation, and fibrosis was evaluated in liver tissues. Total RNA sequencing (RNA-seq) was conducted to determine liver transcriptomic changes. Results After being fed the HFCF diet, mice sequentially developed steatosis, early steatohepatitis, steatohepatitis with fibrosis, and eventually spontaneous liver tumor. Hepatic RNA-seq revealed that the key signatures during steatosis progression to early steatohepatitis were pathways related to extracellular matrix organization and immune responses such as T cell migration, arginine biosynthesis, C-type lectin receptor signaling, and cytokine-cytokine receptor interaction. Genes regulated by transcription factors forkhead box M1 (FOXM1) and negative elongation factor complex member E (NELFE) were significantly altered during disease progression. This phenomenon was also observed in patients with NASH. Conclusions In summary, we identified early signatures associated with disease progression from NAFL to early NASH in a mouse model that recapitulated key metabolic, histologic, and transcriptomic changes seen in humans. The findings from our study may shed light on the development of novel preventative, diagnostic, and therapeutic strategies for NASH.
Collapse
Affiliation(s)
- Nancy Magee
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Forkan Ahamed
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Natalie Eppler
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Elizabeth Jones
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Priyanka Ghosh
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lily He
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
- Liver Center, University of Kansas, Kansas City, KS, USA
| |
Collapse
|
21
|
A lignan from Alnus japonica inhibits glioblastoma tumorspheres by suppression of FOXM1. Sci Rep 2022; 12:13990. [PMID: 35978012 PMCID: PMC9385634 DOI: 10.1038/s41598-022-18185-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 08/08/2022] [Indexed: 11/08/2022] Open
Abstract
Forkhead Box M1 (FOXM1) is known to regulate cell proliferation, apoptosis and tumorigenesis. The lignan, (-)-(2R,3R)-1,4-O-diferuloylsecoisolariciresinol (DFS), from Alnus japonica has shown anti-cancer effects against colon cancer cells by suppressing FOXM1. The present study hypothesized that DFS can have anti-cancer effects against glioblastoma (GBM) tumorspheres (TSs). Immunoprecipitation and luciferase reporter assays were performed to evaluate the ability of DFS to suppress nuclear translocation of β-catenin through β-catenin/FOXM1 binding. DFS-pretreated GBM TSs were evaluated to assess the ability of DFS to inhibit GBM TSs and their transcriptional profiles. The in vivo efficacy was examined in orthotopic xenograft models of GBM. Expression of FOXM1 was higher in GBM than in normal tissues. DFS-induced FOXM1 protein degradation blocked β-catenin translocation into the nucleus and consequently suppressed downstream target genes of FOXM1 pathways. DFS inhibited cell viability and ATP levels, while increasing apoptosis, and it reduced tumorsphere formation and the invasiveness of GBM TSs. And DFS reduced the activities of transcription factors related to tumorigenesis, stemness, and invasiveness. DFS significantly inhibited tumor growth and prolonged the survival rate of mice in orthotopic xenograft models of GBM. It suggests that DFS inhibits the proliferation of GBM TSs by suppressing FOXM1. DFS may be a potential therapeutic agent to treat GBM.
Collapse
|
22
|
Zhang Z, Liu W, Bao X, Sun T, Wang J, Li M, Liu C. USP39 facilitates breast cancer cell proliferation through stabilization of FOXM1. Am J Cancer Res 2022; 12:3644-3661. [PMID: 36119839 PMCID: PMC9442023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/09/2022] [Indexed: 06/15/2023] Open
Abstract
Deubiquitinating enzyme dysregulation has been linked to the development of a variety of human malignancies, including breast cancer. However, the exact involvement of the deubiquitinating enzyme USP39 in the progression of breast cancer is yet unknown. Cell viability and colony formation analysis was used to assess the effects of USP39 knockdown on breast cancer cells in this study. The interaction between USP39 and FOXM1 was investigated using co-immunoprecipitation (co-IP) and in vitro deubiquitination analysis. The expression of USP39 and FOXM1 in breast cancer tissues was studied using the TCGA database. According to our findings, USP39 deubiquitinates and stabilizes FOXM1, promoting breast cancer cell proliferation, colony formation, and tumor growth in vivo. Furthermore, elevated USP39 expression lowers FOXM1 ubiquitination, resulting in increased transcriptional activity. In addition, the high expression of USP39 reduces the ubiquitination of FOXM1, thereby enhancing the transcriptional activity of FOXM1 and regulating the expression of downstream genes Cdc25b and Plk1. USP39 is positively correlated with the expression level of FOXM1 in breast cancer cells. In general, our research revealed the USP39-FOXM1 axis as a critical driver of breast cancer cell proliferation and provided a theoretical foundation for targeting the USP39-FOXM1 axis for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Zhenwang Zhang
- Medicine Research Institute/Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and TechnologyXianning 437000, Hubei, China
- Hubei University of Science and Technology of Medicine, Xianning Medical CollegeXianning 437000, Hubei, China
| | - Wu Liu
- Medicine Research Institute/Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and TechnologyXianning 437000, Hubei, China
- Hubei University of Science and Technology of Medicine, Xianning Medical CollegeXianning 437000, Hubei, China
| | - Xiajun Bao
- Medicine Research Institute/Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and TechnologyXianning 437000, Hubei, China
- Hubei University of Science and Technology of Medicine, Xianning Medical CollegeXianning 437000, Hubei, China
| | - Tian Sun
- Medicine Research Institute/Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and TechnologyXianning 437000, Hubei, China
- Hubei University of Science and Technology of Medicine, Xianning Medical CollegeXianning 437000, Hubei, China
| | - Jiawei Wang
- Medicine Research Institute/Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and TechnologyXianning 437000, Hubei, China
- Hubei University of Science and Technology of Medicine, Xianning Medical CollegeXianning 437000, Hubei, China
| | - Mengxi Li
- Science and Technology Industry Management Office, Hubei University of Science and TechnologyXianning 437000, Hubei, China
| | - Chao Liu
- Medicine Research Institute/Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and TechnologyXianning 437000, Hubei, China
- Hubei University of Science and Technology of Medicine, Xianning Medical CollegeXianning 437000, Hubei, China
| |
Collapse
|
23
|
Sawaya AP, Stone RC, Mehdizadeh S, Pastar I, Worrell S, Balukoff NC, Kaplan MJ, Tomic‐Canic M, Morasso MI. FOXM1
network in association with
TREM1
suppression regulates
NET
formation in diabetic foot ulcers. EMBO Rep 2022; 23:e54558. [PMID: 35856334 PMCID: PMC9346470 DOI: 10.15252/embr.202154558] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/26/2022] [Accepted: 06/08/2022] [Indexed: 11/09/2022] Open
Abstract
Diabetic foot ulcers (DFU) are a serious complication of diabetes mellitus and associated with reduced quality of life and high mortality rate. DFUs are characterized by a deregulated immune response with decreased neutrophils due to loss of the transcription factor, FOXM1. Diabetes primes neutrophils to form neutrophil extracellular traps (NETs), contributing to tissue damage and impaired healing. However, the role of FOXM1 in priming diabetic neutrophils to undergo NET formation remains unknown. Here, we found that FOXM1 regulates reactive oxygen species (ROS) levels in neutrophils and inhibition of FOXM1 results in increased ROS leading to NET formation. Next generation sequencing revealed that TREM1 promoted the recruitment of FOXM1+ neutrophils and reversed effects of diabetes and promoted wound healing in vivo. Moreover, we found that TREM1 expression correlated with clinical healing outcomes of DFUs, indicating TREM1 may serve as a useful biomarker or a potential therapeutic target. Our findings highlight the clinical relevance of TREM1, and indicates FOXM1 pathway as a novel regulator of NET formation during diabetic wound healing, revealing new therapeutic strategies to promote healing in DFUs.
Collapse
Affiliation(s)
- Andrew P Sawaya
- Laboratory of Skin Biology National Institute of Arthritis and Musculoskeletal and Skin Diseases National Institutes of Health Bethesda MD USA
| | - Rivka C Stone
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery University of Miami Miller School of Medicine Miami FL USA
| | - Spencer Mehdizadeh
- Laboratory of Skin Biology National Institute of Arthritis and Musculoskeletal and Skin Diseases National Institutes of Health Bethesda MD USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery University of Miami Miller School of Medicine Miami FL USA
| | - Stephen Worrell
- Laboratory of Skin Biology National Institute of Arthritis and Musculoskeletal and Skin Diseases National Institutes of Health Bethesda MD USA
| | - Nathan C Balukoff
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery University of Miami Miller School of Medicine Miami FL USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases National Institutes of Health Bethesda MD USA
| | - Marjana Tomic‐Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery University of Miami Miller School of Medicine Miami FL USA
- John P. Hussman Institute for Human Genomics University of Miami Miller School of Medicine Miami FL USA
| | - Maria I Morasso
- Laboratory of Skin Biology National Institute of Arthritis and Musculoskeletal and Skin Diseases National Institutes of Health Bethesda MD USA
| |
Collapse
|
24
|
Yang EJ, Park JH, Cho HJ, Hwang JA, Woo SH, Park CH, Kim SY, Park JT, Park SC, Hwang D, Lee YS. Co-inhibition of ATM and ROCK synergistically improves cell proliferation in replicative senescence by activating FOXM1 and E2F1. Commun Biol 2022; 5:702. [PMID: 35835838 PMCID: PMC9283421 DOI: 10.1038/s42003-022-03658-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 06/29/2022] [Indexed: 12/30/2022] Open
Abstract
The multifaceted nature of senescent cell cycle arrest necessitates the targeting of multiple factors arresting or promoting the cell cycle. We report that co-inhibition of ATM and ROCK by KU-60019 and Y-27632, respectively, synergistically increases the proliferation of human diploid fibroblasts undergoing replicative senescence through activation of the transcription factors E2F1 and FOXM1. Time-course transcriptome analysis identified FOXM1 and E2F1 as crucial factors promoting proliferation. Co-inhibition of the kinases ATM and ROCK first promotes the G2/M transition via FOXM1 activation, leading to accumulation of cells undergoing the G1/S transition via E2F1 activation. The combination of both inhibitors increased this effect more significantly than either inhibitor alone, suggesting synergism. Our results demonstrate a FOXM1- and E2F1-mediated molecular pathway enhancing cell cycle progression in cells with proliferative potential under replicative senescence conditions, and treatment with the inhibitors can be tested for senomorphic effect in vivo.
Collapse
Affiliation(s)
- Eun Jae Yang
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Ji Hwan Park
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Hyun-Ji Cho
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Jeong-A Hwang
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Seung-Hwa Woo
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Chi Hyun Park
- Department of Computer Science and Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Seoul, 05029, Korea
| | - Joon Tae Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Sang Chul Park
- Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988, Republic of Korea.
- The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju, 61469, Republic of Korea.
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Young-Sam Lee
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea.
- Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988, Republic of Korea.
- New Biology Research Center, DGIST, Daegu, 42988, Republic of Korea.
| |
Collapse
|
25
|
Distinct Roles of NANOS1 and NANOS3 in the Cell Cycle and NANOS3-PUM1-FOXM1 Axis to Control G2/M Phase in a Human Primordial Germ Cell Model. Int J Mol Sci 2022; 23:ijms23126592. [PMID: 35743036 PMCID: PMC9223905 DOI: 10.3390/ijms23126592] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 12/20/2022] Open
Abstract
Nanos RNA-binding proteins are critical factors of germline development throughout the animal kingdom and their dysfunction causes infertility. During evolution, mammalian Nanos paralogues adopted divergent roles in germ cell biology. However, the molecular basis behind this divergence, such as their target mRNAs, remains poorly understood. Our RNA-sequencing analysis in a human primordial germ cell model-TCam-2 cell line revealed distinct pools of genes involved in the cell cycle process downregulated upon NANOS1 and NANOS3 overexpression. We show that NANOS1 and NANOS3 proteins influence different stages of the cell cycle. Namely, NANOS1 is involved in the G1/S and NANOS3 in the G2/M phase transition. Many of their cell cycle targets are known infertility and cancer-germ cell genes. Moreover, NANOS3 in complex with RNA-binding protein PUM1 causes 3′UTR-mediated repression of FOXM1 mRNA encoding a transcription factor crucial for G2/M phase transition. Interestingly, while NANOS3 and PUM1 act as post-transcriptional repressors of FOXM1, FOXM1 potentially acts as a transcriptional activator of NANOS3, PUM1, and itself. Finally, by utilizing publicly available RNA-sequencing datasets, we show that the balance between FOXM1-NANOS3 and FOXM1-PUM1 expression levels is disrupted in testis cancer, suggesting a potential role in this disease.
Collapse
|
26
|
Zhu J, Zhao J, Luo C, Zhu Z, Peng X, Zhu X, Lin K, Bu F, Zhang W, Li Q, Wang K, Hu Z, Yu X, Chen L, Yuan R. FAT10 promotes chemotherapeutic resistance in pancreatic cancer by inducing epithelial-mesenchymal transition via stabilization of FOXM1 expression. Cell Death Dis 2022; 13:497. [PMID: 35614040 PMCID: PMC9132907 DOI: 10.1038/s41419-022-04960-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer (PC) is one of the deadliest malignant tumors, and its resistance to gemcitabine chemotherapy is the primary reason for poor prognosis in patients. Ubiquitin-like protein FAT10 has recently been reported to promote tumor chemotherapy resistance. In this study, the expression of FAT10 in PC was significantly higher than that in adjacent noncancerous tissues. Increased expression of FAT10 in PC was related to a late TNM stage and decreased overall survival. Functional experiments revealed that downregulating the expression of FAT10 inhibits the proliferation and epithelial-mesenchymal transition (EMT) of PC cells, promotes the apoptosis of PC cells, and enhances sensitivity to gemcitabine chemotherapy. In addition, upregulation of FAT10 increased the expression of FOXM1 protein. The effect of downregulating FAT10 was reversed by FOXM1 overexpression, and FOXM1 knockdown inhibited EMT driven by FAT10 overexpression. Mechanistically, FAT10 stabilized the expression of FOXM1 by competing with ubiquitin to bind FOXM1 and inhibiting the ubiquitination-mediated degradation of FOXM1. In conclusion, the FAT10-FOXM1 axis is a pivotal driver of PC proliferation and gemcitabine resistance, and the results provide novel insights into chemotherapy resistance in PC.
Collapse
Affiliation(s)
- Jinfeng Zhu
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Jiefeng Zhao
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Chen Luo
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Zhengming Zhu
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Xingyu Peng
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Xiaojian Zhu
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Kang Lin
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Fanqin Bu
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Wenjun Zhang
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Qing Li
- Department of Pathology, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Kai Wang
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Jiangxi Provincial Clinical Research Center for General Surgery Disease, Nanchang, 330006, Jiangxi Province, China
| | - Zhigang Hu
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Xin Yu
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Jiangxi Provincial Clinical Research Center for General Surgery Disease, Nanchang, 330006, Jiangxi Province, China
| | - Leifeng Chen
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Rongfa Yuan
- Department of General Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Jiangxi Provincial Clinical Research Center for General Surgery Disease, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
27
|
Mondal A, Das S, Samanta J, Chakraborty S, sengupta A. YAP1 induces hyperglycemic stress-mediated cardiac hypertrophy and fibrosis in an AKT-FOXM1 dependent signaling pathway. Arch Biochem Biophys 2022; 722:109198. [DOI: 10.1016/j.abb.2022.109198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 11/28/2022]
|
28
|
Sun H, Zhang N, Jin Y, Xu H. Cardamonin Promotes the Apoptosis and Chemotherapy Sensitivity to Gemcitabine of Pancreatic Cancer Through Modulating the FOXO3a-FOXM1 Axis. Dose Response 2022; 19:15593258211042163. [PMID: 34987330 PMCID: PMC8725235 DOI: 10.1177/15593258211042163] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022] Open
Abstract
Cardamonin (CAR), a flavone existing in the Alpinia plant, has been found to
modulate multiple biological activities, including antioxidant,
anti-inflammatory, and anti-tumor effects. Nevertheless, the influence of CAR on
pancreatic cancer (PC) is less understood. Here, we conducted in
vitro and in vivo experiments to explore the
functions of CAR on PC cells’ proliferation, apoptosis and chemosensitivity to
gemcitabine (GEM). The growth of PC cells (including PANC-1 and SW1990) was
evaluated by the cell counting kit-8 assay, colony formation assay and xenograft
tumor experiment. Besides, the apoptosis was determined by flow cytometry and
western blot (WB). Moreover, the FOXO3a-FOXM1 pathway expression was tested by
reverse transcription-polymerase chain reaction and WB. Our data suggested that
CAR restrained cell proliferation, growth and expedited apoptosis both
in vitro and in vivo. Moreover, CAR
sensitized PC cells to GEM. Mechanistically, CAR heightened FOXO3a while
repressed FOXM1. Further loss-of-function assays revealed that down-regulating
FOXO3a markedly dampened the anti-tumor effect induced by CAR and accelerated
the FOXM1 expression. Our data confirmed that CAR exerted an anti-tumor function
in PC dependently by modulating the FOXO3a-FOXM1 axis.
Collapse
Affiliation(s)
- Huapeng Sun
- Department of General Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Na Zhang
- Department of Pathology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yiqiang Jin
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Haisheng Xu
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
29
|
Shabani S, Elahi E, Bahraniasl M, Babaheidarian P, Sadeghpour A, Majidzadeh T, Talebi A, Mahjoubi F. Multi-stage analysis of FOXM1, PYROXD1, hTERT, PPARA, PIM3, BMI1 and MCTP1 expression patterns in colorectal cancer. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2022; 15:120-130. [PMID: 35845311 PMCID: PMC9275742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/29/2022] [Indexed: 10/27/2022]
Abstract
Aim To explore biomarkers with a tumor stage-dependent expression pattern in patients with colorectal cancer (CRC). Background The fourth most common cancer in the world is colorectal cancer (CRC). A variation in the gene expression rate is a common change in cancers initiation and the accumulation of these variation changes the behavior of normal cells and turns them into cancer cells. Methods Real-time RT-PCR was used to investigate the expression patterns of the FOXM1, PYROXD1, hTERT, BMI, PPARA, PIM3 and MCTP1 genes in 54 patients with stage I to IV CRC and their relation with clinicopathological features of CRC were analyzed. Results: FOXM1, hTERT and MCTP1 genes are overexpressed in CRC tumor tissues when compared to normal adjacent tissues in all the stages. Results FOXM1, PYROXD1, hTERT, PIM3, BMI1, PPARA and MCTP1 had-stage dependent expression. Investigation of the association between clinicopathological features and expression pattern of the studied genes revealed; a) a significant relationship between FOXM1 gene expression level and tumor stage, tumor size and lymph node involvement, b) a considerable association between alterations in PPARA and PIM3 expression and lymph node involvement, c) a notable correlation between hTERT expression level and the tumor stage and d) a strong correlation between MCTP1 expression and patient's age only. Conclusion Our study indicates that expression profiles of these genes either individually or together can be applied as potential biomarkers for prognosis of CRC.
Collapse
Affiliation(s)
- Samira Shabani
- Department of Clinical Genetic, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Elahe Elahi
- Department of Biotechnology, University College of Science, University of Tehran, Tehran, Iran
| | - Mandana Bahraniasl
- Department of Clinical Genetic, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Pegah Babaheidarian
- Department of Pathology, Hazrate-Rasoule Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Sadeghpour
- Department of Pathology, Hazrate-Rasoule Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Majidzadeh
- Department of Clinical Genetic, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Atefeh Talebi
- Colorectal Research Centre (CRRC), Hazrate-Rasoule Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Frouzandeh Mahjoubi
- Department of Clinical Genetic, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
30
|
Nandi D, Cheema PS, Singal A, Bharti H, Nag A. Artemisinin Mediates Its Tumor-Suppressive Activity in Hepatocellular Carcinoma Through Targeted Inhibition of FoxM1. Front Oncol 2021; 11:751271. [PMID: 34900697 PMCID: PMC8652299 DOI: 10.3389/fonc.2021.751271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/04/2021] [Indexed: 12/28/2022] Open
Abstract
The aberrant up-regulation of the oncogenic transcription factor Forkhead box M1 (FoxM1) is associated with tumor development, progression and metastasis in a myriad of carcinomas, thus establishing it as an attractive target for anticancer drug development. FoxM1 overexpression in hepatocellular carcinoma is reflective of tumor aggressiveness and recurrence, poor prognosis and low survival in patients. In our study, we have identified the antimalarial natural product, Artemisinin, to efficiently curb FoxM1 expression and activity in hepatic cancer cells, thereby exhibiting potential anticancer efficacy. Here, we demonstrated that Artemisinin considerably mitigates FoxM1 transcriptional activity by disrupting its interaction with the promoter region of its downstream targets, thereby suppressing the expression of numerous oncogenic drivers. Augmented level of FoxM1 is implicated in drug resistance of cancer cells, including hepatic tumor cells. Notably, FoxM1 overexpression rendered HCC cells poorly responsive to Artemisinin-mediated cytotoxicity while FoxM1 depletion in resistant liver cancer cells sensitized them to Artemisinin treatment, manifested in lower proliferative and growth index, drop in invasive potential and repressed expression of EMT markers with a concomitantly increased apoptosis. Moreover, Artemisinin, when used in combination with Thiostrepton, an established FoxM1 inhibitor, markedly reduced anchorage-independent growth and displayed more pronounced death in liver cancer cells. We found this effect to be evident even in the resistant HCC cells, thereby putting forth a novel combination therapy for resistant cancer patients. Altogether, our findings provide insight into the pivotal involvement of FoxM1 in the tumor suppressive activities of Artemisinin and shed light on the potential application of Artemisinin for improved therapeutic response, especially in resistant hepatic malignancies. Considering that Artemisinin compounds are in current clinical use with favorable safety profiles, the results from our study will potentiate its utility in juxtaposition with established FoxM1 inhibitors, promoting maximal therapeutic efficacy with minimal adverse effects in liver cancer patients.
Collapse
Affiliation(s)
| | | | - Aakriti Singal
- Department of Biochemistry, University of Delhi, New Delhi, India
| | - Hina Bharti
- Department of Biochemistry, University of Delhi, New Delhi, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi, New Delhi, India
| |
Collapse
|
31
|
Liu C, Barger CJ, Karpf AR. FOXM1: A Multifunctional Oncoprotein and Emerging Therapeutic Target in Ovarian Cancer. Cancers (Basel) 2021; 13:3065. [PMID: 34205406 PMCID: PMC8235333 DOI: 10.3390/cancers13123065] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 02/08/2023] Open
Abstract
Forkhead box M1 (FOXM1) is a member of the conserved forkhead box (FOX) transcription factor family. Over the last two decades, FOXM1 has emerged as a multifunctional oncoprotein and a robust biomarker of poor prognosis in many human malignancies. In this review article, we address the current knowledge regarding the mechanisms of regulation and oncogenic functions of FOXM1, particularly in the context of ovarian cancer. FOXM1 and its associated oncogenic transcriptional signature are enriched in >85% of ovarian cancer cases and FOXM1 expression and activity can be enhanced by a plethora of genomic, transcriptional, post-transcriptional, and post-translational mechanisms. As a master transcriptional regulator, FOXM1 promotes critical oncogenic phenotypes in ovarian cancer, including: (1) cell proliferation, (2) invasion and metastasis, (3) chemotherapy resistance, (4) cancer stem cell (CSC) properties, (5) genomic instability, and (6) altered cellular metabolism. We additionally discuss the evidence for FOXM1 as a cancer biomarker, describe the rationale for FOXM1 as a cancer therapeutic target, and provide an overview of therapeutic strategies used to target FOXM1 for cancer treatment.
Collapse
Affiliation(s)
| | | | - Adam R. Karpf
- Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68918-6805, USA; (C.L.); (C.J.B.)
| |
Collapse
|
32
|
Lv S, Liu M, Xu L, Zhang C. Downregulation of decidual SKP2 is associated with human recurrent miscarriage. Reprod Biol Endocrinol 2021; 19:88. [PMID: 34116705 PMCID: PMC8194034 DOI: 10.1186/s12958-021-00775-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 06/01/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Recurrent miscarriage (RM) is a very frustrating problem for both couples and clinicians. To date, the etiology of RM remains poorly understood. Decidualization plays a critical role in implantation and the maintenance of pregnancy, and its deficiency is closely correlated with RM. The F-box protein S-phase kinase associated protein 2 (SKP2) is a key component of the SCF-type E3 ubiquitin ligase complex, which is critically involved in ErbB family-induced Akt ubiquitination, aerobic glycolysis and tumorigenesis. SKP2 is pivotal for reproduction, and SKP2-deficient mice show impaired ovarian development and reduced fertility. METHODS Here, we investigated the expression and function of SKP2 in human decidualization and its relation with RM. A total of 40 decidual samples were collected. Quantitative PCR analysis, western blot analysis and immunohistochemistry analysis were performed to analyze the differential expression of SKP2 between RM and control cells. For in vitro induction of decidualization, both HESCs (human endometrial stromal cells) cell line and primary ESCs (endometrial stromal cells) were used to analyze the effects of SKP2 on decidualization via siRNA transfection. RESULTS Compared to normal pregnant women, the expression of SKP2 was reduced in the decidual tissues from individuals with RM. After in vitro induction of decidualization, knockdown of SKP2 apparently attenuated the decidualization of HESCs and resulted in the downregulation of HOXA10 and FOXM1, which are essential for normal human decidualization. Moreover, our experiments demonstrated that SKP2 silencing reduced the expression of its downstream target GLUT1. CONCLUSIONS Our study indicates a functional role of SKP2 in RM: downregulation of SKP2 in RM leads to impaired decidualization and downregulation of GLUT1 and consequently predisposes individuals to RM.
Collapse
Affiliation(s)
- Shijian Lv
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Mei Liu
- Department of Obstetrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42 Wenhua Xi Road, Jinan, 250011, Shandong, China
| | - Lizhen Xu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
| |
Collapse
|
33
|
Lemay SE, Awada C, Shimauchi T, Wu WH, Bonnet S, Provencher S, Boucherat O. Fetal Gene Reactivation in Pulmonary Arterial Hypertension: GOOD, BAD, or BOTH? Cells 2021; 10:1473. [PMID: 34208388 PMCID: PMC8231250 DOI: 10.3390/cells10061473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension is a debilitating chronic disorder marked by the progressive obliteration of the pre-capillary arterioles. This imposes a pressure overload on the right ventricle (RV) pushing the latter to undergo structural and mechanical adaptations that inexorably culminate in RV failure and death. Thanks to the advances in molecular biology, it has been proposed that some aspects of the RV and pulmonary vascular remodeling processes are orchestrated by a subversion of developmental regulatory mechanisms with an upregulation of a suite of genes responsible for the embryo's early growth and normally repressed in adults. In this review, we present relevant background regarding the close relationship between overactivation of fetal genes and cardiopulmonary remodeling, exploring whether the reawakening of developmental factors plays a causative role or constitutes a protective mechanism in the setting of PAH.
Collapse
Affiliation(s)
- Sarah-Eve Lemay
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (S.-E.L.); (C.A.); (T.S.); (W.-H.W.); (S.B.); (S.P.)
| | - Charifa Awada
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (S.-E.L.); (C.A.); (T.S.); (W.-H.W.); (S.B.); (S.P.)
| | - Tsukasa Shimauchi
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (S.-E.L.); (C.A.); (T.S.); (W.-H.W.); (S.B.); (S.P.)
| | - Wen-Hui Wu
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (S.-E.L.); (C.A.); (T.S.); (W.-H.W.); (S.B.); (S.P.)
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (S.-E.L.); (C.A.); (T.S.); (W.-H.W.); (S.B.); (S.P.)
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (S.-E.L.); (C.A.); (T.S.); (W.-H.W.); (S.B.); (S.P.)
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (S.-E.L.); (C.A.); (T.S.); (W.-H.W.); (S.B.); (S.P.)
| |
Collapse
|
34
|
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
|
35
|
Ferreira FJ, Carvalho L, Logarinho E, Bessa J. foxm1 Modulates Cell Non-Autonomous Response in Zebrafish Skeletal Muscle Homeostasis. Cells 2021; 10:cells10051241. [PMID: 34070077 PMCID: PMC8158134 DOI: 10.3390/cells10051241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/01/2021] [Accepted: 05/11/2021] [Indexed: 12/23/2022] Open
Abstract
foxm1 is a master regulator of the cell cycle, contributing to cell proliferation. Recent data have shown that this transcription factor also modulates gene networks associated with other cellular mechanisms, suggesting non-proliferative functions that remain largely unexplored. In this study, we used CRISPR/Cas9 to disrupt foxm1 in the zebrafish terminally differentiated fast-twitching muscle cells. foxm1 genomic disruption increased myofiber death and clearance. Interestingly, this contributed to non-autonomous satellite cell activation and proliferation. Moreover, we observed that Cas9 expression alone was strongly deleterious to muscle cells. Our report shows that foxm1 modulates a muscle non-autonomous response to myofiber death and highlights underreported toxicity to high expression of Cas9 in vivo.
Collapse
Affiliation(s)
- Fábio J. Ferreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.J.F.); (L.C.)
- Vertebrate Development and Regeneration Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Aging and Aneuploidy Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Graduate Program in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal
| | - Leonor Carvalho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.J.F.); (L.C.)
- Vertebrate Development and Regeneration Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Elsa Logarinho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.J.F.); (L.C.)
- Aging and Aneuploidy Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence: (E.L.); (J.B.)
| | - José Bessa
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (F.J.F.); (L.C.)
- Vertebrate Development and Regeneration Group, IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence: (E.L.); (J.B.)
| |
Collapse
|
36
|
O'Reilly E, Zeinabad HA, Szegezdi E. Hematopoietic versus leukemic stem cell quiescence: Challenges and therapeutic opportunities. Blood Rev 2021; 50:100850. [PMID: 34049731 DOI: 10.1016/j.blre.2021.100850] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/22/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSC) are responsible for the production of mature blood cells. To ensure that the HSC pool does not get exhausted over the lifetime of an individual, most HSCs are in a state of quiescence with only a small proportion of HSCs dividing at any one time. HSC quiescence is carefully controlled by both intrinsic and extrinsic, niche-driven mechanisms. In acute myeloid leukemia (AML), the leukemic cells overtake the hematopoietic bone marrow niche where they acquire a quiescent state. These dormant AML cells are resistant to chemotherapeutics. Because they can re-establish the disease after therapy, they are often termed as quiescent leukemic stem cells (LSC) or leukemia-initiating cells. While advancements are being made to target particular driver mutations in AML, there is less focus on how to tackle the drug resistance of quiescent LSCs. This review summarises the current knowledge on the biochemical characteristics of quiescent HSCs and LSCs, the intracellular signaling pathways and the niche-driven mechanisms that control quiescence and the key differences between HSC- and LSC-quiescence that may be exploited for therapy.
Collapse
Affiliation(s)
- Eimear O'Reilly
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Hojjat Alizadeh Zeinabad
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Eva Szegezdi
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
37
|
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
|
38
|
Hsu CC, Liao WY, Chang KY, Chan TS, Huang PJ, Chiang CT, Shan YS, Cheng LH, Liao TY, Tsai KK. A multi-mode Wnt- and stemness-regulatory module dictated by FOXM1 and ASPM isoform I in gastric cancer. Gastric Cancer 2021; 24:624-639. [PMID: 33515163 DOI: 10.1007/s10120-020-01154-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gastric cancer (GC) is the third leading cause of cancer mortality globally and a molecularly heterogeneous disease. Identifying the driver pathways in GC progression is crucial to improving the clinical outcome. Recent studies identified ASPM (abnormal spindle-like microcephaly-associated) and FOXM1 (Forkhead box protein M1) as novel Wnt and cancer stem cell (CSC) regulators; their pathogenetic roles and potential crosstalks in GC remain unclarified. METHODS The expression patterns of ASPM isoforms and FOXM1 were profiled in normal gastric epithelial and GC tissues. The functional roles of ASPM and FOXM1 in Wnt activity, cancer stemness and GC progression, and the underlying signaling processes were investigated. RESULTS Approximately one third of GC cells upregulate the expression of ASPM isoform I (ASPMiI) in their cytoplasm; the tumors with a high ASPMiI positive score (≥ 10%) are associated with a poor prognosis of the patients. Mechanistically, the molecular interplay among FOXM1, ASPMiI and DVL3 was found to converge on β-catenin to control the Wnt activity and the stemness property of GC cells. This multi-mode Wnt-regulatory module serves to reinforce Wnt signals in CSCs by transcriptional regulation (FOXM1-ASPM), protein-protein interactions (ASPMiI-DVL3-β-catenin), and nuclear translocation (FOXM1-β-catenin). CONCLUSIONS This study illuminates a novel Wnt- and stemness-regulatory mechanism in GC cells and identifies a novel subset of FOXM1highASPMiIhigh GC with potential to guide Wnt- and stemness-related diagnostics and therapies.
Collapse
Affiliation(s)
- Chung-Chi Hsu
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City, 824410, Taiwan
| | - Wen-Ying Liao
- Laboratory of Advanced Molecular Therapeutics, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Xinyi Dist., Taipei City, 110301, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes (NHRIs), Tainan City, 704016, Taiwan
| | - Tze-Sian Chan
- Laboratory of Advanced Molecular Therapeutics, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Xinyi Dist., Taipei City, 110301, Taiwan
- School of Medicine, College of Medicine, Taipei Medical University, Taipei City, 110301, Taiwan
- Division of Gastroenterology, Wan Fang Hospital, Taipei Medical University, Taipei City, 110301, Taiwan
- Integrative Therapy Center for Gastroenterological Cancers, Wan Fang Hospital, Taipei Medical University, Taipei City, 110301, Taiwan
| | - Po-Jui Huang
- Laboratory of Advanced Molecular Therapeutics, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Xinyi Dist., Taipei City, 110301, Taiwan
- Division of Gastroenterology, Wan Fang Hospital, Taipei Medical University, Taipei City, 110301, Taiwan
- Integrative Therapy Center for Gastroenterological Cancers, Wan Fang Hospital, Taipei Medical University, Taipei City, 110301, Taiwan
| | - Chun-Ting Chiang
- Department of Pathology, National Cheng-Kung University Hospital, Tainan City, 704302, Taiwan
| | - Yan-Shen Shan
- Department of Surgery, National Cheng-Kung University Hospital, Tainan City, 704302, Taiwan
| | - Lin-Hsin Cheng
- Laboratory of Advanced Molecular Therapeutics, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Xinyi Dist., Taipei City, 110301, Taiwan
| | - Tai-Yan Liao
- Laboratory of Advanced Molecular Therapeutics, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Xinyi Dist., Taipei City, 110301, Taiwan
| | - Kelvin K Tsai
- Laboratory of Advanced Molecular Therapeutics, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Xinyi Dist., Taipei City, 110301, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes (NHRIs), Tainan City, 704016, Taiwan.
- Division of Gastroenterology, Wan Fang Hospital, Taipei Medical University, Taipei City, 110301, Taiwan.
- Integrative Therapy Center for Gastroenterological Cancers, Wan Fang Hospital, Taipei Medical University, Taipei City, 110301, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei City, 110301, Taiwan.
- TMU and Affiliated Hospitals Pancreatic Cancer Group, Taipei Medical University, Taipei City, 110301, Taiwan.
| |
Collapse
|
39
|
Forkhead Transcription Factors in Health and Disease. Trends Genet 2021; 37:460-475. [DOI: 10.1016/j.tig.2020.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
|
40
|
Allaman-Pillet N, Schorderet DF. Piperlongumine promotes death of retinoblastoma cancer cells. Oncotarget 2021; 12:907-916. [PMID: 33953844 PMCID: PMC8092346 DOI: 10.18632/oncotarget.27947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 04/01/2021] [Indexed: 01/08/2023] Open
Abstract
Retinoblastoma is the most common pediatric intraocular malignant tumor. While retinoblastoma initiation is triggered by the inactivation of both alleles of the retinoblastoma tumor suppressor gene (RB1) in the developing retina, tumor progression requires additional epigenetic changes, retinoblastoma genomes being quite stable. Although the management of RB has recently improved, new therapeutic agents are necessary to improve the treatment of advanced forms of retinoblastoma. In this report, we analyzed the pro-death effect of piperlongumine (PL), a natural compound isolated from Piper longum L., on two human retinoblastoma cell lines, WERI-Rb and Y79. The effects of PL on cell proliferation, cell death and cell cycle were investigated. PL effectively inhibited cell growth, impacted the cell cycle by decreasing the level of cyclins and CDK1 and increasing CDKN1A and triggered a caspase-3 independant cell death process in which reactive oxygen species (ROS) production is a major player. Indeed, PL toxicity in retinoblastoma cell lines was inhibited by a ROS scavenger N-acetyl-l-cysteine (NAC) treatment. These findings suggest that PL reduces tumor growth and induces cell death by regulating the cell cycle.
Collapse
Affiliation(s)
| | - Daniel F Schorderet
- Institute for Research in Ophthalmology, Sion, Switzerland.,University of Lausanne, Faculty of Biology and Medicine, Lausanne, Switzerland.,Ecole Polytechnique Fédérale de Lausanne, Faculty of Life Sciences, Lausanne, Switzerland
| |
Collapse
|
41
|
Chen Q, Zhou L, Chen F, Hu A, Wang K, Liang H, Dong J. Forkhead box F2 as a novel prognostic biomarker and potential therapeutic target in human cancers prone to bone metastasis: a meta-analysis. J Int Med Res 2021; 49:3000605211002372. [PMID: 33845605 PMCID: PMC8047092 DOI: 10.1177/03000605211002372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/08/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To undertake a systematic review and meta-analysis to evaluate the prognostic value of Forkhead box F2 (FOXF2) levels in different types of cancers prone to bone metastasis. METHODS A systematic search of publications listed in electronic databases (The Web of Science, EMBASE®, PubMed®, PMC, Science Direct and CNKI) from inception to 5 November 2020 was conducted. The hazard ratios (HRs) and 95% confidence intervals (95% CIs) were used to assess the relationship between FOXF2 levels and patient prognosis including overall survival (OS) and disease-free survival (DFS). RESULTS Sixteen studies enrolling 8461 participants were included in the meta-analysis. High levels of FOXF2 were a predictor of OS (HR: 0.66; 95% CI 0.51, 0.86) and DFS (HR: 0.60; 95% CI 0.48, 0.76). The trim-and-fill analysis, sensitivity analysis and subgroup analyses stratified by the study characteristics confirmed the robustness of the results. CONCLUSION These current findings indicate that high FOXF2 levels could be an indicator of a good prognosis in cancer patients with tumours that are prone to bone metastasis. FOXF2 levels might be a clinically important prognostic biomarker.
Collapse
Affiliation(s)
| | | | - Fancheng Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Annan Hu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ketao Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haifeng Liang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Curcio C, Brugiapaglia S, Bulfamante S, Follia L, Cappello P, Novelli F. The Glycolytic Pathway as a Target for Novel Onco-Immunology Therapies in Pancreatic Cancer. Molecules 2021; 26:1642. [PMID: 33804240 PMCID: PMC7998946 DOI: 10.3390/molecules26061642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal forms of human cancer, characterized by unrestrained progression, invasiveness and treatment resistance. To date, there are limited curative options, with surgical resection as the only effective strategy, hence the urgent need to discover novel therapies. A platform of onco-immunology targets is represented by molecules that play a role in the reprogrammed cellular metabolism as one hallmark of cancer. Due to the hypoxic tumor microenvironment (TME), PDA cells display an altered glucose metabolism-resulting in its increased uptake-and a higher glycolytic rate, which leads to lactate accumulation and them acting as fuel for cancer cells. The consequent acidification of the TME results in immunosuppression, which impairs the antitumor immunity. This review analyzes the genetic background and the emerging glycolytic enzymes that are involved in tumor progression, development and metastasis, and how this represents feasible therapeutic targets to counteract PDA. In particular, as the overexpressed or mutated glycolytic enzymes stimulate both humoral and cellular immune responses, we will discuss their possible exploitation as immunological targets in anti-PDA therapeutic strategies.
Collapse
Affiliation(s)
- Claudia Curcio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (C.C.); (S.B.); (S.B.); (L.F.); (P.C.)
- Centro Ricerche Medicina Sperimentale, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Silvia Brugiapaglia
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (C.C.); (S.B.); (S.B.); (L.F.); (P.C.)
- Centro Ricerche Medicina Sperimentale, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Sara Bulfamante
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (C.C.); (S.B.); (S.B.); (L.F.); (P.C.)
- Centro Ricerche Medicina Sperimentale, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Laura Follia
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (C.C.); (S.B.); (S.B.); (L.F.); (P.C.)
- Computer Science Department, University of Turin, 10126 Turin, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (C.C.); (S.B.); (S.B.); (L.F.); (P.C.)
- Centro Ricerche Medicina Sperimentale, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (C.C.); (S.B.); (S.B.); (L.F.); (P.C.)
- Centro Ricerche Medicina Sperimentale, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| |
Collapse
|
43
|
Yang X, Yang S, Song J, Yang W, Ji Y, Zhang F, Rao J. Dysregulation of miR-23b-5p promotes cell proliferation via targeting FOXM1 in hepatocellular carcinoma. Cell Death Discov 2021; 7:47. [PMID: 33723252 PMCID: PMC7960996 DOI: 10.1038/s41420-021-00440-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/29/2021] [Accepted: 02/13/2021] [Indexed: 12/12/2022] Open
Abstract
Growing evidence demonstrates that MicroRNAs (miRNAs) play an essential role in contributing to tumor development and progression. However, the underlying role and mechanisms of miR-23b-5p in hepatocellular carcinoma (HCC) formation remain unclear. Our study showed that miR-23b-5p was downregulated in the HCC tissues and cell lines, and lower expression of miR-23b-5p was associated with more severe tumor size and poorer survival. Gain- or loss-of-function assays demonstrated that miR-23b-5p induced G0/G1 cell cycle arrest and inhibited cell proliferation both in vitro and in vivo. qRT-PCR, western blot and luciferase assays verified that Mammalian transcription factor Forkhead Box M1 (FOXM1), upregulated in HCC specimens, was negatively correlated with miR-23b-5p expression and acted as a direct downstream target of miR-23b-5p. In addition, miR-23b-5p could regulate cyclin D1 and c-MYC expression by directly targeting FOXM1. Further study revealed that restoration of FOXM1 neutralized the cell cycle arrest and cell proliferation inhibition caused by miR-23b-5p. Taken together, our findings suggest that miR-23b-5p acted as a tumor suppressor role in HCC progression by targeting FOXM1 and may serve as a potential novel biomarker for HCC diagnosis and prognosis.
Collapse
Affiliation(s)
- Xinchen Yang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, China
| | - Shikun Yang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, China
| | - Jinhua Song
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, China
| | - Wenjie Yang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, China
| | - Yang Ji
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, China
| | - Feng Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, China.
| | - Jianhua Rao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, China.
| |
Collapse
|
44
|
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
|
45
|
Characterization of pathways involved in colorectal cancer using real-time RT-PCR gene expression data. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2021; 14:123-131. [PMID: 33968339 PMCID: PMC8101527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AIM Efforts to explore biomarkers and biological pathways involved in the disease are needed to improve colorectal cancer (CRC) diagnosis and alternative treatments. BACKGROUND The fourth common malignancy in the world is colorectal cancer. The over-all burden is predicted to rise by 2030. METHODS In the current study, nine genes were selected. Previously, a panel of genes by Agendia, a classifier of robust gene expression (ColoPrint), was determined to significantly improve the prognostic accuracy of pathologic factors in stage II and III colorectal cancer patients. Five genes, including Ppara, Mctp1, Pyroxd1, Il2r, and Cyfip2, from this panel and four other genes which were not in this panel but were cited abundantly in the literature were selected. Then, expression levels of the selected genes in CRC tissue were compared with levels in adjacent normal tissue. To identify the pathways involved in CRC, gene set enrichment analysis was subsequently performed. Furthermore, to illustrate the relationship between genes in this disease, the cross-shaped co-expression pattern and gene regulatory network were determined using computational methods. RESULTS This research found that the pairs of genes: {IL2R, CYFIP2}, {FOXM1, PPARA}, {MCTP1, CTSC}, and {PYROXD1, CYF1P2} are functionally related. Furthermore, two differentially expressed gene pairs ({FOXM1, PPARA} and {IL2R, CYFIP2}) are involved in the vascular endothelial growth factor receptor signaling pathway and the purine ribonucleoside diphosphate metabolic process, respectively. CONCLUSION This research found that the combination of computational analysis and laboratory data provided the opportunity to better characterize the relation between central colorectal cancer genes as well as possible pathways involved in the colorectal cancer.
Collapse
|
46
|
Chen M, Zhao S, Guo WH, Zhu YP, Pan L, Xie ZW, Sun WL, Jiang JT. Maternal exposure to Di-n-butyl phthalate (DBP) aggravate gestational diabetes mellitus via FoxM1 suppression by pSTAT1 signalling. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 205:111154. [PMID: 32810643 DOI: 10.1016/j.ecoenv.2020.111154] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/05/2020] [Accepted: 08/10/2020] [Indexed: 06/11/2023]
Abstract
The study focused on the toxicological effect of Di-n-butyl phthalate (DBP) on the expression of Phosphorylated signal transducer and activator of transcription 1 (pSTAT1) -regulated Forkhead box protein M1 (FoxM1), which might provide a new understanding of gestational diabetes mellitus (GDM) development and a potential target for treatment. Streptozotocin (STZ) (40 mg/kg) was introduced in maternal rats by intraperitoneal injection on gestation day 0 (GD 0) in the STZ and STZ + DBP groups. DBP was introduced in maternal rats by oral feeding in the STZ + DBP group over the following 3 days (750 mg/kg/day). The changes in fasting blood glucose level in rats were detected on GD 1 and GD 5. The insulin levels in maternal rats and PIBCs were measured on GD 18. The Oral Glucose Tolerance Test (OGTT) test was performed on GD 18 to check the stability of the GDM model. The primary islet β cells (PIBCs) were established for in vitro experiments. We examined the FoxM1 and pSTAT1 expression in pancreas by immunohistochemistry. Real-time PCR and Western blot were used to detect the pSTAR1 and FoxM1 protein and mRNA gene expression levels in PIBCs. Cell Counting Kit-8 (CCK-8) and flow cytometric analysis was used to test the viability and apoptosis of cells. The results showed that the STZ + DBP group had higher glucose and lower insulin secretion levels than the other groups by both fasting test and OGTT. FoxM1 was significantly suppressed while pSTAT1 was highly expressed after DBP exposure. FoxM1 could be regulated by pSTAT1. DBP can influence the progression of GDM through its toxicological effect, which significantly increases the expression of pSTAT1 and suppresses FoxM1, causing a decline in β cell viability.
Collapse
Affiliation(s)
- Min Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China
| | - Sheng Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China
| | - Wen-Huan Guo
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China
| | - Yi-Ping Zhu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China
| | - Lei Pan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China
| | - Zhi-Wen Xie
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China
| | - Wen-Lan Sun
- Department of Geriatrics, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China.
| | - Jun-Tao Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
47
|
Li Z, Yu DS, Doetsch PW, Werner E. Replication stress and FOXM1 drive radiation induced genomic instability and cell transformation. PLoS One 2020; 15:e0235998. [PMID: 33253193 PMCID: PMC7703902 DOI: 10.1371/journal.pone.0235998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/07/2020] [Indexed: 12/25/2022] Open
Abstract
In contrast to the vast majority of research that has focused on the immediate effects of ionizing radiation, this work concentrates on the molecular mechanism driving delayed effects that emerge in the progeny of the exposed cells. We employed functional protein arrays to identify molecular changes induced in a human bronchial epithelial cell line (HBEC3-KT) and osteosarcoma cell line (U2OS) and evaluated their impact on outcomes associated with radiation induced genomic instability (RIGI) at day 5 and 7 post-exposure to a 2Gy X-ray dose, which revealed replication stress in the context of increased FOXM1b expression. Irradiated cells had reduced DNA replication rate detected by the DNA fiber assay and increased DNA resection detected by RPA foci and phosphorylation. Irradiated cells increased utilization of homologous recombination-dependent repair detected by a gene conversion assay and DNA damage at mitosis reflected by RPA positive chromosomal bridges, micronuclei formation and 53BP1 positive bodies in G1, all known outcomes of replication stress. Interference with the function of FOXM1, a transcription factor widely expressed in cancer, employing an aptamer, decreased radiation-induced micronuclei formation and cell transformation while plasmid-driven overexpression of FOXM1b was sufficient to induce replication stress, micronuclei formation and cell transformation.
Collapse
Affiliation(s)
- Zhentian Li
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - David S. Yu
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Paul W. Doetsch
- Laboratory of Genomic Integrity and Structural Biology, NIH, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Erica Werner
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
48
|
FAM188B Expression Is Critical for Cell Growth via FOXM1 Regulation in Lung Cancer. Biomedicines 2020; 8:biomedicines8110465. [PMID: 33142744 PMCID: PMC7693245 DOI: 10.3390/biomedicines8110465] [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: 10/07/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/31/2022] Open
Abstract
Although family with sequence similarity 188 member B (FAM188B) is known to be a member of a novel putative deubiquitinase family, its biological role has not been fully elucidated. Here, we demonstrate the oncogenic function of FAM188B via regulation of forkhead box M1 (FOXM1), another oncogenic transcription factor, in lung cancer cells. FAM188B knockdown induced the inhibition of cell growth along with the downregulation of mRNA and protein levels of FOXM1. FAM188B knockdown also resulted in downregulation of Survivin and cell cycle-related proteins, which are direct targets of FOXM1. Interestingly, FOXM1 co-immunoprecipitated with FAM188B, and the levels of FOXM1 ubiquitination increased with FAM188B knockdown but decreased with FAM188B overexpression. In addition, in vivo xenograft of FAM188B siRNA (siFAM188B) RNA-treated cells resulted in the retardation of tumor growth compared with that in the control. Furthermore, protein levels of FAM188B and FOXM1 were elevated in the human lung cancer tissues, and FAM188B expression was negatively correlated with the overall survival of lung cancer patients. These results indicate that FAM188B exerts its oncogenic effects by regulating FOXM1 deubiquitination and thus its stability. Therefore, FAM188B might be a potential therapeutic target to control lung cancer progression.
Collapse
|
49
|
Abstract
Forkhead box O (FOXO) transcription factors regulate diverse biological processes, affecting development, metabolism, stem cell maintenance and longevity. They have also been increasingly recognised as tumour suppressors through their ability to regulate genes essential for cell proliferation, cell death, senescence, angiogenesis, cell migration and metastasis. Mechanistically, FOXO proteins serve as key connection points to allow diverse proliferative, nutrient and stress signals to converge and integrate with distinct gene networks to control cell fate, metabolism and cancer development. In consequence, deregulation of FOXO expression and function can promote genetic disorders, metabolic diseases, deregulated ageing and cancer. Metastasis is the process by which cancer cells spread from the primary tumour often via the bloodstream or the lymphatic system and is the major cause of cancer death. The regulation and deregulation of FOXO transcription factors occur predominantly at the post-transcriptional and post-translational levels mediated by regulatory non-coding RNAs, their interactions with other protein partners and co-factors and a combination of post-translational modifications (PTMs), including phosphorylation, acetylation, methylation and ubiquitination. This review discusses the role and regulation of FOXO proteins in tumour initiation and progression, with a particular emphasis on cancer metastasis. An understanding of how signalling networks integrate with the FOXO transcription factors to modulate their developmental, metabolic and tumour-suppressive functions in normal tissues and in cancer will offer a new perspective on tumorigenesis and metastasis, and open up therapeutic opportunities for malignant diseases.
Collapse
Affiliation(s)
- Yannasittha Jiramongkol
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK.
| |
Collapse
|
50
|
Vilgelm AE, Saleh N, Shattuck-Brandt R, Riemenschneider K, Slesur L, Chen SC, Johnson CA, Yang J, Blevins A, Yan C, Johnson DB, Al-Rohil RN, Halilovic E, Kauffmann RM, Kelley M, Ayers GD, Richmond A. MDM2 antagonists overcome intrinsic resistance to CDK4/6 inhibition by inducing p21. Sci Transl Med 2020; 11:11/505/eaav7171. [PMID: 31413145 DOI: 10.1126/scitranslmed.aav7171] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/17/2019] [Accepted: 07/12/2019] [Indexed: 12/13/2022]
Abstract
Intrinsic resistance of unknown mechanism impedes the clinical utility of inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6i) in malignancies other than breast cancer. Here, we used melanoma patient-derived xenografts (PDXs) to study the mechanisms for CDK4/6i resistance in preclinical settings. We observed that melanoma PDXs resistant to CDK4/6i frequently displayed activation of the phosphatidylinositol 3-kinase (PI3K)-AKT pathway, and inhibition of this pathway improved CDK4/6i response in a p21-dependent manner. We showed that a target of p21, CDK2, was necessary for proliferation in CDK4/6i-treated cells. Upon treatment with CDK4/6i, melanoma cells up-regulated cyclin D1, which sequestered p21 and another CDK inhibitor, p27, leaving a shortage of p21 and p27 available to bind and inhibit CDK2. Therefore, we tested whether induction of p21 in resistant melanoma cells would render them responsive to CDK4/6i. Because p21 is transcriptionally driven by p53, we coadministered CDK4/6i with a murine double minute (MDM2) antagonist to stabilize p53, allowing p21 accumulation. This resulted in improved antitumor activity in PDXs and in murine melanoma. Furthermore, coadministration of CDK4/6 and MDM2 antagonists with standard of care therapy caused tumor regression. Notably, the molecular features associated with response to CDK4/6 and MDM2 inhibitors in PDXs were recapitulated by an ex vivo organotypic slice culture assay, which could potentially be adopted in the clinic for patient stratification. Our findings provide a rationale for cotargeting CDK4/6 and MDM2 in melanoma.
Collapse
Affiliation(s)
- Anna E Vilgelm
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA. .,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| | - Nabil Saleh
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Rebecca Shattuck-Brandt
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kelsie Riemenschneider
- Department of Dermatology, University of Texas Southwestern, Medical Center, Dallas, TX 75390, USA
| | - Lauren Slesur
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sheau-Chiann Chen
- Division of Cancer Biostatistics, Department of Biostatistics, Vanderbilt University Center for Quantitative Sciences, Nashville, TN 37232, USA
| | - C Andrew Johnson
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jinming Yang
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Ashlyn Blevins
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Chi Yan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Douglas B Johnson
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rami N Al-Rohil
- Department of Pathology, Duke University, Durham, NC 27708, USA
| | - Ensar Halilovic
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Rondi M Kauffmann
- Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Mark Kelley
- Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Gregory D Ayers
- Division of Cancer Biostatistics, Department of Biostatistics, Vanderbilt University Center for Quantitative Sciences, Nashville, TN 37232, USA
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|