1
|
Lu X, Friedrich LJ, Efferth T. Natural products targeting tumour angiogenesis. Br J Pharmacol 2025; 182:2094-2136. [PMID: 37680009 DOI: 10.1111/bph.16232] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
Tumour angiogenesis is the formation of new blood vessels to support the growth of a tumour. This process is critical for tumour progression and metastasis, making it an attractive approach to cancer therapy. Natural products derived from plants, animals or microorganisms exert anti-angiogenic properties and can be used to inhibit tumour growth and progression. In this review, we comprehensively report on the current status of natural products against tumour angiogenesis from four perspectives until March 2023: (1) the role of pro-angiogenic factors and antiangiogenic factors in tumour angiogenesis; (2) the development of anti-tumour angiogenesis therapy (monoclonal antibodies, VEGFR-targeted small molecules and fusion proteins); (3) the summary of anti-angiogenic natural agents, including polyphenols, polysaccharides, alkaloids, terpenoids, saponins and their mechanisms of action, and (4) the future perspectives of anti-angiogenic natural products (bioavailability improvement, testing of dosage and side effects, combination use and discovery of unique natural-based compounds). Our review aims to better understand the potential of natural products for drug development in inhibiting tumour angiogenesis and further aid the effective transition of these outcomes into clinical trials. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lara Johanna Friedrich
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
2
|
Jawwad T, Kamkaew M, Phongkitkarun K, Chusorn P, Jamnongsong S, Lam EWF, Sampattavanich S. Exploring the Single-Cell Dynamics of FOXM1 Under Cell Cycle Perturbations. Cell Prolif 2025:e70019. [PMID: 40091487 DOI: 10.1111/cpr.70019] [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: 07/11/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
The cell cycle is crucial for maintaining normal cellular functions and preventing replication errors. FOXM1, a key transcription factor, plays a pivotal role in regulating cell cycle progression and is implicated in various physiological and pathological processes, including cancers like liver, prostate, breast, lung and colon cancer. Despite previous research, our understanding of FOXM1 dynamics under different cell cycle perturbations and its connection to heterogeneous cell fate decisions remains limited. In this study, we investigated FOXM1 behaviour in individual cells exposed to various perturbagens. We found that different drugs induce diverse responses due to heterogeneous FOXM1 dynamics at the single-cell level. Single-cell analysis identified six distinct cellular phenotypes: on-time cytokinesis, cytokinesis delay, cell cycle delay, G1 arrest, G2 arrest and cell death, observed across different drug types and doses. Specifically, treatments with PLK1, CDK1, CDK1/2 and Aurora kinase inhibitors revealed varied FOXM1 dynamics leading to heterogeneous cellular outcomes. Our findings affirm that the dynamics of FOXM1 are essential in shaping cellular outcomes, influencing the signals that dictate responses to various stimuli. Our results gave insights into how FOXM1 dynamics contribute to cell cycle fate decisions, especially under different cell cycle perturbations.
Collapse
Affiliation(s)
- Tooba Jawwad
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Maliwan Kamkaew
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kriengkrai Phongkitkarun
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Porncheera Chusorn
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Faculty of Liberal Arts and Science, Roi Et Rajabhat University, Roi Et, Thailand
| | - Supawan Jamnongsong
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Somponnat Sampattavanich
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
3
|
Greco F, Mallio CA. Radiomics and Radiogenomics Toward Personalized Management of Clear Cell Renal Cell Carcinoma: The Importance of FOXM1. Acad Radiol 2024; 31:3647-3649. [PMID: 39097509 DOI: 10.1016/j.acra.2024.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Affiliation(s)
- Federico Greco
- Department of Radiology, Cittadella della Salute, Azienda Sanitaria Locale di Lecce, Piazza Filippo Bottazzi, 2, 73100 Lecce, Italy; Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy.
| | - Carlo Augusto Mallio
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| |
Collapse
|
4
|
Shibui Y, Kohashi K, Hino Y, Tamaki A, Kinoshita I, Yamamoto H, Nakashima Y, Tajiri T, Oda Y. Expression of Forkhead Box M1 and Anticancer Effects of FOXM1 Inhibition in Epithelioid Sarcoma. J Transl Med 2024; 104:102093. [PMID: 38857782 DOI: 10.1016/j.labinv.2024.102093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
Epithelioid sarcoma (ES) is a rare aggressive sarcoma that, unlike most soft-tissue sarcomas, shows a tendency toward local recurrence and lymph node metastasis. Novel antitumor agents are needed for ES patients. Forkhead box transcription factor 1 (FOXM1) is a member of the Forkhead transcription factor family and is associated with multiple oncogenic functions; FOXM1 is known to be overexpressed and correlated with pathogenesis in various malignancies. In this study, we immunohistochemically analyzed FOXM1 expression levels and their clinical, clinicopathologic, and prognostic significance in 38 ES specimens. In addition, to investigate potential correlations between FOXM1 downregulation and oncologic characteristics, we treated ES cell lines with thiostrepton, a naturally occurring antibiotic that inhibits both small interfering RNA (siRNA) and FOXM1. In the analyses using ES samples, all 38 specimens were diagnosed as positive for FOXM1 by immunohistochemistry. We separated specimens into high (n = 19) and low (n = 19) FOXM1-protein expression groups by staining index score, and into large (n = 12), small (n = 25), and unknown (n = 1) tumor-size groups using a cutoff of 5 cm maximum diameter. Although there were significantly more samples with high FOXM1 expression in the large tumor group (P = .013), there were no significant differences with respect to age (P = 1.00), sex (P = .51), primary site of origin (P = .74), histologic subtypes (P = 1.00), depth (P = .74), or survival rate (P = .288) between the high and low FOXM1-protein expression groups. In the in vitro experiments using ES cell lines, FOXM1 siRNA and thiostrepton successfully downregulated FOXM1 mRNA and protein expression. Furthermore, downregulation of FOXM1 inhibited cell proliferation, drug resistance against chemotherapeutic agents, migration, and invasion and caused cell cycle arrest in the ES cell lines. Finally, cDNA microarray analysis data showed that FOXM1 regulated cIAP2, which is one of the apoptosis inhibitors activated by the TNFα-mediated NF-κB pathway. In conclusion, the FOXM1 gene may be a promising therapeutic target for ES.
Collapse
Affiliation(s)
- Yuichi Shibui
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba Hospital, Ibaraki, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Pathology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yuko Hino
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihiko Tamaki
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Izumi Kinoshita
- Department of Pathology, Kokura Memorial Hospital, Fukuoka, Japan
| | - Hidetaka Yamamoto
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuro Tajiri
- Department of Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
5
|
Ibrahim HM, Abdelrahman AE, Elwan A, Gharieb SA, Refaat M, Elmesallamy W, Salem AA. Clinicopathological Impact of FOXM1 and MMP-9 Immunohistochemical Expression in Different Grades of Intracranial Meningioma. Appl Immunohistochem Mol Morphol 2024; 32:292-304. [PMID: 38863278 DOI: 10.1097/pai.0000000000001205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 04/05/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVES To find predictive biomarkers for recurrence and progression of meningioma. BACKGROUND Despite great advances in meningioma treatment, the prognosis remained unfavorable due to the high recurrence rate. METHODS In this study, we evaluated the immunohistochemical expression of FOXM1, MMP-9, and Ki67 in 50 cases of intracranial meningioma to detect its potential role in meningioma progression, recurrence, and patients' survival. RESULTS Strong FOXM1 expression was detected in 20% of the cases and was significantly associated with meningioma grade ( P = 0.002) and peritumoral brain edema (PTBE; P <0.001). Strong MMP-9 expression was noted in 32% of the cases and was significantly associated with meningioma grade and PTBE ( P <0.001, P <0.001, respectively). High Ki67 was noted in 50% and significantly associated with tumor grade and PTBE ( P <0.001, P = 0.002, respectively). The follow-up period revealed that meningiomas with strong FOXM1, strong MMP-9, and high Ki67 expression were associated with tumor recurrence, shorter OS, and recurrence-free survival. Furthermore, up-regulation of FOXM1 and MMP-9 expression had a significant relation with poor clinical response to the therapy ( P = 0.010, P = 0. 001, respectively). However, high Ki67 cases were more sensitive to clinical therapy ( P = 0.005). CONCLUSION Strong FOXM1, strong MMP-9, and high Ki67 in meningiomas indicate highly aggressive tumors with a shortened survival rate, dismal outcome, and high risk of recurrence after the standard protocol of therapy.
Collapse
Affiliation(s)
| | | | - Amira Elwan
- Department of Clinical Oncology and Nuclear Medicine
| | | | | | - Wael Elmesallamy
- Department of Neurosurgery, Faculty of Medicine, Zagazig University, Egypt
| | | |
Collapse
|
6
|
Galal MA, Al-Rimawi M, Hajeer A, Dahman H, Alouch S, Aljada A. Metformin: A Dual-Role Player in Cancer Treatment and Prevention. Int J Mol Sci 2024; 25:4083. [PMID: 38612893 PMCID: PMC11012626 DOI: 10.3390/ijms25074083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer continues to pose a significant global health challenge, as evidenced by the increasing incidence rates and high mortality rates, despite the advancements made in chemotherapy. The emergence of chemoresistance further complicates the effectiveness of treatment. However, there is growing interest in the potential of metformin, a commonly prescribed drug for type 2 diabetes mellitus (T2DM), as an adjuvant chemotherapy agent in cancer treatment. Although the precise mechanism of action of metformin in cancer therapy is not fully understood, it has been found to have pleiotropic effects, including the modulation of metabolic pathways, reduction in inflammation, and the regulation of cellular proliferation. This comprehensive review examines the anticancer properties of metformin, drawing insights from various studies conducted in vitro and in vivo, as well as from clinical trials and observational research. This review discusses the mechanisms of action involving both insulin-dependent and independent pathways, shedding light on the potential of metformin as a therapeutic agent for different types of cancer. Despite promising findings, there are challenges that need to be addressed, such as conflicting outcomes in clinical trials, considerations regarding dosing, and the development of resistance. These challenges highlight the importance of further research to fully harness the therapeutic potential of metformin in cancer treatment. The aims of this review are to provide a contemporary understanding of the role of metformin in cancer therapy and identify areas for future exploration in the pursuit of effective anticancer strategies.
Collapse
Affiliation(s)
- Mariam Ahmed Galal
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Mohammed Al-Rimawi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | | | - Huda Dahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Samhar Alouch
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| |
Collapse
|
7
|
Chen J, Yang P, Li S, Feng Y. Increased FOXM1 Expression was Associated with the Prognosis and the Recruitment of Neutrophils in Endometrial Cancer. J Immunol Res 2023; 2023:5437526. [PMID: 37159818 PMCID: PMC10163965 DOI: 10.1155/2023/5437526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/27/2022] [Accepted: 04/05/2023] [Indexed: 05/11/2023] Open
Abstract
Background Although the biological functions of Forkhead box protein M1 (FOXM1) were explored in a variety of cancer, to date, however, little attention has been paid to the situation of FOXM1 in EC endometrial cancer (EC). Method Bioinformatics analysis, including GEPIA, TIMER, cBioPortal, LinkedOmics, and STRING were used to analyze the FOXM1 gene expression, genetic alteration, and immune cell infiltration in EC. IHC staining, qPCR, cell viability, and migration assay were applied to identify the functions of FOXM1 in EC. Results FOXM1 was highly expressed in EC tissues and closely correlated with the prognosis of EC patients. FOXM1 knockdown inhibited EC cell proliferation and invasion as well as migration. FOXM1 genetic alteration was verified in EC patients. Coexpression network of FOXM1 indicated that it had roles in the EC cell cycle and the infiltration of immune cells in EC. Furthermore, bioinformatic and immunohistochemical analysis indicated that FOXM1 induced the increased CD276 expression and also enhanced the neutrophil recruitment in EC. Conclusion Our present study discovered a novel role of FOXM1 in EC, suggesting FOXM1 could be treated as a potential prognostic biomarker and immunotherapeutic target in EC diagnosis and treatment.
Collapse
Affiliation(s)
- Jing Chen
- Department of Obstetrics and Gynecology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Pusheng Yang
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecology Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shaojing Li
- Department of Obstetrics and Gynecology, Shanghai Fengxian District Central Hospital, Shanghai, China
| | - Yichen Feng
- Department of Obstetrics and Gynecology, Shanghai Fengxian District Central Hospital, Shanghai, China
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
Aljerian K. FOXO1 and PAX5 Rearrangement in Alveolar Rhabdomyosarcoma in Saudi Pediatric Patients. Fetal Pediatr Pathol 2022; 42:385-393. [PMID: 36484735 DOI: 10.1080/15513815.2022.2154134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective: In this study, we investigate the molecular rearrangement of FOXO1 in alveolar rhabdomyosarcoma (ARHS) in Saudi pediatric patients. Method: We performed a molecular detection of molecular translocation in 30 pediatric cases of ARHS using FOXO1 dual color break-apart FISH probe (ZytoLight®, 13q14.11) and PAX5 dual color break-apart FISH probe (ZytoLight®, 9p13.2). Results: All analyzable cases of ARHS demonstrated FOXO1 translocation whereas PAX5 translocation was not detected in any case. Conclusion: Although the testing for PAX5 rearrangement was based on protein-protein network analysis, our study showed that PAX5 translocation is not conspicuous in ARHS. PAX7/3::FOXO1 fusion genes feature ARMS, rendering crossreactivity between PAX7 and PAX3 a possible explanation. Nevertheless, PAX5 immunoreactivity and molecular translocation could be an adjunctive pathway that is confined to aggressive ARMS.
Collapse
Affiliation(s)
- Khaldoon Aljerian
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Demirtas Korkmaz F, Dogan Turacli I, Esendagli G, Ekmekci A. Effects of thiostrepton alone or in combination with selumetinib on triple-negative breast cancer metastasis. Mol Biol Rep 2022; 49:10387-10397. [PMID: 36097108 DOI: 10.1007/s11033-022-07751-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE FoxM1 transcription factor contributes to tumor metastasis and poor prognosis in many cancers including triple-negative breast cancer (TNBC). In this study, we examined the effects of FoxM1 inhibitor Thiostrepton (THIO) alone or in combination with MEK inhibitor Selumetinib (SEL) on metastatic parameters in vitro and in vivo. METHODS Cell viability was determined by MTT assay. Immunoblotting and immunohistochemistry was used to assess metastasis-related protein expressions in 4T1 cells and its allograft tumor model in BALB/c mice. In vivo uPA activity was determined by enzymatic methods. RESULTS Both inhibitors were effective on the expressions of FoxM1, ERK, p-ERK, Twist, E-cadherin, and Vimentin alone or in combination in vitro. THIO significantly decreased 4T1 cell migration and changed the cell morphology from mesenchymal-like to epithelial-like structure. THIO was more effective than in combination with SEL in terms of metastatic protein expressions in vivo. THIO alone significantly inhibited mean tumor growth, decreased lung metastasis rate and tumor foci, however, no significant changes in these parameters were observed in the combined group. Immunohistochemically, FoxM1 expression intensity was decreased with THIO and its combination with SEL in the tumors. CONCLUSIONS This study suggests that inhibiting FoxM1 as a single target is more effective than combined treatment with MEK in theTNBC allograft model. The therapeutic efficacy of THIO should be investigated with further studies on appropriate drug delivery systems.
Collapse
Affiliation(s)
- Funda Demirtas Korkmaz
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara, Turkey. .,Department of Medical Biology, Faculty of Medicine, Giresun University, Giresun, 28100, Turkey.
| | - Irem Dogan Turacli
- Department of Medical Biology, Faculty of Medicine, Ufuk University, Ankara, Turkey
| | - Guldal Esendagli
- Department of Medical Pathology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Abdullah Ekmekci
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
10
|
Aberrant transcription factors in the cancers of the pancreas. Semin Cancer Biol 2022; 86:28-45. [PMID: 36058426 DOI: 10.1016/j.semcancer.2022.08.011] [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: 06/13/2022] [Revised: 08/15/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022]
Abstract
Transcription factors (TFs) are essential for proper activation of gene set during the process of organogenesis, differentiation, lineage specificity. Reactivation or dysregulation of TFs regulatory networks could lead to deformation of organs, diseases including various malignancies. Currently, understanding the mechanism of oncogenesis became necessity for the development of targeted therapeutic strategy for different cancer types. It is evident that many TFs go awry in cancers of the pancreas such as pancreatic ductal adenocarcinoma (PDAC) and pancreatic neuroendocrine neoplasms (PanNENs). These mutated or dysregulated TFs abnormally controls various signaling pathways in PDAC and PanNENs including RTK, PI3K-PTEN-AKT-mTOR, JNK, TGF-β/SMAD, WNT/β-catenin, SHH, NOTCH and VEGF which in turn regulate different hallmarks of cancer. Aberrant regulation of such pathways have been linked to the initiation, progression, metastasis, and resistance in pancreatic cancer. As of today, a number of TFs has been identified as crucial regulators of pancreatic cancer and a handful of them shown to have potential as therapeutic targets in pre-clinical and clinical settings. In this review, we have summarized the current knowledge on the role and therapeutic usefulness of TFs in PDAC and PanNENs.
Collapse
|
11
|
Li X, Liu W, Tao W. LINC00174 promotes cell proliferation and metastasis in renal clear cell carcinoma by regulating miR-612/FOXM1 axis. Immunopharmacol Immunotoxicol 2022; 44:746-756. [PMID: 35616230 DOI: 10.1080/08923973.2022.2082303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Kidney renal clear cell carcinoma (KIRC) is the most common pathological subtype of kidney tumor. Reportedly, LINC00174 is a key regulator in cancer progression. This study aims to clarify the role and molecular mechanism of LINC00174 in the progression of KIRC. METHODS LINC00174 expression in KIRC and its prognostic value were analyzed by bioinformatics. LINC00174, miR-612 and FOXM1 mRNA expression levels in KIRC clinical samples and cell lines were detected by qRT-PCR. After LINC00174 was overexpressed or knocked down, CCK-8, BrdU and Transwell assays were adopted to evaluate the proliferation and metastatic potential of KIRC cells. Bioinformatics and dual luciferase reporter assays were employed to validate the targeting relationship between miR-612 and LINC00174 or FOXM1 mRNA, respectively. Western blot assay was performed to detect FOXM1 protein expression in KIRC cells. RESULTS LINC00174 expression and FOXM1 expression were up-regulated in 42 cases of KIRC tissues (P < 0.001), while miR-612 expression was down-regulated (P < 0.001). LINC00174 overexpression or miR-612 inhibitor promoted the viability and proliferation of KIRC cells (P < 0.01). Migration and invasion of KIRC cells were promoted when the cells were transfected with LINC00174 overexpression or miR-612 inhibitor (P < 0.05). LINC00174 can competitively bind with miR-612 to repress the expression of miR-612, in turn up-regulate the expression of FOXM1 mRNA. CONCLUSION LINC00174 facilitates the proliferation and metastatic potential of KIRC cells via regulating the miR-612/FOXM1 axis.
Collapse
Affiliation(s)
- Xiaoshan Li
- Department of Urology, Yangtze River Shipping General Hospital, Wuhan 430010, Hubei, China
| | - Wei Liu
- Department of Urology, Yangtze River Shipping General Hospital, Wuhan 430010, Hubei, China
| | - Weixiong Tao
- Department of Urology, Yangtze River Shipping General Hospital, Wuhan 430010, Hubei, China
| |
Collapse
|
12
|
Okuni N, Honma Y, Urano T, Tamura K. Romidepsin and tamoxifen cooperatively induce senescence of pancreatic cancer cells through downregulation of FOXM1 expression and induction of reactive oxygen species/lipid peroxidation. Mol Biol Rep 2022; 49:3519-3529. [PMID: 35099714 DOI: 10.1007/s11033-022-07192-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although improvement has been made in therapeutic strategies against pancreatic carcinoma, overall survival has not significantly enhanced over the past decade. Thus, the establishment of better therapeutic regimens remains a high priority. METHODS Pancreatic cancer cell lines were incubated with romidepsin, an inhibitor of histone deacetylase, and tamoxifen, and their effects on cell growth, signaling and gene expression were analyzed. Xenografts of human pancreatic cancer CFPAC1 cells were medicated with romidepsin and tamoxifen to evaluate their effects on tumor growth. RESULTS The inhibition of the growth of pancreatic cancer cells induced by romidepsin and tamoxifen was effectively reduced by N-acetyl cysteine and α-tocopherol, respectively. The combined treatment greatly induced reactive oxygen species production and mitochondrial lipid peroxidation, and these effects were prevented by N-acetyl cysteine and α-tocopherol. Tamoxifen enhanced romidepsin-induced cell senescence. FOXM1 expression was markedly downregulated in pancreatic cancer cells treated with romidepsin, and tamoxifen further reduced FOXM1 expression in cells treated with romidepsin. Siomycin A, an inhibitor of FOXM1, induced senescence in pancreatic cancer cells. Similar results were obtained in knockdown of FOXM1 expression by siRNA. CONCLUSION Since FOXM1 is used as a prognostic marker and therapeutic target for pancreatic cancer, a combination of the clinically available drugs romidepsin and tamoxifen might be considered for the treatment of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Noriko Okuni
- Innovative Cancer Center, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Yoshio Honma
- Innovative Cancer Center, Shimane University, Izumo, Shimane, 693-8501, Japan.
- Department of Biochemistry, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan.
| | - Takeshi Urano
- Department of Biochemistry, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Kenji Tamura
- Innovative Cancer Center, Shimane University, Izumo, Shimane, 693-8501, Japan
| |
Collapse
|
13
|
Hasegawa T, Ishii M. Pathological Osteoclasts and Precursor Macrophages in Inflammatory Arthritis. Front Immunol 2022; 13:867368. [PMID: 35464401 PMCID: PMC9024112 DOI: 10.3389/fimmu.2022.867368] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/18/2022] [Indexed: 01/10/2023] Open
Abstract
Macrophages comprise a variety of subsets with diverse biological functions, including inflammation, tissue repair, regeneration, and fibrosis. In the bone marrow, macrophages differentiate into multinucleated osteoclasts, which have a unique bone-destroying capacity and play key roles in physiological bone remodelling. In contrast, osteoclasts are also involved in inflammatory bone erosion in arthritis and it has been unclear whether the osteoclasts in different tissue settings arise from similar monocytoid precursors and share similar phenotypes. Rapid progresses in the sequencing technologies have provided many important insights regarding the heterogeneity of different types of osteoclasts. The application of single-cell RNA sequencing (scRNA-seq) to the osteoclast precursor-containing macrophages enabled to identify the specific subpopulation differentiating into pathological mature osteoclasts in joints. Furthermore, an intravital imaging technology using two-photon microscopy has succeeded in visualizing the real-time dynamics of immune cells in the synovial microenvironment. These technologies together contributed to characterize the unique macrophages in the inflamed synovium, termed “arthritis-associated osteoclastogenic macrophages (AtoMs)”, causing the pathological bone destruction in inflammatory arthritis. Here, we review and discuss how novel technologies help to better understand the role of macrophages in inflammatory arthritis, especially focusing of osteoclastogenesis at the pannus-bone interface.
Collapse
Affiliation(s)
- Tetsuo Hasegawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- World Premier International Research Center Initiative (WPI)-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- *Correspondence: Masaru Ishii,
| |
Collapse
|
14
|
Pan C, Zhang L, Meng X, Qin H, Xiang Z, Gong W, Luo W, Li D, Han X. Chronic exposure to microcystin-LR increases the risk of prostate cancer and induces malignant transformation of human prostate epithelial cells. CHEMOSPHERE 2021; 263:128295. [PMID: 33297237 DOI: 10.1016/j.chemosphere.2020.128295] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/02/2020] [Accepted: 09/06/2020] [Indexed: 06/12/2023]
Abstract
Microcystins-LR (MC-LR) acts as a possible carcinogen for humans and causes a serious risk to public environmental health. The current study aimed to evaluate the interaction between MC-LR exposure and prostate cancer development and elucidate the underlying mechanism. In this study, mice were exposed to MC-LR at various doses for 180 days. MC-LR was able to induce the progression of prostatic intraepithelial neoplasia (PIN) and microinvasion. Furthermore, MC-LR notably increased angiogenesis and susceptibility to prostate cancer in vivo. In vitro, over 25 weeks of MC-LR exposure, normal human prostate epithelial (RWPE-1) cells increased secretion of matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9), and colony formation, features typical for cancer cells. These MC-LR-transformed prostate epithelial cells displayed increased expression of forkhead box M1 (FOXM1) and cyclooxygenase-2 (COX-2); abrogation of FOXM1 or COX-2 activity by specific inhibitors could abolish the invasion and migration of MC-LR-treated cells. In conclusion, we have provided compelling evidence demonstrating the induction of a malignant phenotype in human prostate epithelial cells and the in vivo development of prostate cancer by exposure to MC-LR, which might be a potential tumor promoter in the progression of prostate cancer.
Collapse
Affiliation(s)
- Chun Pan
- Immunology and Reproduction Biology Laboratory, State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China
| | - Ling Zhang
- Immunology and Reproduction Biology Laboratory, State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China
| | - Xiannan Meng
- Immunology and Reproduction Biology Laboratory, State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China
| | - Haixiang Qin
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Zou Xiang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Wenyue Gong
- Immunology and Reproduction Biology Laboratory, State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China
| | - Wenxin Luo
- Immunology and Reproduction Biology Laboratory, State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China
| | - Dongmei Li
- Immunology and Reproduction Biology Laboratory, State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory, State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
15
|
Shibui Y, Kohashi K, Tamaki A, Kinoshita I, Yamada Y, Yamamoto H, Taguchi T, Oda Y. The forkhead box M1 (FOXM1) expression and antitumor effect of FOXM1 inhibition in malignant rhabdoid tumor. J Cancer Res Clin Oncol 2020; 147:1499-1518. [PMID: 33221995 DOI: 10.1007/s00432-020-03438-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Malignant rhabdoid tumor (MRT) is a rare, highly aggressive sarcoma with an uncertain cell of origin. Despite the existing standard of intensive multimodal therapy, the prognosis of patients with MRT is very poor. Novel antitumor agents are needed for MRT patients. Forkhead box transcription factor 1 (FOXM1) is overexpressed and is correlated with the pathogenesis in several human malignancies. In this study, we identified the clinicopathological and prognostic values of the expression of FOXM1 and its roles in the progression of MRT. METHODS We investigated the FOXM1 expression levels and their clinical significance in 23 MRT specimens using immunohistochemistry and performed clinicopathologic and prognostic analyses. We also demonstrated correlations between the downregulation of FOXM1 and oncological characteristics using small interfering RNA (siRNA) and FOXM1 inhibitor in MRT cell lines. RESULTS Histopathological analyses revealed that primary renal MRTs showed significantly low FOXM1 protein expression levels (p = 0.032); however, there were no significant differences in other clinicopathological characteristics or the survival rate. FOXM1 siRNA and FOXM1 inhibitor (thiostrepton) successfully downregulated the mRNA and protein expression of FOXM1 in vitro and the downregulation of FOXM1 inhibited cell proliferation, drug resistance to chemotherapeutic agents, migration, invasion, and caused the cell cycle arrest and apoptosis of MRT cell lines. A cDNA microarray analysis showed that FOXM1 regulated FANCD2 and NBS1, which are key genes for DNA damage repair. CONCLUSION This study demonstrates that FOXM1 may serve as a promising therapeutic target for MRT.
Collapse
Affiliation(s)
- Yuichi Shibui
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akihiko Tamaki
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Izumi Kinoshita
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
16
|
Dey P, Wang A, Ziegler Y, Kim SH, El-Ashry D, Katzenellenbogen JA, Katzenellenbogen BS. Suppression of Tumor Growth, Metastasis, and Signaling Pathways by Reducing FOXM1 Activity in Triple Negative Breast Cancer. Cancers (Basel) 2020; 12:cancers12092677. [PMID: 32961773 PMCID: PMC7565743 DOI: 10.3390/cancers12092677] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Triple negative breast cancer is an aggressive subtype of breast cancer that frequently metastasizes. Because the transcription factor FOXM1 is highly upregulated in triple negative breast cancer and controls many cell activities that lead to cancer progression and metastasis, we sought to determine if FOXM1 inhibitory compounds could effectively suppress the invasiveness and progression of triple negative breast cancer cells and tumors. Our findings show that these compounds inhibit cell motility, invasiveness, and the expression of important proteins associated with epithelial to mesenchymal transition. These compounds also suppressed the proliferation and metastatic outgrowth of triple negative breast tumors. Thus, these findings highlight the crucial role of FOXM1 in promoting the progression and metastasis of these cancers, and suggest that FOXM1 inhibitory compounds may have therapeutic potential and prove beneficial in intervention against triple negative breast cancer. Abstract Metastasis-related complications account for the overwhelming majority of breast cancer mortalities. Triple negative breast cancer (TNBC), the most aggressive breast cancer subtype, has a high propensity to metastasize to distant organs, leading to poor patient survival. The forkhead transcription factor, FOXM1, is especially upregulated and overexpressed in TNBC and is known to regulate multiple signaling pathways that control many key cancer properties, including proliferation, invasiveness, stem cell renewal, and therapy resistance, making FOXM1 a critical therapeutic target for TNBC. In this study, we test the effectiveness of a novel class of 1,1-diarylethylene FOXM1 inhibitory compounds in suppressing TNBC cell migration, invasion, and metastasis using in vitro cell culture and in vivo tumor models. We show that these compounds inhibit the motility and invasiveness of TNBC MDA-MB-231 and DT28 cells, along with reducing the expression of important epithelial to mesenchymal transition (EMT) associated genes. Further, orthotopic tumor studies in NOD-SCID-gamma (NSG) mice demonstrate that these compounds reduce FOXM1 expression and suppress TNBC tumor growth as well as distant metastasis. Gene expression and protein analyses confirm the decreased levels of EMT factors and FOXM1-regulated target genes in tumors and metastatic lesions in the inhibitor-treated animals. The findings suggest that these FOXM1 suppressive compounds may have therapeutic potential in treating triple negative breast cancer, with the aim of reducing tumor progression and metastatic outgrowth.
Collapse
Affiliation(s)
- Parama Dey
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (P.D.); (A.W.); (Y.Z.)
| | - Alexander Wang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (P.D.); (A.W.); (Y.Z.)
| | - Yvonne Ziegler
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (P.D.); (A.W.); (Y.Z.)
| | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (S.H.K.); (J.A.K.)
| | - Dorraya El-Ashry
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - John A. Katzenellenbogen
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (S.H.K.); (J.A.K.)
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Benita S. Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (P.D.); (A.W.); (Y.Z.)
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Correspondence: ; Tel.: +1-217-333-9769
| |
Collapse
|
17
|
Jeong JH, Ryu JH. Broussoflavonol B from Broussonetia kazinoki Siebold Exerts Anti-Pancreatic Cancer Activity through Downregulating FoxM1. Molecules 2020; 25:E2328. [PMID: 32429421 PMCID: PMC7287790 DOI: 10.3390/molecules25102328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/11/2020] [Accepted: 05/15/2020] [Indexed: 11/22/2022] Open
Abstract
Pancreatic cancer has a high mortality rate due to poor rates of early diagnosis. One tumor suppressor gene in particular, p53, is frequently mutated in pancreatic cancer, and mutations in p53 can inactivate normal wild type p53 activity and increase expression of transcription factor forkhead box M1 (FoxM1). Overexpression of FoxM1 accelerates cellular proliferation and cancer progression. Therefore, inhibition of FoxM1 represents a therapeutic strategy for treating pancreatic cancer. Broussoflavonol B (BF-B), isolated from the stem bark of Broussonetia kazinoki Siebold has previously been shown to inhibit the growth of breast cancer cells. This study aimed to investigate whether BF-B exhibits anti-pancreatic cancer activity and if so, identify the underlying mechanism. BF-B reduced cell proliferation, induced cell cycle arrest, and inhibited cell migration and invasion of human pancreatic cancer PANC-1 cells (p53 mutated). Interestingly, BF-B down-regulated FoxM1 expression at both the mRNA and protein level. It also suppressed the expression of FoxM1 downstream target genes, such as cyclin D1, cyclin B1, and survivin. Cell cycle analysis showed that BF-B induced the arrest of G0/G1 phase. BF-B reduced the phosphorylation of extracellular signal-regulated kinase ½ (ERK½) and expression of ERK½ downstream effector c-Myc, which regulates cell proliferation. Furthermore, BF-B inhibited cell migration and invasion, which are downstream functional properties of FoxM1. These results suggested that BF-B could repress pancreatic cancer cell proliferation by inactivation of the ERK/c-Myc/FoxM1 signaling pathway. Broussoflavonol B from Broussonetia kazinoki Siebold may represent a novel chemo-therapeutic agent for pancreatic cancer.
Collapse
Affiliation(s)
| | - Jae-Ha Ryu
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Sookmyung Women’s University, Seoul 04310, Korea;
| |
Collapse
|
18
|
Baicalin suppresses the cell cycle progression and proliferation of prostate cancer cells through the CDK6/FOXM1 axis. Mol Cell Biochem 2020; 469:169-178. [DOI: 10.1007/s11010-020-03739-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
|
19
|
Sun HL, Men JR, Liu HY, Liu MY, Zhang HS. FOXM1 facilitates breast cancer cell stemness and migration in YAP1-dependent manner. Arch Biochem Biophys 2020; 685:108349. [DOI: 10.1016/j.abb.2020.108349] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/15/2020] [Accepted: 03/19/2020] [Indexed: 12/11/2022]
|
20
|
Yu JH, Im CY, Min SH. Function of PIN1 in Cancer Development and Its Inhibitors as Cancer Therapeutics. Front Cell Dev Biol 2020; 8:120. [PMID: 32258027 PMCID: PMC7089927 DOI: 10.3389/fcell.2020.00120] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/11/2020] [Indexed: 12/15/2022] Open
Abstract
Peptidyl-prolyl isomerase (PIN1) specifically binds and isomerizes the phosphorylated serine/threonine-proline (pSer/Thr-Pro) motif, which results in the alteration of protein structure, function, and stability. The altered structure and function of these phosphorylated proteins regulated by PIN1 are closely related to cancer development. PIN1 is highly expressed in human cancers and promotes cancer as well as cancer stem cells by breaking the balance of oncogenes and tumor suppressors. In this review, we discuss the roles of PIN1 in cancer and PIN1-targeted small-molecule compounds.
Collapse
Affiliation(s)
- Ji Hoon Yu
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, South Korea
| | - Chun Young Im
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, South Korea
| | - Sang-Hyun Min
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, South Korea
| |
Collapse
|
21
|
Chattopadhyay M, Kodela R, Santiago G, Le TTC, Nath N, Kashfi K. NOSH-aspirin (NBS-1120) inhibits pancreatic cancer cell growth in a xenograft mouse model: Modulation of FoxM1, p53, NF-κB, iNOS, caspase-3 and ROS. Biochem Pharmacol 2020; 176:113857. [PMID: 32061771 DOI: 10.1016/j.bcp.2020.113857] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer has poor survival rates and largely ineffective therapies. Aspirin is the prototypical anti-cancer agent but its long-term use is associated with significant side effects. NOSH-aspirin belongs to a new class of anti-inflammatory agents that were designed to be safer alternatives by releasing nitric oxide and hydrogen sulfide. In this study we evaluated the effects of NOSH-aspirin against pancreatic cancer using cell lines and a xenograft mouse model. NOSH-aspirin inhibited growth of MIA PaCa-2 and BxPC-3 pancreatic cancer cells with IC50s of 47 ± 5, and 57 ± 4 nM, respectively, while it did not inhibit growth of a normal pancreatic epithelial cell line at these concentrations. NOSH-aspirin inhibited cell proliferation, caused G0/G1 phase cycle arrest, leading to increased apoptosis. Treated cells displayed increases in reactive oxygen species (ROS) and caspase-3 activity. In MIA PaCa-2 cell xenografts, NOSH-aspirin significantly reduced tumor growth and tumor mass. Growth inhibition was due to reduced proliferation (decreased PCNA expression) and induction of apoptosis (increased TUNEL positive cells). Expressions of ROS, iNOS, and mutated p53 were increased; while that of NF-κB and FoxM1 that were high in vehicle-treated xenografts were significantly inhibited by NOSH-aspirin. Taken together, these molecular events and signaling pathways contribute to NOSH-aspirin mediated growth inhibition and apoptotic death of pancreatic cancer cells in vitro and in vivo.
Collapse
Affiliation(s)
- Mitali Chattopadhyay
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, United States
| | - Ravinder Kodela
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, United States
| | - Gabriela Santiago
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, United States
| | - Thuy Tien C Le
- Department of Biological and Chemical Sciences, New York Institute of Technology, NY 10023, United States
| | - Niharika Nath
- Department of Biological and Chemical Sciences, New York Institute of Technology, NY 10023, United States
| | - 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, United States; Avicenna Pharmaceuticals Inc., New York NY, United States; Graduate Program in Biology, City University of New York Graduate Center, New York NY, United States.
| |
Collapse
|
22
|
Chen Y, Mo L, Wang X, Chen B, Hua Y, Gong L, Yang F, Li Y, Chen F, Zhu G, Ni W, Zhang C, Cheng Y, Luo Y, Shi J, Qiu M, Wu S, Tan Z, Wang K. TPGS-1000 exhibits potent anticancer activity for hepatocellular carcinoma in vitro and in vivo. Aging (Albany NY) 2020; 12:1624-1642. [PMID: 31986488 PMCID: PMC7053644 DOI: 10.18632/aging.102704] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 01/02/2020] [Indexed: 12/15/2022]
Abstract
D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS1000) is the most active water-soluble derivative of vitamin E and has been widely used as a carrier of solvents, plasticizers, emulsifiers, absorbent agents and refractory drug delivery systems. However, its anti-hepatocellular carcinoma (HCC) properties have not been explored. HCC cells were treated with different concentrations of TPGS1000. Cell survival was tested by CCK8 assay, and cell migration was tested by wound healing and Transwell assay. EdU staining verified cell proliferation, and signalling pathway was assayed by Western blot analysis. The BALB/c-nu mouse xenograft model was established to test HCC cell growth in vivo. In vitro TPGS1000 significantly inhibited the viability and mobility of HCC cells (HepG2, Hep3B and Huh7) in a dose-dependent manner. Cell cycle analysis indicated that TPGS1000 treatment arrested the HCC cell cycle in the G0/G1 phase, and induction of cell apoptosis was confirmed by TUNEL and Annexin V-7-AAD staining. Further pharmacological analysis indicated that collapse of the transmembrane potential of mitochondria, increased ROS generation, PARP-induced cell apoptosis and FoxM1-p21-mediated cell cycle arresting, were involved in the anti-HCC activity of TPGS1000. Moreover, treatment in vivo with TPGS1000 effectively impaired the growth of HCC xenografts in nude mice.
Collapse
Affiliation(s)
- Yidan Chen
- Cancer Research Institute, Hangzhou Cancer Hospital, Zhejiang, China
| | - Liqin Mo
- Cancer Research Institute, Hangzhou Cancer Hospital, Zhejiang, China
| | - Xuan Wang
- Life Sciences Research Institute, College of Life and Environmental Sciences, Hangzhou Normal University, Zhejiang, China
| | - Bi Chen
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Yunfen Hua
- College of Pharmaceutical Science, Zhejiang University of Technology, Zhejiang, China
| | - Linyan Gong
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Fei Yang
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Yongqiang Li
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Fangfang Chen
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Guiting Zhu
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Wei Ni
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Cheng Zhang
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Yuming Cheng
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Yan Luo
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Junping Shi
- Oncology Department, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Mengsheng Qiu
- Life Sciences Research Institute, College of Life and Environmental Sciences, Hangzhou Normal University, Zhejiang, China
| | - Shixiu Wu
- Cancer Research Institute, Hangzhou Cancer Hospital, Zhejiang, China
| | - Zhou Tan
- Life Sciences Research Institute, College of Life and Environmental Sciences, Hangzhou Normal University, Zhejiang, China
| | - Kaifeng Wang
- Cancer Research Institute, Hangzhou Cancer Hospital, Zhejiang, China.,State Key Laboratory for Oncogenes and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai Cancer Institute, Shanghai, China
| |
Collapse
|
23
|
Hasegawa T, Kikuta J, Sudo T, Matsuura Y, Matsui T, Simmons S, Ebina K, Hirao M, Okuzaki D, Yoshida Y, Hirao A, Kalinichenko VV, Yamaoka K, Takeuchi T, Ishii M. Identification of a novel arthritis-associated osteoclast precursor macrophage regulated by FoxM1. Nat Immunol 2019; 20:1631-1643. [DOI: 10.1038/s41590-019-0526-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 09/24/2019] [Indexed: 12/12/2022]
|
24
|
Kim E, Zucconi BE, Wu M, Nocco SE, Meyers DJ, McGee JS, Venkatesh S, Cohen DL, Gonzalez EC, Ryu B, Cole PA, Alani RM. MITF Expression Predicts Therapeutic Vulnerability to p300 Inhibition in Human Melanoma. Cancer Res 2019; 79:2649-2661. [PMID: 30910803 PMCID: PMC6522293 DOI: 10.1158/0008-5472.can-18-2331] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/27/2018] [Accepted: 03/21/2019] [Indexed: 01/17/2023]
Abstract
Histone modifications, largely regulated by histone acetyltransferases (HAT) and histone deacetylases, have been recognized as major regulatory mechanisms governing human diseases, including cancer. Despite significant effort and recent advances, the mechanism by which the HAT and transcriptional coactivator p300 mediates tumorigenesis remains unclear. Here, we use a genetic and chemical approach to identify the microphthalmia-associated transcription factor (MITF) as a critical downstream target of p300 driving human melanoma growth. Direct transcriptional control of MITF by p300-dependent histone acetylation within proximal gene regulatory regions was coupled to cellular proliferation, suggesting a significant growth regulatory axis. Further analysis revealed forkhead box M1 (FOXM1) as a key effector of the p300-MITF axis driving cell growth that is selectively activated in human melanomas. Targeted chemical inhibition of p300 acetyltransferase activity using a potent and selective catalytic p300/CBP inhibitor demonstrated significant growth inhibitory effects in melanoma cells expressing high levels of MITF. Collectively, these data confirm the critical role of the p300-MITF-FOXM1 axis in melanoma and support p300 as a promising novel epigenetic therapeutic target in human melanoma. SIGNIFICANCE: These results show that MITF is a major downstream target of p300 in human melanoma whose expression is predictive of melanoma response to small-molecule inhibition of p300 HAT activity.
Collapse
Affiliation(s)
- Edward Kim
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Beth E. Zucconi
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| | - Muzhou Wu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Sarah E. Nocco
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - David J. Meyers
- Department of Pharmacology & Molecular Sciences, Johns Hopkins School of Medicine, 725 N. Wolfe Street, 316 Hunterian Building, Baltimore, Maryland 21205
| | - Jean S. McGee
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Samantha Venkatesh
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Daniel L. Cohen
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Estela C. Gonzalez
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Byungwoo Ryu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118
| | - Philip A. Cole
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115;,Corresponding Authors: Rhoda M. Alani 617-638-5517, Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118 (Lead Contact), Philip A. Cole 617-525-5208, Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| | - Rhoda M. Alani
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118;,Corresponding Authors: Rhoda M. Alani 617-638-5517, Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA 02118 (Lead Contact), Philip A. Cole 617-525-5208, Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
25
|
Lee HJ, Jeong JH, Ryu JH. Anti-pancreatic cancer activity of Z-ajoene from garlic: An inhibitor of the Hedgehog/Gli/FoxM1 axis. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
26
|
Yang K, Jiang B, Lu Y, Shu Q, Zhai P, Zhi Q, Li Q. FOXM1 promotes the growth and metastasis of colorectal cancer via activation of β-catenin signaling pathway. Cancer Manag Res 2019; 11:3779-3790. [PMID: 31118796 PMCID: PMC6501701 DOI: 10.2147/cmar.s185438] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Purpose Our previous study proved that FOXM1 regulates colorectal cancer (CRC) cell metastasis through epithelial–mesenchymal transition program. The aim of this study is to further explore the underlying mechanism of FOXM1 in CRC. Materials and methods In this study, we detected the mRNA and protein expressions of FOXM1 and β-catenin in CRC tissues and their corresponding normal-appearing tissues (NATs) by quantitative reverse transcription-PCR and western blot analysis, respectively. Then the potential link between FOXM1 and β-catenin in CRC tissues was analyzed. Furthermore, we systematically analyzed the biological functions of FOXM1 in CRC cells after reconstitution of FOXM1 expression in vitro. Moreover, the mechanism of FOXM1-promoted CRC progression by improving β-catenin nuclear translocation was also discussed. Results Our data demonstrated that FOXM1 and β-catenin were upregulated in CRC tissues compared with the corresponding NATs (P<0.05). Clinicopathologic analysis revealed that increased FOXM1 (or β-catenin) expression positively correlated with some clinicopathologic features, such as tumor size, TNM stage, lymphatic metastasis, and distant metastasis (P<0.05). Meanwhile, the possible relationships between FOXM1 and β-catenin in CRC samples were evaluated using SPSS software, and a significant positive correlation was found (P<0.05). In vitro data demonstrate that elevated FOXM1 expression exerted oncogenic effects on CRC via activation of β-catenin signaling pathway. The inhibition of β-catenin by siRNAs significantly attenuates FOXM1-induced malignant activities. Conclusion The data suggested that FOXM1/β-catenin is critical for malignancy of CRC, which may constitute a potential therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Kankan Yang
- Department of Gastrointestinal Surgery, Chaohu Hospital of Anhui Medical University, Hefei 238000, Anhui, China,
| | - Bing Jiang
- Department of Gastrointestinal Surgery, Chaohu Hospital of Anhui Medical University, Hefei 238000, Anhui, China,
| | - Yecai Lu
- Department of Gastrointestinal Surgery, Chaohu Hospital of Anhui Medical University, Hefei 238000, Anhui, China,
| | - Qingbing Shu
- Department of Gastrointestinal Surgery, Chaohu Hospital of Anhui Medical University, Hefei 238000, Anhui, China,
| | - Pan Zhai
- Department of Gastrointestinal Surgery, Chaohu Hospital of Anhui Medical University, Hefei 238000, Anhui, China,
| | - Qiaoming Zhi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China,
| | - Qixin Li
- Department of Gastrointestinal Surgery, Chaohu Hospital of Anhui Medical University, Hefei 238000, Anhui, China,
| |
Collapse
|
27
|
Chen C, Xu ZQ, Zong YP, Ou BC, Shen XH, Feng H, Zheng MH, Zhao JK, Lu AG. CXCL5 induces tumor angiogenesis via enhancing the expression of FOXD1 mediated by the AKT/NF-κB pathway in colorectal cancer. Cell Death Dis 2019; 10:178. [PMID: 30792394 PMCID: PMC6385313 DOI: 10.1038/s41419-019-1431-6] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/15/2022]
Abstract
The mechanisms underlying the role of CXCL5 in tumor angiogenesis have not been fully defined. Here, we examined the effect of CXCL5 on tumor angiogenesis in colorectal cancer (CRC). Immunohistochemistry was used to monitor the expression of CXCL5 and CD31 in CRC patients’ tissues. HUVEC cell lines stably transfected with shCXCR2 and shFOXD1 lentivirus plasmids were used in an in vitro study. Based on some molecular biological experiments in vitro and in vivo, we found that CXCL5 was upregulated in tumor tissues and that its level positively correlated with the expression of CD31. Next, we used recombinant human CXCL5 (rhCXCL5) to stimulate HUVECs and found that their tube formation ability, proliferation, and migration were enhanced by the activation of the AKT/NF-κB/FOXD1/VEGF-A pathway in a CXCR2-dependent manner. However, silencing of CXCR2 and FOXD1 or inhibition of the AKT and NF-κB pathways could attenuate the tube formation ability, proliferation, and migration of rhCXCL5-stimulated HUVECs in vitro. rhCXCL5 can promote angiogenesis in vivo in Matrigel plugs, and the overexpression of CXCL5 can also increase microvessel density in vivo in a subcutaneous xenotransplanted tumor model in nude mice. Taken together, our findings support CXCL5 as an angiogenic factor that can promote cell metastasis through tumor angiogenesis in CRC. Furthermore, we propose that FOXD1 is a novel regulator of VEGF-A. These observations open new avenues for therapeutic application of CXCL5 in tumor anti-angiogenesis.
Collapse
Affiliation(s)
- Chun Chen
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Gastroenterology Surgery Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute of Digestive Surgery, Shanghai, China
| | - Zhuo-Qing Xu
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Gastroenterology Surgery Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute of Digestive Surgery, Shanghai, China
| | - Ya-Ping Zong
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Gastroenterology Surgery Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bao-Chi Ou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao-Hui Shen
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Gastroenterology Surgery Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hao Feng
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Gastroenterology Surgery Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min-Hua Zheng
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Gastroenterology Surgery Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jing-Kun Zhao
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China. .,Gastroenterology Surgery Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Ai-Guo Lu
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China. .,Gastroenterology Surgery Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
28
|
Devor E, Santillan D, Scroggins S, Warrier A, Santillan M. Trimester-specific plasma exosome microRNA expression profiles in preeclampsia. J Matern Fetal Neonatal Med 2019; 33:3116-3124. [PMID: 30700172 DOI: 10.1080/14767058.2019.1569614] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective: To identify microRNAs (miRNAs) differentially expressed in plasma exosomes collected in women diagnosed with preeclampsia compared with women with uncomplicated pregnancies.Materials and methods: Exosomes were purified from plasma samples obtained at each trimester from four women subsequently diagnosed with preeclampsia and from five matched healthy controls. RNA was purified from the exosomes, and expression of 368 miRNAs was profiled using A-Set TaqMan low density array (TLDA).Results: One-third of the 368 miRNAs profiled are not expressed in exosomes. Further, those that are not expressed tend to be evolutionarily younger and have a significantly different mature sequence signature than do miRNAs that are expressed in exosomes. Among miRNAs that are expressed in exosomes, a total of eight (miR-134, miR-196b, miR-302c, miR-346, miR-376c, miR-486-3p, miR-590-5p, and miR-618) were found to display statistically significant differential expression between women who developed preeclampsia as compared with those who did not. Moreover, half of these miRNAs (miR-134, miR-376c, miR-486-3p, and miR-590-5p) displayed statistically significant differential expression in the first trimester.Conclusions: Not all miRNAs are expressed in exosomes. Those that tend to be evolutionarily older and have a significantly different mature sequence signature than those that are not. A few exosome-expressed miRNAs do display expression patterns in women subsequently diagnosed with preeclampsia that are significantly different than in women having an uncomplicated and, among these, several appear in the first trimester. These miRNAs are potential early markers of preeclampsia risk.
Collapse
Affiliation(s)
- Eric Devor
- Obstetrics and Gynecology, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Donna Santillan
- Obstetrics and Gynecology, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sabrina Scroggins
- Obstetrics and Gynecology, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Akshaya Warrier
- Obstetrics and Gynecology, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mark Santillan
- Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
29
|
Bai C, Liu X, Qiu C, Zheng J. FoxM1 is regulated by both HIF-1α and HIF-2α and contributes to gastrointestinal stromal tumor progression. Gastric Cancer 2019; 22:91-103. [PMID: 29948390 DOI: 10.1007/s10120-018-0846-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/04/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND FoxM1 plays important regulatory roles in a variety of diseases. However, the functional role of FoxM1 and mechanisms responsible for its expression in gastrointestinal stromal tumor (GIST) is not thoroughly understood. METHODS FoxM1 protein expression and biological function were examined in human GIST tissues and cells using immunohistochemistry, quantitative real-time PCR, western blot, CCK-8, wound-healing- and Matrigel invasion assays, respectively. The role of hypoxia-inducible factor (HIF) signaling in FoxM1 expression was investigated using chromatin immunoprecipitation and luciferase reporter and in vivo tumor growth assays. RESULTS FoxM1 was highly expressed in highly proliferative and migratory/invasive GIST specimens. Upregulation of FoxM1 was positively correlated with the expression of HIF-1α and HIF-2α in GIST specimens, and hypoxia-induced FoxM1 expression in GIST cells. Functionally, ectopic expression of FoxM1 significantly promoted GIST cell proliferation, cell cycle progression, migration and invasion, whereas the knockdown of endogenous FoxM1 of hypoxic GIST cells had the opposite effects. Molecularly, FoxM1 was transcriptionally regulated by HIF-2α under normoxia, whereas it was upregulated by both HIF-1α and HIF-2α under hypoxia. The xenograft tumor data further confirmed the regulated effect of HIF-1α and HIF-2α on FoxM1, and demonstrated that the simultaneous downregulation of both HIF-1α and HIF-2α inhibited GIST tumor growth. CONCLUSIONS Our data demonstrated the critical role of FoxM1 in promoting GIST progression and uncovered a novel HIF-1α/HIF-2α-FoxM1 axis. These findings identify FoxM1 as a possible new molecular target for designing novel therapeutic treatments to control GIST progression.
Collapse
Affiliation(s)
- Chenguang Bai
- Department of Pathology, Changhai Hospital, Second Military Medical University, 168, Changhai Rd., Shanghai, 200433, China
| | - Xiaohong Liu
- Department of Pathology, Changhai Hospital, Second Military Medical University, 168, Changhai Rd., Shanghai, 200433, China
| | - Cen Qiu
- Department of Pathology, Changhai Hospital, Second Military Medical University, 168, Changhai Rd., Shanghai, 200433, China
| | - Jianming Zheng
- Department of Pathology, Changhai Hospital, Second Military Medical University, 168, Changhai Rd., Shanghai, 200433, China.
| |
Collapse
|
30
|
Cao J, Li J, Sun L, Qin T, Xiao Y, Chen K, Qian W, Duan W, Lei J, Ma J, Ma Q, Han L. Hypoxia-driven paracrine osteopontin/integrin αvβ3 signaling promotes pancreatic cancer cell epithelial-mesenchymal transition and cancer stem cell-like properties by modulating forkhead box protein M1. Mol Oncol 2018; 13:228-245. [PMID: 30367545 PMCID: PMC6360359 DOI: 10.1002/1878-0261.12399] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/05/2018] [Accepted: 09/19/2018] [Indexed: 12/30/2022] Open
Abstract
Pancreatic stellate cells (PSCs), a key component of the tumor microenvironment, contribute to tumor invasion, metastasis, and chemoresistance. Osteopontin (OPN), a phosphorylated glycoprotein, is overexpressed in pancreatic cancer. However, OPN expression in PSCs and its potential roles in tumor–stroma interactions remain unclear. The present study first showed that OPN is highly expressed and secreted in activated PSCs driven by hypoxia, and this process is in a ROS‐dependent manner; in addition, OPN was shown to be involved in the PSC‐induced epithelial–mesenchymal transition (EMT) and cancer stem cell (CSC)‐like properties of pancreatic cancer cells (PCCs). Mechanistically, OPN from activated PSCs interacts with the transmembrane receptor integrin αvβ3 on PCCs to upregulate forkhead box protein M1 (FOXM1) expression and induce malignant phenotypes of PCCs. Moreover, the Akt and Erk pathways participate in OPN/integrin αvβ3 axis‐induced FOXM1 expression of PCCs. Our further analysis showed that OPN and FOXM1 are significantly upregulated in pancreatic cancer tissues and are associated with poor clinical outcome, indicating that OPN and FOXM1 might be considered as diagnostic and prognostic biomarkers for patients with pancreatic cancer. In conclusion, we show here for the first time that OPN promotes the EMT and CSC‐like properties of PCCs by activating the integrin αvβ3‐Akt/Erk‐FOXM1 cascade in a paracrine manner, suggesting that targeting the tumor microenvironment represents a promising therapeutic strategy in pancreatic cancer.
Collapse
Affiliation(s)
- Junyu Cao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Jie Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Liankang Sun
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Ke Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Jiguang Ma
- Department of Anesthesiology, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, China
| |
Collapse
|
31
|
Liu L, Wu J, Guo Y, Xie W, Chen B, Zhang Y, Li S, Hua Y, Peng B, Shen S. Overexpression of FoxM1 predicts poor prognosis of intrahepatic cholangiocarcinoma. Aging (Albany NY) 2018; 10:4120-4140. [PMID: 30580327 PMCID: PMC6326658 DOI: 10.18632/aging.101706] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/12/2018] [Indexed: 12/23/2022]
Abstract
FoxM1 is an oncoprotein that is significantly overexpressed in many malignancies including hepatocellular carcinoma, but its role in intrahepatic cholangiocarcinoma (ICC) remains unclear. This study explores the expression of FoxM1 in human ICC, its relationships with clinical outcomes, and its role in the proliferation, migration, and invasion of ICC in vitro and in vivo. The results show that FoxM1 was markedly elevated in tumor tissues versus the paired peritumoral tissues. Overexpression of FoxM1 was correlated with multiple tumor nodules, tumor size > 5 cm, positive lymph node metastasis and advanced TNM stage. Cox analysis revealed that overexpression of FoxM1 is an independent prognostic indicator for both the overall survival and disease-free survival of ICC patients after hepatectomy. Furthermore, up/downregulation of FoxM1 markedly promoted/inhibited ICC cell proliferation, migration, and invasion in vitro and in vivo. Bioinformatic analysis indicated that overexpression of FoxM1 resulted in the dysregulation of multiple signaling pathways in ICC, and selected components of some key signaling pathways such as c-Myc signaling were confirmed in vitro. In addition, overexpression of FoxM1 enhanced MMP-9 and MMP-2 protein expression in ICC cells. In conclusion, FoxM1 promotes ICC progression and is a reliable predictor of poor prognosis in ICC.
Collapse
Affiliation(s)
- Lingyun Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541000, Guangxi, China
- Equal contribution
| | - Jian Wu
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Equal contribution
| | - Yu Guo
- Department of General Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Equal contribution
| | - Wenxuan Xie
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Bin Chen
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yi Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Shaoqiang Li
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Yunpeng Hua
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Baogang Peng
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Shunli Shen
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
32
|
Wang Y, Wu M, Lei Z, Huang M, Li Z, Wang L, Cao Q, Han D, Chang Y, Chen Y, Liu X, Xue L, Mao X, Geng J, Chen Y, Dai T, Ren L, Wang Q, Yu H, Chen C, Chu X. Dysregulation of miR-6868-5p/FOXM1 circuit contributes to colorectal cancer angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:292. [PMID: 30486864 PMCID: PMC6264626 DOI: 10.1186/s13046-018-0970-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/19/2018] [Indexed: 11/10/2022]
Abstract
Background Transcription factor forkhead box M1 (FOXM1) is a crucial regulator in colorectal cancer (CRC) progression. However, the regulatory mechanisms causing dysregulation of FOXM1 in CRC remain unclear. Methods Dual-luciferase reporter assay was conducted to determine FOXM1 as miR-6868-5p target. The function of miR-6868-5p and FOXM1 in CRC angiogenesis was verified in vitro. Intratumoral injection model was constructed to explore the effect of miR-6868-5p on angiogenesis in vivo. Chromatin immunoprecipitation assays were used to assess direct binding of H3K27me3 to the miR-6868 promoter. Results Through integrated analysis, we identified miR-6868-5p as the potent regulator of FOXM1. Overexpression of miR-6868-5p in CRC cells inhibited the angiogenic properties of co-cultured endothelial cells, whereas silencing of miR-6868-5p had opposite effects. In vivo delivery of miR-6868-5p blocked tumor angiogenesis in nude mice, resulting in tumor growth inhibition. Rescue of FOXM1 reversed the effect of miR-6868-5p on tumor angiogenesis. Further mechanistic study revealed that FOXM1 promoted the production of IL-8, which was responsible for the miR-6868-5p/FOXM1 axis-regulated angiogenesis. Reciprocally, FOXM1 inhibited miR-6868-5p expression through EZH2-mediated H3K27me3 on miR-6868-5p promoter, thus forming a feedback circuit. Clinically, the level of miR-6868-5p was downregulated in CRC tissues and inversely correlated with microvessel density as well as levels of FOXM1 and IL-8 in tumor specimens. Conclusions Together, these data identify miR-6868-5p as a novel determinant of FOXM1 expression and establish a miR-6868-5p/FOXM1 regulatory circuit for CRC angiogenesis, providing potential target for CRC treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-0970-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ye Wang
- Departments of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing, Jiangsu Province, China
| | - Meijuan Wu
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zengjie Lei
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Mengxi Huang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhiping Li
- Departments of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing, Jiangsu Province, China
| | - Liya Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qijun Cao
- Departments of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing, Jiangsu Province, China
| | - Dong Han
- Departments of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing, Jiangsu Province, China
| | - Yue Chang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yanyan Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaobei Liu
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Lijun Xue
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaobei Mao
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Jian Geng
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yanan Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tingting Dai
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Lili Ren
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qian Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hongju Yu
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Cheng Chen
- Departments of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing, Jiangsu Province, China. .,Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Xiaoyuan Chu
- Departments of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical University, Nanjing, Jiangsu Province, China. .,Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
33
|
Harada K, Ferdous T, Minami H, Mishima K. Prognostic significance of FOXM1 in oral squamous cell carcinoma patients treated by docetaxel-containing regimens. Mol Clin Oncol 2018; 10:29-36. [PMID: 30655974 PMCID: PMC6314082 DOI: 10.3892/mco.2018.1770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 09/21/2018] [Indexed: 01/27/2023] Open
Abstract
Forkhead box protein M1 (FOXM1) is an oncoprotein that is involved in cell proliferation, differentiation and aging, and overexpression of FOXM1 is thought to be associated with the development and progression of various types of cancer. The expression of FOXM1 was retrospectively examined in tumor tissues taken from 56 oral squamous cell carcinoma (OSCC) patients by immunohistochemical staining. All of these patients received docetaxel (Doc)-containing regimens as treatments against OSCC. The association between FOXM1 expression and the clinicopathological characteristics and prognosis of these patients was then examined. FOXM1 was expressed in the nucleus and cytoplasm of OSCC tissues samples. There was a significant association between FOXM1 expression in tumor tissues and N classification (P=0.0395), stage (P=0.004), therapeutic efficacy (P=0.0113) and outcome (P=0.0134) of patients. However, FOXM1 expression had no association with patients' sex, age or T classification. Additionally, high expression of FOXM1 in tumor cells was associated with a shorter overall survival (P=0.0257) of patients. Multivariate analysis also revealed that elevated expression of FOXM1 was a predictor of patients' poor survival (P=0.0327). The results suggested that high expression of FOXM1 in OSCC tumors may result in reduced therapeutic effects and poor clinical outcomes of patients receiving Doc-based treatment regimens.
Collapse
Affiliation(s)
- Koji Harada
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Tarannum Ferdous
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Haruyasu Minami
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Katsuaki Mishima
- Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
34
|
Ramezani A, Nikravesh H, Faghihloo E. The roles of FOX proteins in virus-associated cancers. J Cell Physiol 2018; 234:3347-3361. [PMID: 30362516 DOI: 10.1002/jcp.27295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022]
Abstract
Forkhead box (FOX) proteins play a crucial role in regulating the expression of genes involved in multiple biological processes, such as metabolism, development, differentiation, proliferation, apoptosis, migration, invasion, and longevity. Deregulation of FOX proteins is commonly associated with cancer initiation, progression, and chemotherapeutic drug resistance in many human tumors. FOX proteins deregulate through genetic events and the perturbation of posttranslational modification. The purpose of the present review is to describe the deregulation of FOX proteins by oncoviruses. Oncoviruses utilize various mechanisms to deregulate FOX proteins, including alterations in posttranslational modifications, cellular localization independently of posttranslational modifications, virus-encoded miRNAs, activation or suppression of a series of cell signaling pathways. This deregulation can affect proliferation, metastasis, chemotherapy resistance, and immunosuppression in virus-induced cancers and help to chronic viral infection, development of gluconeogenic responses, and inflammation. Since the PI3K/Akt/mTOR signaling pathway is the upstream FOXO, suppressing it can cause FOXO function to return, and this can be one of the reasons for patients to recover from the infection of the viruses used to treat these inhibitors. Hence, FOX proteins could serve as prognosis markers and target therapy specifically in cancers caused by oncoviruses.
Collapse
Affiliation(s)
- Ali Ramezani
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hojatolla Nikravesh
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ebrahim Faghihloo
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Retraction: Down-regulation of Forkhead Box M1 Transcription Factor Leads to the Inhibition of Invasion and Angiogenesis of Pancreatic Cancer Cells. Cancer Res 2018; 78:5470. [PMID: 30217880 DOI: 10.1158/0008-5472.can-18-1172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Prolyl isomerase Pin1: a promoter of cancer and a target for therapy. Cell Death Dis 2018; 9:883. [PMID: 30158600 PMCID: PMC6115400 DOI: 10.1038/s41419-018-0844-y] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/15/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022]
Abstract
Pin1 is the only known peptidyl-prolyl cis–trans isomerase (PPIase) that specifically recognizes and isomerizes the phosphorylated Serine/Threonine-Proline (pSer/Thr-Pro) motif. The Pin1-mediated structural transformation posttranslationally regulates the biofunctions of multiple proteins. Pin1 is involved in many cellular processes, the aberrance of which lead to both degenerative and neoplastic diseases. Pin1 is highly expressed in the majority of cancers and its deficiency significantly suppresses cancer progression. According to the ground-breaking summaries by Hanahan D and Weinberg RA, the hallmarks of cancer comprise ten biological capabilities. Multiple researches illuminated that Pin1 contributes to these aberrant behaviors of cancer via promoting various cancer-driving pathways. This review summarized the detailed mechanisms of Pin1 in different cancer capabilities and certain Pin1-targeted small-molecule compounds that exhibit anticancer activities, expecting to facilitate anticancer therapies by targeting Pin1.
Collapse
|
37
|
Wu M, Zhao H, Guo L, Wang Y, Song J, Zhao X, Li C, Hao L, Wang D, Tang J. Ultrasound-mediated nanobubble destruction (UMND) facilitates the delivery of A10-3.2 aptamer targeted and siRNA-loaded cationic nanobubbles for therapy of prostate cancer. Drug Deliv 2018; 25:226-240. [PMID: 29313393 PMCID: PMC6058493 DOI: 10.1080/10717544.2017.1422300] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The Forkhead box M1 (FoxM1) transcription factor is an important anti-tumor target. A novel targeted ultrasound (US)-sensitive nanobubble that is likely to make use of the physical energy of US exposure for the improvement of delivery efficacy to target tumors and specifically silence FoxM1 expression appears as among the most potential nanocarriers in respect of drug delivery. In this study, we synthesized a promising anti-tumor targeted FoxM1 siRNA-loaded cationic nanobubbles (CNBs) conjugated with an A10-3.2 aptamer (siFoxM1-Apt-CNBs), which demonstrate high specificity when binding to prostate-specific membrane antigen (PSMA) positive LNCaP cells. Uniform nanoscaled siFoxM1-Apt-CNBs were developed using a thin-film hydration sonication, carbodiimide chemistry approaches, and electrostatic adsorption methods. Fluorescence imaging as well as flow cytometry evidenced the fact that the siFoxM1-Apt-CNBs were productively developed and that they specifically bound to PSMA-positive LNCaP cells. siFoxM1-Apt-CNBs combined with ultrasound-mediated nanobubble destruction (UMND) significantly improved transfection efficiency, cell apoptosis, and cell cycle arrest in vitro while reducing FoxM1 expression. In vivo xenografts tumors in nude-mouse model results showed that siFoxM1-Apt-CNBs combined with UMND led to significant inhibition of tumor growth and prolonged the survival of the mice, with low toxicity, an obvious reduction in FoxM1 expression, and a higher apoptosis index. Our study suggests that siFoxM1-Apt-CNBs combined with UMND might be a promising targeted gene delivery strategy for therapy of prostate cancer.
Collapse
Affiliation(s)
- Meng Wu
- a Department of Ultrasound , Chinese PLA General Hospital , Beijing , China.,b School of Medicine , Nankai University , Tianjin , China
| | - Hongyun Zhao
- c Department of Gastroenterology , The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging , Chongqing , China
| | - Liang Guo
- a Department of Ultrasound , Chinese PLA General Hospital , Beijing , China
| | - Yiru Wang
- a Department of Ultrasound , Chinese PLA General Hospital , Beijing , China
| | - Jiao Song
- d Department of Obstetrics and Gynecology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Xueli Zhao
- e Ultrasound Department , Xijing Hospital, Fourth Military Medical University , Xi'an , China
| | - Chongyan Li
- f State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology , Chongqing Medical University , Chongqing , China
| | - Lan Hao
- g Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Dong Wang
- h Department of Ultrasound , The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Jie Tang
- a Department of Ultrasound , Chinese PLA General Hospital , Beijing , China
| |
Collapse
|
38
|
Xiao Z, Jia Y, Jiang W, Wang Z, Zhang Z, Gao Y. FOXM1: A potential indicator to predict lymphatic metastatic recurrence in stage IIA esophageal squamous cell carcinoma. Thorac Cancer 2018; 9:997-1004. [PMID: 29877046 PMCID: PMC6068428 DOI: 10.1111/1759-7714.12776] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/03/2018] [Accepted: 05/05/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Previous studies have elucidated that FOXM1 may predict poor prognosis in patients with multiple solid malignant tumors. In this study we explored the differential expression of FOXM1 in stage IIA esophageal squamous cell carcinoma (ESCC) and investigated its prognostic value. METHODS Immunohistochemistry (IHC) and Western blot were used to detect FOXM1 expression in ESCC. Correlations between FOXM1 expression and clinicopathological variables, and five-year lymphatic metastatic recurrence (LMR) and overall survival (OS) of patients were analyzed. RESULTS FOXM1 was aberrantly expressed in ESCC. Statistical analysis revealed a close relationship between FOXM1 expression and tumor size (P = 0.024), depth of invasion (P = 0.048), and degree of differentiation (P = 0.043). The five-year LMR of patients in the FOXM1 overexpression group was significantly increased compared to the low expression group (P = 0.001). The five-year OS of patients in the FOXM1 overexpression group was significantly reduced compared to the low expression group (P = 0.007). Log-rank tests demonstrated that large tumor size (P = 0.044), poor differentiation degree (P = 0.005), deep invasion (P = 0.000), and FOXM1 overexpression (P = 0.007) may indicate poor prognosis in stage IIA ESCC. Cox multivariate regression analysis revealed that all of these variables were independent predictors of unfavorable outcome (P < 0.05). CONCLUSION FOXM1 could be a predictor of lymphatic metastatic recurrence in stage IIA ESCC after Ivor Lewis esophagectomy.
Collapse
Affiliation(s)
- Zhaohua Xiao
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yang Jia
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Wenpeng Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhou Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhiping Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Department of Thoracic Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Yanyun Gao
- Department of Gynaecology and Obstetrics, Jining Traditional Chinese Medicine Hospital, Jining, China
| |
Collapse
|
39
|
Bell R, Barraclough R, Vasieva O. Gene Expression Meta-Analysis of Potential Metastatic Breast Cancer Markers. Curr Mol Med 2018; 17:200-210. [PMID: 28782484 PMCID: PMC5748874 DOI: 10.2174/1566524017666170807144946] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/20/2017] [Accepted: 07/30/2017] [Indexed: 12/15/2022]
Abstract
Background: Breast cancer metastasis is a highly prevalent cause of death for European females. DNA microarray analysis has established that primary tumors, which remain localized, differ in gene expression from those that metastasize. Cross-analysis of these studies allow to revile the differences that may be used as predictive in the disease prognosis and therapy. Objective: The aim of the project was to validate suggested prognostic and therapeutic markers using meta-analysis of data on gene expression in metastatic and primary breast cancer tumors. Method: Data on relative gene expression values from 12 studies on primary breast cancer and breast cancer metastasis were retrieved from Genevestigator (Nebion) database. The results of the data meta-analysis were compared with results of literature mining for suggested metastatic breast cancer markers and vectors and consistency of their reported differential expression. Results: Our analysis suggested that transcriptional expression of the COX2 gene is significantly downregulated in metastatic tissue compared to normal breast tissue, but is not downregulated in primary tumors compared with normal breast tissue and may be used as a differential marker in metastatic breast cancer diagnostics. RRM2 gene expression decreases in metastases when compared to primary breast cancer and could be suggested as a marker to trace breast cancer evolution. Our study also supports MMP1, VCAM1, FZD3, VEGFC, FOXM1 and MUC1 as breast cancer onset markers, as these genes demonstrate significant differential expression in breast neoplasms compared with normal breast tissue. Conclusion: COX2 and RRM2 are suggested to be prominent markers for breast cancer metastasis. The crosstalk between upstream regulators of genes differentially expressed in primary breast tumors and metastasis also suggests pathways involving p53, ER1, ERB-B2, TNF and WNT, as the most promising regulators that may be considered for new complex drug therapeutic interventions in breast cancer metastatic progression.
Collapse
Affiliation(s)
- R Bell
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB. United Kingdom
| | - R Barraclough
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB. United Kingdom
| | - O Vasieva
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB. United Kingdom
| |
Collapse
|
40
|
Cai P, Xiao Z, Pan T, Wen X, Cao J, Ouyang B. Lx2-32c inhibits the formation of mammosphere from MDA-MB-231 cells and induces apoptosis involving in down-regulating FoxM1. Biomed Pharmacother 2018; 102:1176-1181. [PMID: 29710535 DOI: 10.1016/j.biopha.2018.03.143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/22/2018] [Accepted: 03/23/2018] [Indexed: 11/18/2022] Open
Abstract
Cancer stem cells (CSCs) are a subset of cancer cells which have self-renewal ability and exist in various tumors. Inhibition of CSCs self-renewal is considered as a new method for tumor therapy. A novel semi-synthetic taxane analogue, Lx2-32c, could overcome drug resistance in various cancer cell lines. In this study, it was found that Lx2-32c inhibited the proliferation and mammosphere formation of MDA-MB-231-derived cancer stem cell-like cells (MCSCLCs) and induced apoptosis, as well as down-regulated the expression of FoxM1 and CD44 in MCSCLCs. Simultaneously, it was proved that Lx2-32c combined with thiostreption, a FoxM1 inhibitor inhibited proliferation and mammosphere formation of MCSCLCs and induced apoptosis to a more extent than Lx2-32c alone; thiostreption could also enhance the effect of Lx2-32c of reduction of the expression of FoxM1 and CD44. All of these results indicated that Lx2-32c is a novel semi-synthetic taxane analogue which inhibits the self-renewal of MCSCLCs cells and induces apoptosis involving in down-regulating FoxM1.
Collapse
Affiliation(s)
- Pei Cai
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, PR China
| | - Zuoqi Xiao
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, PR China
| | - Tao Pan
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, PR China
| | - Xiaoke Wen
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, PR China.
| | - Jianguo Cao
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, PR China.
| | - Bo Ouyang
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, PR China.
| |
Collapse
|
41
|
Rinaldetti S, Wirtz RM, Worst TS, Eckstein M, Weiss CA, Breyer J, Otto W, Bolenz C, Hartmann A, Erben P. FOXM1 predicts overall and disease specific survival in muscle-invasive urothelial carcinoma and presents a differential expression between bladder cancer subtypes. Oncotarget 2018; 8:47595-47606. [PMID: 28498805 PMCID: PMC5564590 DOI: 10.18632/oncotarget.17394] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/10/2017] [Indexed: 02/07/2023] Open
Abstract
Forkhead box M1 (FOXM1) is a late cell cycle gene that plays a crucial role in carcinogenesis and chemotherapeutic drug resistance. In this study, the impact of FOXM1 expression on patient outcome was investigated for the first time in formalin fixed and paraffin embedded (FFPE) samples of chemotherapy naïve muscle-invasive bladder cancer (MIBC) patients. Expression analyses were performed on the Mannheim cohort (n=84) and validated on the independent Chungbuk cohort (n=61). In a Cox’ proportional hazards model, a distinct FOXM1 expression cut-off dividing both cohorts in a ‘high-risk’ and ‘low-risk’ group has been determined. Multivariate analyses showed that FOXM1 is an independent risk factor for outcome prediction superior to the TNM system. The FOXM1 ‘high-risk’ group had a 4- to 7-fold increased risk of death (p<0.03) and presented further an overexpression of MKI67. Recent studies showed that MIBCs can be subclassified in breast cancer-like subtypes: basal, luminal and p53-like. Here we demonstrated that FOXM1 was differentially expressed between MIBC subtypes concordant to its subtype specific expression in breast cancer. Since the proto-oncogene FOXM1 is known to play an important role in cisplatin resistance and to be a promising drug target, this study supports FOXM1 as a crucial biomarker in the personalization of MIBC therapy and urges prospective translational studies.
Collapse
Affiliation(s)
- Sebastien Rinaldetti
- Department of Hematology and Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | | | - Thomas Stefan Worst
- Department of Urology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Markus Eckstein
- Institute of Pathology, University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Cleo Aaron Weiss
- Institute of Pathology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Johannes Breyer
- Department of Urology, University of Regensburg, 93053 Regensburg, Germany
| | - Wolfgang Otto
- Department of Urology, University of Regensburg, 93053 Regensburg, Germany
| | | | - Arndt Hartmann
- Institute of Pathology, University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Philipp Erben
- Department of Urology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
42
|
Nicolau-Neto P, Da Costa NM, de Souza Santos PT, Gonzaga IM, Ferreira MA, Guaraldi S, Moreira MA, Seuánez HN, Brewer L, Bergmann A, Boroni M, Mencalha AL, Kruel CDP, Lima SCS, Esposito D, Simão TA, Pinto LFR. Esophageal squamous cell carcinoma transcriptome reveals the effect of FOXM1 on patient outcome through novel PIK3R3 mediated activation of PI3K signaling pathway. Oncotarget 2018; 9:16634-16647. [PMID: 29682174 PMCID: PMC5908275 DOI: 10.18632/oncotarget.24621] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/22/2018] [Indexed: 12/31/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) presents poor prognosis, and patients diagnosed with this tumor currently lack target treatments. Therefore, in order to identify potential targets for ESCC treatment, we carried out a transcriptome analysis with ESCC and paired nonmalignant surrounding mucosa samples, followed by a master regulator analysis, and further explored the role of the identified central regulatory genes through in vivo and in vitro assays. Among the transcription factors deregulated/enriched in ESCC, we focused on FOXM1 because of its involvement in the regulation of critical biological processes. A new transcriptome analysis performed with ESCC cell lineage TE-1 showed that the modulation of FOXM1 expression resulted in PIK3R3 expression changes, whereas chromatin immunoprecipitation assay revealed that FOXM1 was capable of binding onto PIK3R3 promoter, thus demonstrating that PIK3R3 is a new FOXM1 target. Furthermore, FOXM1 overexpression resulted in the activation of PIK3/AKT signaling pathway through PIK3R3-mediated AKT phosphorylation. Finally, the analysis of the clinic-pathological data of ESCC patients revealed that overexpression of both FOXM1 and PIK3R3 was associated with poor prognosis, but only the latter was an independent prognosis factor for ESCC patients. In conclusion, our results show that FOXM1 seems to play a central role in ESCC carcinogenesis by upregulating many oncogenes found overexpressed in this tumor. Furthermore, PIK3R3 is a novel FOXM1 target that triggers the activation of the PI3K/AKT pathway in ESCC cells.
Collapse
Affiliation(s)
- Pedro Nicolau-Neto
- Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil
| | - Nathalia Meireles Da Costa
- Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil
| | | | - Isabela Martins Gonzaga
- Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil
| | - Maria Aparecida Ferreira
- Endoscopy Section, Instituto Nacional de Câncer (INCA), Praça Cruz Vermelha, 20230-130 RJ, Brasil
| | - Simone Guaraldi
- Endoscopy Section, Instituto Nacional de Câncer (INCA), Praça Cruz Vermelha, 20230-130 RJ, Brasil
| | - Miguel Angelo Moreira
- Genetic Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil
| | - Hector N Seuánez
- Genetic Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil
| | - Lilian Brewer
- Biochemistry Department, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, 20551-030 RJ, Brasil
| | - Anke Bergmann
- Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil
| | - Mariana Boroni
- Genetic Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil
| | - Andre Luiz Mencalha
- Biophysics and Biometry Department, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, 20551-030 RJ, Brasil
| | - Cleber Dario Pinto Kruel
- Surgery Department, Faculty of Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90035-003 RS, Brasil
| | - Sheila Coelho Soares Lima
- Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil
| | - Dominic Esposito
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, 21701 MD, USA
| | - Tatiana Almeida Simão
- Biochemistry Department, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, 20551-030 RJ, Brasil
| | - Luis Felipe Ribeiro Pinto
- Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, 20231-050 RJ, Brasil.,Biochemistry Department, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, 20551-030 RJ, Brasil
| |
Collapse
|
43
|
Li L, Wu D, Yu Q, Li L, Wu P. Prognostic value of FOXM1 in solid tumors: a systematic review and meta-analysis. Oncotarget 2018; 8:32298-32308. [PMID: 28427178 PMCID: PMC5458285 DOI: 10.18632/oncotarget.15764] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
Accumulated studies have provided controversial evidences of the association between Forkhead Box M1 (FOXM1) expression and survival of human solid tumors. To address this inconsistency, we performed a meta-analysis with 23 studies identified from PubMed and Medline. We found elevated FOXM1-protein expression was significantly associated with worse 3-year overall survival (OS) (OR = 3.30, 95% CI = 2.56 to 4.25, P < 0.00001) 5-year OS (OR =3.35, 95% CI = 2.64 to 4.26, P < 0.00001) and 10-year OS (OR = 5.24, 95% CI = 2.61 to 10.52, P < 0.00001) of human solid tumors. Similar results were observed when disease free survival (DFS) were analyzed. Subgroup analysis showed that FOXM1 overexpression was associated with poor prognosis of colorectal cancer, gastric cancer, hepatic cancer, lung cancer and ovarian cancer. High expression level of FOXM1 was also associated with advanced tumor stage. In conclusion, elevated FOXM1 expression is associated with poor survival in most solid tumors. FOXM1 is a potential biomarker for prognosis prediction and a promising therapeutic target in human solid tumors.
Collapse
Affiliation(s)
- Lijun Li
- Department of Surgery, Hangzhou Xixi Hospital, Hangzhou, China
| | - Dang Wu
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Qun Yu
- Fourth Ward of Neurosurgery, Division of Nursing, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Lingdi Li
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, China
| | - Pin Wu
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Zhong X, Liu X, Li Y, Cheng M, Wang W, Tian K, Mu L, Zeng T, Liu Y, Jiang X, Yu L, Gao L, Zhou Y. HMGA2 sustains self-renewal and invasiveness of glioma-initiating cells. Oncotarget 2018; 7:44365-44380. [PMID: 27259253 PMCID: PMC5190103 DOI: 10.18632/oncotarget.9744] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/20/2016] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of brain tumors with dismal outcomes. The mesenchymal phenotype is the hallmark of tumor aggressiveness in GBMs. Perivascular smooth muscle cells (pericytes) are essential in homeostasis of normal and glioma tissues. Here we found HMGA2, an architectural transcription factor that promotes mesenchymal phenotypes in a number of solid tumors, is highly expressed in mesenchymal subtype of GBMs and labels both glioma pericytes and glioma-initiating cells (GICs). Accordingly, depletion of HMGA2 in GICs resulted in compromised self-renewal and tumorigenic capability, as well as undermined mesenchymal or pericyte differentiation. We further showed HMGA2 allows expressions of FOXM1 and PLAU to maintain GIC propagation, gliomagenesis and aggressiveness both in vitro and in vivo. Therefore, suppressing HMGA2-mediated GIC self-renewal and invasiveness might be a promising means to treat GBMs.
Collapse
Affiliation(s)
- Xiaoling Zhong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| | - Xuan Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| | - Yamu Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| | - Man Cheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| | - Wen Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| | - Kuan Tian
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| | - Lili Mu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| | - Tao Zeng
- Department of Neurosurgery, The Tenth Affiliated Hospital, Tongji University, Shanghai 200072, China
| | - Ying Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Luyang Yu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Liang Gao
- Department of Neurosurgery, The Tenth Affiliated Hospital, Tongji University, Shanghai 200072, China
| | - Yan Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences at Wuhan University, Wuhan 430072, China
| |
Collapse
|
45
|
Han S, Ma X, Zhao Y, Zhao H, Batista A, Zhou S, Zhou X, Yang Y, Wang T, Bi J, Xia Z, Bai Z, Garkavtsev I, Zhang Z. Identification of Glypican-3 as a potential metastasis suppressor gene in gastric cancer. Oncotarget 2018; 7:44406-44416. [PMID: 27259271 PMCID: PMC5190106 DOI: 10.18632/oncotarget.9763] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 05/23/2016] [Indexed: 01/07/2023] Open
Abstract
Gastric cancer is a prevalent tumor that is usually detected at an advanced metastatic stage. Currently, standard therapies are mostly ineffective. Here, we report that Glypican-3 (GPC3) is absent in invasive tumors and metastatic lymph nodes, in particular in aggressive and highly disseminated signet ring cell carcinomas. We demonstrate that loss of GPC3 correlates with poor overall survival in patients. Moreover, we show that absence of GPC3 causes up-regulation of MAPK/FoxM1 signaling and that blockade of this pathway alters cellular invasion. An inverse correlation between GPC3 and FoxM1 is also shown in patient samples. These data identify GPC3 as a potential metastasis suppressor gene and suggest its value as a prognostic marker in gastric cancer. Development of therapies targeting signaling downstream of GPC3 are warranted.
Collapse
Affiliation(s)
- Shiwei Han
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xuemei Ma
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yanxia Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongying Zhao
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ana Batista
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Sheng Zhou
- Institute of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaona Zhou
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yao Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Tingting Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jingtao Bi
- Department of General Surgery, Beijing Jishuitan Hospital, The Fourth Medical College of Peking University, Beijing, China
| | - Zheng Xia
- Department of Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Igor Garkavtsev
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
46
|
Tabatabaei-Dakhili SA, Aguayo-Ortiz R, Domínguez L, Velázquez-Martínez CA. Untying the knot of transcription factor druggability: Molecular modeling study of FOXM1 inhibitors. J Mol Graph Model 2018; 80:197-210. [PMID: 29414039 DOI: 10.1016/j.jmgm.2018.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/15/2017] [Accepted: 01/15/2018] [Indexed: 12/27/2022]
Abstract
The FOXM1 protein is a relevant transcription factor involved in cancer cell proliferation. The direct or indirect inhibition of this protein's transcriptional activity by small molecule drugs correlates well with a potentially significant anti-cancer profile, making this macro molecule a promising drug target. There are a few drug molecules reported to interact with (and inhibit) the FOXM1 DNA binding domain (FOXM1-BD), causing downregulation of protein expression and cancer cell proliferation inhibition. Among these drug molecules are the proteasome inhibitor thiostrepton, the former antidiabetic drug troglitazone, and the new FDI-6 molecule. Despite their structural differences, these drugs exert a similar inhibitory profile, and this observation prompted us to study a possible similar mechanism of action. Using a series of molecular dynamics simulations and docking protocols, we identified essential binding interactions exerted by all three classes of drugs, among which, a π-sulfur interaction (between a His287 and a sulfur-containing heterocycle) was the most important. In this report, we describe the preliminary evidence suggesting the presence of a drug-binding pocket within FOXM1 DNA binding domain, in which inhibitors fit to dissociate the protein-DNA complex. This finding suggests a common mechanism of action and a basic framework to design new FOXM1 inhibitors.
Collapse
Affiliation(s)
| | - Rodrigo Aguayo-Ortiz
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, CDMX 04510, Mexico
| | - Laura Domínguez
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, CDMX 04510, Mexico
| | | |
Collapse
|
47
|
Ma J, Qi G, Xu J, Ni H, Xu W, Ru G, Zhao Z, Xu W, He X. Overexpression of forkhead box M1 and urokinase-type plasminogen activator in gastric cancer is associated with cancer progression and poor prognosis. Oncol Lett 2017; 14:7288-7296. [PMID: 29344165 PMCID: PMC5754915 DOI: 10.3892/ol.2017.7136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 07/03/2017] [Indexed: 01/26/2023] Open
Abstract
Forkhead box M1 (FOXM1) and urokinase-type plasminogen activator (uPA) are overexpressed and associated with the pathogenesis of multiple types of human malignancy. The aims of the present study were to investigate FOXM1 and uPA expression levels in human gastric cancer using tissue microarray techniques; determining their association with clinicopathological characteristics as well as their prognostic value. Tissue microarray blocks, comprising 436 gastric cancer cases and 92 non-cancerous adjacent normal gastric tissues, were analyzed for FOXM1 and uPA protein expression levels using immunohistochemistry. The results were analyzed statistically in association with various clinicopathological characteristics and overall survival rates. FOXM1 and uPA were detected in 78.67 (343/436) and 83.26% (363/436) of cancer samples, respectively. FOXM1 and uPA were not expressed in the 92 normal gastric tissue samples. In gastric cancer, FOXM1 and uPA levels were associated with tumor size, depth of invasion, tumor-node-metastasis (TNM) stage, lymph node metastasis, vessel invasion and distant metastases. The overall survival rate was significantly decreased in patients expressing FOXM1 and uPA compared with FOXM1- and uPA-negative patients. Coxs multivariate analysis revealed that age, depth of invasion and expression levels of FOXM1 and uPA are independent predictors of survival in patients with gastric cancer. These results indicated that increased FOXM1 and uPA expression levels are associated with the invasive and metastatic processes in human gastric cancer, and inversely associated with patient prognosis. Therefore, FOXM1 and uPA may serve as novel prognostic markers independent of, but supplementing, the TNM staging system.
Collapse
Affiliation(s)
- Jie Ma
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Guangwei Qi
- Department of Pathology, Hangzhou Children's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Ji Xu
- Department of Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Haibing Ni
- Department of Surgery, Tongde Hospital of Zhejiang, Hangzhou, Zhejiang 310012, P.R. China
| | - Wulin Xu
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Guoqing Ru
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Zhongsheng Zhao
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Wenjuan Xu
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xujun He
- Key Laboratory of Gastroenterology of Zhejiang, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
48
|
Zingales V, Distefano A, Raffaele M, Zanghi A, Barbagallo I, Vanella L. Metformin: A Bridge between Diabetes and Prostate Cancer. Front Oncol 2017; 7:243. [PMID: 29075616 PMCID: PMC5641539 DOI: 10.3389/fonc.2017.00243] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) has become the most frequent type of cancer in men. Recent data suggest that diabetic patients taking metformin have a lower incidence of certain cancer, including PCa. Metformin is the most common drug used in type II diabetes mellitus; its use has been shown to lower the incidence of several cancers, although there are ambiguous data about the anticancer activity of metformin. A large number of studies examined the potential antineoplastic mechanism of metformin although it is not still completely understood. This review summarizes the literature concerning the effects of metformin on PCa cells, highlighting its numerous mechanisms of action through which it can act. We analyze the possible causes of the discordances regarding the impact of metformin on risk of PCa; we discuss the latest findings in this field, suggesting that metformin may have a future role in the management of PCa both as monotherapy and in combination with other drugs.
Collapse
Affiliation(s)
- Veronica Zingales
- Department of Drug Science, Biochemistry Section, University of Catania, Catania, Italy
| | - Alfio Distefano
- Department of Drug Science, Biochemistry Section, University of Catania, Catania, Italy
| | - Marco Raffaele
- Department of Drug Science, Biochemistry Section, University of Catania, Catania, Italy
| | - Antonio Zanghi
- Department of Surgery, Azienda Ospedaliero Universitaria Policlinico Vittorio Emanuele, Catania, Italy
| | - Ignazio Barbagallo
- Department of Drug Science, Biochemistry Section, University of Catania, Catania, Italy
| | - Luca Vanella
- Department of Drug Science, Biochemistry Section, University of Catania, Catania, Italy
| |
Collapse
|
49
|
Hasanpourghadi M, Pandurangan AK, Mustafa MR. Modulation of oncogenic transcription factors by bioactive natural products in breast cancer. Pharmacol Res 2017; 128:376-388. [PMID: 28923544 DOI: 10.1016/j.phrs.2017.09.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/11/2017] [Accepted: 09/14/2017] [Indexed: 12/17/2022]
Abstract
Carcinogenesis, a multi-step phenomenon, characterized by alterations at genetic level and affecting the main intracellular pathways controlling cell growth and development. There are growing number of evidences linking oncogenes to the induction of malignancies, especially breast cancer. Modulations of oncogenes lead to gain-of-function signals in the cells and contribute to the tumorigenic phenotype. These signals yield a large number of proteins that cause cell growth and inhibit apoptosis. Transcription factors such as STAT, p53, NF-κB, c-JUN and FOXM1, are proteins that are conserved among species, accumulate in the nucleus, bind to DNA and regulate the specific genes targets. Oncogenic transcription factors resulting from the mutation or overexpression following aberrant gene expression relay the signals in the nucleus and disrupt the transcription pattern. Activation of oncogenic transcription factors is associated with control of cell cycle, apoptosis, migration and cell differentiation. Among different cancer types, breast cancer is one of top ten cancers worldwide. There are different subtypes of breast cancer cell-lines such as non-aggressive MCF-7 and aggressive and metastatic MDA-MB-231 cells, which are identified with distinct molecular profile and different levels of oncogenic transcription factor. For instance, MDA-MB-231 carries mutated and overexpressed p53 with its abnormal, uncontrolled downstream signalling pathway that account for resistance to several anticancer drugs compared to MCF-7 cells with wild-type p53. Appropriate enough, inhibition of oncogenic transcription factors has become a potential target in discovery and development of anti-tumour drugs against breast cancer. Plants produce diverse amount of organic metabolites. Universally, these metabolites with biological activities are known as "natural products". The chemical structure and function of natural products have been studied since 1850s. Investigating these properties leaded to recognition of their molecular effects as anticancer drugs. Numerous natural products extracted from plants, fruits, mushrooms and mycelia, show potential inhibitory effects against several oncogenic transcription factors in breast cancer. Natural compounds that target oncogenic transcription factors have increased the number of candidate therapeutic agents. This review summarizes the current findings of natural products in targeting specific oncogenic transcription factors in breast cancer.
Collapse
Affiliation(s)
- Mohadeseh Hasanpourghadi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ashok Kumar Pandurangan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia; Centre for Natural Products Research and Drug Discovery, Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
50
|
Nandi D, Cheema PS, Jaiswal N, Nag A. FoxM1: Repurposing an oncogene as a biomarker. Semin Cancer Biol 2017; 52:74-84. [PMID: 28855104 DOI: 10.1016/j.semcancer.2017.08.009] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 08/08/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022]
Abstract
The past few decades have witnessed a tremendous progress in understanding the biology of cancer, which has led to more comprehensive approaches for global gene expression profiling and genome-wide analysis. This has helped to determine more sophisticated prognostic and predictive signature markers for the prompt diagnosis and precise screening of cancer patients. In the search for novel biomarkers, there has been increased interest in FoxM1, an extensively studied transcription factor that encompasses most of the hallmarks of malignancy. Considering the attractive potential of this multifarious oncogene, FoxM1 has emerged as an important molecule implicated in initiation, development and progression of cancer. Bolstered with the skill to maneuver the proliferation signals, FoxM1 bestows resistance to contemporary anti-cancer therapy as well. This review sheds light on the large body of literature that has accumulated in recent years that implies that FoxM1 neoplastic functions can be used as a novel predictive, prognostic and therapeutic marker for different cancers. This assessment also highlights the key features of FoxM1 that can be effectively harnessed to establish FoxM1 as a strong biomarker in diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Deeptashree Nandi
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Pradeep Singh Cheema
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Neha Jaiswal
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|