1
|
YANG PIAO, SHEYKHHASAN MOHSEN, HEIDARI REZA, CHAMANARA MOHSEN, DAMA PAOLA, AHMADIEH-YAZDI AMIRHOSSEIN, MANOOCHEHRI HAMED, TANZADEHPANAH HAMID, MAHAKI HANIE, KALHOR NASER, DIRBAZIYAN ASHKAN, AL-MUSAWI SHARAFALDIN. FOXR2 in cancer development: emerging player and therapeutic opportunities. Oncol Res 2025; 33:283-300. [PMID: 39866234 PMCID: PMC11753994 DOI: 10.32604/or.2024.052939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/26/2024] [Indexed: 01/28/2025] Open
Abstract
Cancer, a leading cause of global mortality, remains a significant challenge to increasing life expectancy worldwide. Forkhead Box R2 (FOXR2), identified as an oncogene within the FOX gene family, plays a crucial role in developing various endoderm-derived organs. Recent studies have elucidated FOXR2-related pathways and their involvement in both tumor and non-tumor diseases. Dysregulation of FOXR2 has been linked to numerous malignant tumors, spanning the brain, nervous system, thyroid, osteosarcoma, Hodgkin lymphoma, colorectal, liver, pancreatic, lung, breast, ovarian, prostate, female genital tract, endometrial, and uterine cancers. Despite extensive research on FOXR2 dysregulation, its practical applications remain underexplored. This review delves into the mechanisms underlying FOXR2 dysregulation during oncogenesis and its implications for cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- PIAO YANG
- Department of Molecular Genetics, College of Arts and Sciences, The Ohio State University, Columbus, OH, USA
| | - MOHSEN SHEYKHHASAN
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - REZA HEIDARI
- Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - MOHSEN CHAMANARA
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Student Research Committee, AJA University of Medical Sciences, Tehran, Iran
| | - PAOLA DAMA
- Research Fellow School of Life Sciences, University of Sussex, Brighton, UK
| | - AMIRHOSSEIN AHMADIEH-YAZDI
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - HAMED MANOOCHEHRI
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - HAMID TANZADEHPANAH
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - HANIE MAHAKI
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - NASER KALHOR
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | - ASHKAN DIRBAZIYAN
- Department of Microbiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
2
|
Wu X, Wu Y, Tang F, Wang Y, Li C, Wu S, Wang G, Zhang J. Foxq1 activates CB2R with oleamide to alleviate POCD. Brain Pathol 2025; 35:e13289. [PMID: 39046224 PMCID: PMC11669408 DOI: 10.1111/bpa.13289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a major concern, particularly among older adults. This study used social isolation (ISO) and multiomics analyses in aged mice to investigate potential mechanisms underlying POCD development. Aged mice were divided into two groups: ISO and paired housing (PH). Oleamide and the cannabinoid receptor type 2 (CB2R) antagonist AM630 were administered intraperitoneally, while Foxq1 adeno-associated viral (AAV) vector was injected directly into the hippocampus. Intramedullary tibial surgeries were subsequently performed to establish the POCD models. Behavioral tests comprising the Y-maze, open field test, and novel object recognition were conducted 2 days after surgery. Hippocampal and serum inflammatory cytokines were assessed. Following surgery, ISO mice demonstrated intensified cognitive impairments and escalated inflammatory markers. Integrative transcriptomic and metabolomic analysis revealed elevated oleamide concentrations in the hippocampus and serum of PH mice, with associative investigations indicating a close relationship between the Foxq1 gene and oleamide levels. While oleamide administration and Foxq1 gene overexpression substantially ameliorated postoperative cognitive performance and systemic inflammation in mice, CB2R antagonist AM630 impeded these enhancements. The Foxq1 gene and oleamide may be crucial in alleviating POCD. While potentially acting through CB2R-mediated pathways, these factors may modulate neuroinflammation and attenuate proinflammatory cytokine levels within the hippocampus, substantially improving cognitive performance postsurgery. This study lays the groundwork for future research into therapeutic approaches targeting the Foxq1-oleamide-CB2R axis, with the ultimate goal of preventing or mitigating POCD.
Collapse
Affiliation(s)
- Xiaoying Wu
- Department of Anesthesiology and Perioperative MedicinePeople's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouHenanChina
| | - Yuming Wu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Fudong Tang
- Department of Anesthesiology and Perioperative MedicinePeople's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouHenanChina
| | - Yangyang Wang
- Department of Anesthesiology and Perioperative MedicinePeople's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouHenanChina
| | - Chenxi Li
- Department of Anesthesiology and Perioperative MedicinePeople's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouHenanChina
| | - Su Wu
- Department of Anesthesiology and Perioperative MedicinePeople's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouHenanChina
| | - Guangzhi Wang
- Department of Anesthesiology and Perioperative MedicinePeople's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouHenanChina
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative MedicinePeople's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouHenanChina
| |
Collapse
|
3
|
Lotfi M, Maharati A, Hamidi AA, Taghehchian N, Moghbeli M. MicroRNA-532 as a probable diagnostic and therapeutic marker in cancer patients. Mutat Res 2024; 829:111874. [PMID: 38986233 DOI: 10.1016/j.mrfmmm.2024.111874] [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: 08/18/2023] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
The high mortality rate in cancer patients is always one of the main challenges of the health systems globally. Several factors are involved in the high rate of cancer related mortality, including late diagnosis and drug resistance. Cancer is mainly diagnosed in the advanced stages of tumor progression that causes the failure of therapeutic strategies and increases the death rate in these patients. Therefore, assessment of the molecular mechanisms associated with the occurrence of cancer can be effective to introduce early tumor diagnostic markers. MicroRNAs (miRNAs) as the stable non-coding RNAs in the biological body fluids are involved in regulation of cell proliferation, migration, and apoptosis. MiR-532 deregulation has been reported in different tumor types. Therefore, in the present review we discussed the role of miR-532 during tumor growth. It has been shown that miR-532 has mainly a tumor suppressor role through the regulation of transcription factors, chemokines, and signaling pathways such as NF-kB, MAPK, PI3K/AKT, and WNT. In addition to the independent role of miR-532 in regulation of cellular processes, it also functions as a mediator of lncRNAs and circRNAs. Therefore, miR-532 can be considered as a non-invasive diagnostic/prognostic marker as well as a therapeutic target in cancer patients.
Collapse
Affiliation(s)
- Malihe Lotfi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Hamidi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Gharbaran R. Insights into the molecular roles of FOXR2 in the pathology of primary pediatric brain tumors. Crit Rev Oncol Hematol 2023; 192:104188. [PMID: 37879492 DOI: 10.1016/j.critrevonc.2023.104188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/23/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023] Open
Abstract
Forkhead box gene R2 (FOXR2) belongs to the family of FOX genes which codes for highly conserved transcription factors (TFs) with critical roles in biological processes ranging from development to organogenesis to metabolic and immune regulation to cellular homeostasis. A number of FOX genes are associated with cancer development and progression and poor prognosis. A growing body of evidence suggests that FOXR2 is an oncogene. Studies suggested important roles for FOXR2 in cancer cell growth, metastasis, and drug resistance. Recent studies showed that FOXR2 is overexpressed by a subset of newly identified entities of embryonal tumors. This review discusses the role(s) FOXR2 plays in the pathology of pediatric brain cancers and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Rajendra Gharbaran
- Biological Sciences Department, Bronx Community College/City University of New York, 2155 University Avenue, Bronx, NY 10453, USA.
| |
Collapse
|
5
|
Song J, Li L, Fang Y, Lin Y, Wu L, Wan W, Wei G, Hua F, Ying J. FOXN Transcription Factors: Regulation and Significant Role in Cancer. Mol Cancer Ther 2023; 22:1028-1039. [PMID: 37566097 DOI: 10.1158/1535-7163.mct-23-0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/29/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023]
Abstract
A growing number of studies have demonstrated that cancer development is closely linked to abnormal gene expression, including alterations in the transcriptional activity of transcription factors. The Forkhead box class N (FOXN) proteins FOXN1-6 form a highly conserved class of transcription factors, which have been shown in recent years to be involved in the regulation of malignant progression in a variety of cancers. FOXNs mediate cell proliferation, cell-cycle progression, cell differentiation, metabolic homeostasis, embryonic development, DNA damage repair, tumor angiogenesis, and other critical biological processes. Therefore, transcriptional dysregulation of FOXNs can directly affect cellular physiology and promote cancer development. Numerous studies have demonstrated that the transcriptional activity of FOXNs is regulated by protein-protein interactions, microRNAs (miRNA), and posttranslational modifications (PTM). However, the mechanisms underlying the molecular regulation of FOXNs in cancer development are unclear. Here, we reviewed the molecular regulatory mechanisms of FOXNs expression and activity, their role in the malignant progression of tumors, and their value for clinical applications in cancer therapy. This review may help design experimental studies involving FOXN transcription factors, and enhance their therapeutic potential as antitumor targets.
Collapse
Affiliation(s)
- Jiali Song
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Longshan Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yang Fang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Luojia Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Wei Wan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Gen Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| |
Collapse
|
6
|
Liang YX, Zhang LL, Yang L. circANKRD17(has_circ_0007883) confers paclitaxel resistance of ovarian cancer via interacting with FUS to stabilize FOXR2. Mol Cell Biochem 2022; 478:835-850. [PMID: 36107285 DOI: 10.1007/s11010-022-04548-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 08/26/2022] [Indexed: 12/21/2022]
Abstract
Emerging numbers of endogenous circular RNAs (circRNAs) have gained much attention to serve as essential regulators in the carcinogenesis of human cancers. Unfortunately, the occurrence of paclitaxel (PTX) resistance to ovarian cancer remains to be responsible for the poor prognosis. Herein, the aim of our study is to reveal a dysregulation of a particular circRNA, circANKRD17 (has_circ_0007883), and its exact role involving in chemoresistance of ovarian cancer. Expression patterns of circANKRD17 in PTX-resistant ovarian cancer tissues and cell lines was examined using quantitative real-time PCR analysis. Role of circANKRD17 on drug resistance and cell viability was evaluated by CCK-8 assay. Colony formation was subjected to measure cell proliferation. Flow cytometry was employed to evaluate cell cycle either or cell apoptosis. Xenograft models were constructed for further in vivo confirmation. The cicrANKRD17/FUS/FOXR2 axis was demonstrated using bioinformatics analysis, RNA pull-down, as well as RNA immunoprecipitation assays. Dramatically high expressed circANKRD17 observed in ovarian cancer tissues and cells was correlated with PTX resistance, which indicated the poor prognosis. Functionally, knockdown of circANKRD17 decreased PTX resistance via inhibiting cell viability and inducing cell apoptosis. Mechanistically, circANKRD17 interacted with the RNA-binding protein, fused in sarcoma (FUS) to stabilize FOXR2. In summary, our study uncovered a novel machinery of circANKRD17/FUS/FOXR2 referring to ovarian cancer drug sensitivity and tumorigenesis, highlighting a potential strategy for circRNAs in chemoresistance.
Collapse
Affiliation(s)
- Yue-Xiu Liang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, Guangxi, People's Republic of China
| | - Lian-Ling Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, Guangxi, People's Republic of China
| | - Li Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, Guangxi, People's Republic of China.
| |
Collapse
|
7
|
Mahabady MK, Mirzaei S, Saebfar H, Gholami MH, Zabolian A, Hushmandi K, Hashemi F, Tajik F, Hashemi M, Kumar AP, Aref AR, Zarrabi A, Khan H, Hamblin MR, Nuri Ertas Y, Samarghandian S. Noncoding RNAs and their therapeutics in paclitaxel chemotherapy: Mechanisms of initiation, progression, and drug sensitivity. J Cell Physiol 2022; 237:2309-2344. [PMID: 35437787 DOI: 10.1002/jcp.30751] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
Abstract
The identification of agents that can reverse drug resistance in cancer chemotherapy, and enhance the overall efficacy is of great interest. Paclitaxel (PTX) belongs to taxane family that exerts an antitumor effect by stabilizing microtubules and inhibiting cell cycle progression. However, PTX resistance often develops in tumors due to the overexpression of drug transporters and tumor-promoting pathways. Noncoding RNAs (ncRNAs) are modulators of many processes in cancer cells, such as apoptosis, migration, differentiation, and angiogenesis. In the present study, we summarize the effects of ncRNAs on PTX chemotherapy. MicroRNAs (miRNAs) can have opposite effects on PTX resistance (stimulation or inhibition) via influencing YES1, SK2, MRP1, and STAT3. Moreover, miRNAs modulate the growth and migration rates of tumor cells in regulating PTX efficacy. PIWI-interacting RNAs, small interfering RNAs, and short-hairpin RNAs are other members of ncRNAs regulating PTX sensitivity of cancer cells. Long noncoding RNAs (LncRNAs) are similar to miRNAs and can modulate PTX resistance/sensitivity by their influence on miRNAs and drug efflux transport. The cytotoxicity of PTX against tumor cells can also be affected by circular RNAs (circRNAs) and limitation is that oncogenic circRNAs have been emphasized and experiments should also focus on onco-suppressor circRNAs.
Collapse
Affiliation(s)
- Mahmood K Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Hamidreza Saebfar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad H Gholami
- Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Amirhossein Zabolian
- Resident of Orthopedics, Department of Orthopedics, School of Medicine, 5th Azar Hospital, Golestan University of Medical Sciences, Golestan, Iran
| | - Kiavash Hushmandi
- Division of Epidemiology, Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alan P Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amir R Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.,Xsphera Biosciences Inc, Boston, Massachusetts, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul, Turkey
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey.,ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
8
|
Xia D, Chen Z, Liu Q. Circ-PGC increases the expression of FOXR2 by targeting miR-532-3p to promote the development of non-small cell lung cancer. Cell Cycle 2021; 20:2195-2209. [PMID: 34494941 DOI: 10.1080/15384101.2021.1974788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This study was to explore the function of circular progastricsin (circ-PGC) in NSCLC. The histological morphology of tumor tissues was observed by hematoxylin and eosin (HE) staining. The expression of circ-PGC, miR-532-3p and forkhead box R2 (FOXR2) mRNA was measured by real-time quantitative polymerase chain reaction (RT-qPCR). The protein level of FOXR2 was checked by western blot. In functional analyses, cell viability, colony formation, cell apoptosis, cell migration and cell invasion were investigated using cell counting kit-8 (CCK-8) assay, colony formation assay, flow cytometry assay, wound healing assay and transwell assay. Besides, glycolysis metabolism was assessed by the levels of glucose consumption, lactate production and adenosine triphosphate (ATP) production. The predicted relationship between miR-532-3p and circ-PGC and FOXR2 was verified by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. The results showed that circ-PGC and FOXR2 were upregulated in NSCLC tissues and cells. Circ-PGC knockdown or FOXR2 knockdown inhibited NSCLC cell viability, colony formation, cell migration, invasion and glycolysis metabolism, and FOXR2 overexpression rescued these inhibitory effects caused by circ-PGC knockdown. MiR-532-3p harbored the same binding site with circ-PGC and FOXR2, and circ-PGC positively regulated FOXR2 expression by targeting miR-532-3p. The expression of β-catenin and c-Myc was decreased in cells after circ-PGC knockdown but recovered with miR-532-3p inhibition or FOXR2 overexpression. Circ-PGC downregulation also inhibited tumor growth in vivo. In conclusion, circ-PGC positively regulated FOXR2 expression by competitively binding to miR-532-3p, thereby promoting the development of NSCLC, and the Wnt/β-catenin signaling pathway might be activated by the circ-PGC/miR-532-3p/FOXR2 network.
Collapse
Affiliation(s)
- Daokui Xia
- Department of Cardiothoracic Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Zhen Chen
- Department of Cardiothoracic Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Quan Liu
- Department of Cardiothoracic Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
9
|
Schmitt-Hoffner F, van Rijn S, Toprak UH, Mauermann M, Rosemann F, Heit-Mondrzyk A, Hübner JM, Camgöz A, Hartlieb S, Pfister SM, Henrich KO, Westermann F, Kool M. FOXR2 Stabilizes MYCN Protein and Identifies Non- MYCN-Amplified Neuroblastoma Patients With Unfavorable Outcome. J Clin Oncol 2021; 39:3217-3228. [PMID: 34110923 PMCID: PMC8500564 DOI: 10.1200/jco.20.02540] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clinical outcomes of patients with neuroblastoma range from spontaneous tumor regression to fatality. Hence, understanding the mechanisms that cause tumor progression is crucial for the treatment of patients. In this study, we show that FOXR2 activation identifies a subset of neuroblastoma tumors with unfavorable outcome and we investigate the mechanism how FOXR2 relates to poor outcome in patients.
Collapse
Affiliation(s)
- Felix Schmitt-Hoffner
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Sjoerd van Rijn
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Umut H Toprak
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Monika Mauermann
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Felix Rosemann
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Anke Heit-Mondrzyk
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jens-Martin Hübner
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Aylin Camgöz
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Sabine Hartlieb
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kai-Oliver Henrich
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Westermann
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcel Kool
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
10
|
E3 ubiquitin ligase PJA1 regulates lung adenocarcinoma apoptosis and invasion through promoting FOXR2 degradation. Biochem Biophys Res Commun 2021; 556:106-113. [PMID: 33839405 DOI: 10.1016/j.bbrc.2021.03.137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/18/2023]
Abstract
Among all lung cancer cases, lung adenocarcinoma (LAC) represents nearly 40% and remains the leading cause of cancer deaths worldwide. Although the combination therapy of surgical treatment with radiotherapy, chemotherapy, and immunotherapy, has been used to treat LAC, unfortunately, high recurrence rates and poor survival remain. Therefore, novel prognostic markers and new targets for molecular targeted therapy in LAC is urgently needed. Fork-head box R2 (FOXR2) plays a key role in a wide range of cellular processes, including cellular proliferation, invasion, differentiation, and apoptosis, and it has been reported to be implicated in progression of LAC, thus inhibition of FOXR2 may be a novel targeting therapy for lung cancer. This current study found that E3 ligase PJA1 regulates ubiquitin-mediated degradation of FOXR2 and predicts good outcome of patients with LAC. In addition, it was showed force expression of PJA1 significantly inhibited LAC cells invasion and induced apoptosis in vitro through inactivating Wnt/β-catenin signaling pathway. In short, our findings reveal that PJA1 could be a potential diagnostic and prognostic biomarkers and the PJA1- FOXR2 axis could be served as a promising target for LAC therapy.
Collapse
|
11
|
Liao CW, Zheng C, Wang L. Down-regulation of FOXR2 inhibits hypoxia-driven ROS-induced migration and invasion of thyroid cancer cells via regulation of the hedgehog pathway. Clin Exp Pharmacol Physiol 2020; 47:1076-1082. [PMID: 32068268 DOI: 10.1111/1440-1681.13286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 02/08/2020] [Accepted: 02/14/2020] [Indexed: 12/15/2022]
Abstract
Forkhead box R2 (FOXR2), a new member of the FOX family, is involved in a wide range of biological processes such as embryogenesis, differentiation, transformation and metabolic homeostasis. Recently, FOXR2 has been reported to be aberrantly expressed in a variety of cancers and correlated with cancer development. However, the specific role of FOXR2 in thyroid cancer (TC) remains unclear. In this study, we showed that FOXR2 was highly expressed in TC tissues and cell lines. Moreover, down-regulation of FOXR2 inhibited hypoxia-induced reactive oxygen species (ROS) production and migration/invasion of TC cells. We also found that the hedgehog pathway was responsible for the partial mechanisms underlying the inhibitory effect. Taken together, these findings indicated that down-regulation of FOXR2 inhibits hypoxia-driven ROS-induced migration and invasion of TC cells via regulation of the hedgehog pathway. Thus, FOXR2 may hold great potential for TC treatment.
Collapse
Affiliation(s)
- Chong-Wu Liao
- First Department of General Surgery, Xi'an Central Hospital, Xi'an, China
| | - Chen Zheng
- First Department of General Surgery, Xi'an Central Hospital, Xi'an, China
| | - Le Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Forkhead box (FOX) G1 promotes hepatocellular carcinoma epithelial-Mesenchymal transition by activating Wnt signal through forming T-cell factor-4/Beta-catenin/FOXG1 complex. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:475. [PMID: 31771611 PMCID: PMC6880489 DOI: 10.1186/s13046-019-1433-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/01/2019] [Indexed: 11/18/2022]
Abstract
Background Forkhead box G1 (FOXG1) is a member of the Fox transcription factor family involved in regulation of many cancers. However, the role of FOXG1 in hepatocellular carcinogenesisis largely unclear. The present study aimed at examining the biological function and underlying mechanism of FOXG1 on hepatocellular carcinoma (HCC) tumor metastasis as well as its clinical significance. Methods Levels of FOXG1 were determined by immunohistochemical and real-time PCR analysis in HCC cell lines and human HCC samples. The effect of FOXG1 on cancer cell invasion and metastasis was investigated in vitro and in vivo in either FOXG1-silenced or overexpressing human HCC cell lines. Immunoprecipitation and chromatin immunoprecipitation assays were performed to investigate the interaction of FOXG1, β-catenin, TCF4 and the effect on Wnt target-gene promoters. Results In human HCC, the level of FOXG1 progressively increased from surrounding non tumorous livers to HCC, reaching the highest levels in metastatic HCC. Furthermore, expression levels of FOXG1 directly correlated with cancer cell epithelial-mesenchymal transition (EMT) phenotype. In FOXG1-overexpressing cells, FOXG1 promotes the stabilization and nuclear accumulation of β-catenin by directly binding to β-catenin and it associates with the lymphoid enhancer factor/T cell factor proteins (LEF/TCFs) on Wnt responsive enhancers (WREs) in chromatin. Conclusions The results show that FOXG1 plays a key role in mediating cancer cell metastasis through the Wnt/β-catenin pathway in HCC cells and predicts HCC prognosis after surgery. Targeting FOXG1 may provide a new approach for therapeutic treatment in the future.
Collapse
|
13
|
Li Y, Zu X, Hu X, Wang L, He W. Forkhead Box R2 Knockdown Decreases Chemoresistance to Cisplatin via MYC Pathway in Bladder Cancer. Med Sci Monit 2019; 25:8928-8939. [PMID: 31761897 PMCID: PMC6894368 DOI: 10.12659/msm.917345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Bladder cancer is a very common urological cancer globally, and cisplatin- or gemcitabine-based chemotherapy is essential for advanced bladder cancer patients. Many patients with bladder cancer have a relatively poor response to chemotherapy, leading to failure of clinical treatment. We mined the GSE77883 GEO dataset, identifying FoxR2 as being a significantly upregulated gene in T24 chemoresistant cells. Herein, we assessed how FoxR2 functions in bladder cancer cell chemoresistance. Material/Methods Cisplatin-resistant T24 (T24/DDP) cells were constructed by administering increasing concentrations of cisplatin, and differences in expression of FoxR2 were examined in T24/DDP and T24 cells. FoxR2 loss- and gain-of-function cells models were established in T24/DDP and T24 cells, respectively. Cell survival, clone formation, cell cycle, and cell apoptosis were assessed, and the MYC pathway was verified. Results FoxR2 was significantly upregulated in T24/DDP cells compared to T24 cells. Knockdown of FoxR2 in T24/DDP cells, survival rate, and clone formation were decreased, G1/S phase transition was suppressed, and cell apoptosis was promoted. These results were reversed by restoration of FoxR2 levels in T24 cells. We found that FoxR2 knockdown enhanced sensitivity to cisplatin, whereas MYC overexpression antagonized chemosensitivity in T24/DDP cells. Conclusions FoxR2 knockdown decreases chemoresistance to cisplatin via the MYC pathway in bladder cancer cells, and this may be a target for overcoming chemoresistance in bladder cancer.
Collapse
Affiliation(s)
- Yangle Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Xiheng Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Long Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Wei He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
14
|
Zhang G, Zhang G. Upregulation of FoxP4 in HCC promotes migration and invasion through regulation of EMT. Oncol Lett 2019; 17:3944-3951. [PMID: 30930991 DOI: 10.3892/ol.2019.10049] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 11/16/2018] [Indexed: 12/21/2022] Open
Abstract
Previous studies have indicated that FoxP1, FoxP2 and FoxP3 play important roles in hepatocellular carcinoma (HCC). However, the effect of FoxP4 in HCC requires further elucidation. The aim of the present study was to explore the roles of FoxP4 in HCC and further decipher the detailed mechanism. In present study, it was found that FoxP4, which is overexpressed in HCC tissues and cell lines, facilitated EMT in HCC cell lines through regulation of Slug. First, increased expression of FoxP4 was identified in 110 pairs of human HCC tumor and their adjacent normal tissues. In addition, the association between FoxP4 expression and clinicopathological features of HCC patients indicated that FoxP4 played vital roles in HCC development. Subsequently, gain- and loss-of-function experiments indicated that FoxP4 promoted cellular proliferation, migration as well invasion. In addition, EMT, a key mechanism during cancer metastasis, was regulated by FoxP4. Furthermore, ChIP and qChIP as well as luciferase reporter assays indicated that Slug, an EMT-associated transcription factor, was transcriptionally regulated by FoxP4. In conclusion, FoxP4 functioned as a tumor promoter in HCC cells by transcriptionally regulating Slug, and the present study highlighted the potential effects of FoxP4 on the prognosis and treatment of HCC.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Liver Diseases, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, Shandong 276800, P.R. China
| | - Guangye Zhang
- Department of Liver Diseases, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, Shandong 276800, P.R. China
| |
Collapse
|
15
|
Beckmann PJ, Larson JD, Larsson AT, Ostergaard JP, Wagner S, Rahrmann EP, Shamsan GA, Otto GM, Williams RL, Wang J, Lee C, Tschida BR, Das P, Dubuc AM, Moriarity BS, Picard D, Wu X, Rodriguez FJ, Rosemarie Q, Krebs RD, Molan AM, Demer AM, Frees MM, Rizzardi AE, Schmechel SC, Eberhart CG, Jenkins RB, Wechsler-Reya RJ, Odde DJ, Huang A, Taylor MD, Sarver AL, Largaespada DA. Sleeping Beauty Insertional Mutagenesis Reveals Important Genetic Drivers of Central Nervous System Embryonal Tumors. Cancer Res 2019; 79:905-917. [PMID: 30674530 DOI: 10.1158/0008-5472.can-18-1261] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 11/07/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022]
Abstract
Medulloblastoma and central nervous system primitive neuroectodermal tumors (CNS-PNET) are aggressive, poorly differentiated brain tumors with limited effective therapies. Using Sleeping Beauty (SB) transposon mutagenesis, we identified novel genetic drivers of medulloblastoma and CNS-PNET. Cross-species gene expression analyses classified SB-driven tumors into distinct medulloblastoma and CNS-PNET subgroups, indicating they resemble human Sonic hedgehog and group 3 and 4 medulloblastoma and CNS neuroblastoma with FOXR2 activation. This represents the first genetically induced mouse model of CNS-PNET and a rare model of group 3 and 4 medulloblastoma. We identified several putative proto-oncogenes including Arhgap36, Megf10, and Foxr2. Genetic manipulation of these genes demonstrated a robust impact on tumorigenesis in vitro and in vivo. We also determined that FOXR2 interacts with N-MYC, increases C-MYC protein stability, and activates FAK/SRC signaling. Altogether, our study identified several promising therapeutic targets in medulloblastoma and CNS-PNET. SIGNIFICANCE: A transposon-induced mouse model identifies several novel genetic drivers and potential therapeutic targets in medulloblastoma and CNS-PNET.
Collapse
Affiliation(s)
- Pauline J Beckmann
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Jon D Larson
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Alex T Larsson
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Jason P Ostergaard
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Sandra Wagner
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Eric P Rahrmann
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota.,Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, England, United Kingdom
| | - Ghaidan A Shamsan
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - George M Otto
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota.,Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, California
| | - Rory L Williams
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota.,Department of Bioengineering, California Institute of Technology, Pasadena, California
| | - Jun Wang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Catherine Lee
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Barbara R Tschida
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Paramita Das
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Adrian M Dubuc
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Branden S Moriarity
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Daniel Picard
- Department of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Xiaochong Wu
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Quincy Rosemarie
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota.,McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ryan D Krebs
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Amy M Molan
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota.,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Addison M Demer
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Michelle M Frees
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Anthony E Rizzardi
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Stephen C Schmechel
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota.,Department of Clinical Sciences, College of Medicine, Florida State University, Sarasota, Florida
| | - Charles G Eberhart
- Department of Pathology, Ophthalmology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic and Foundation, 200 First Street Southwest, Rochester, Minnesota
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Annie Huang
- Division of Hematology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael D Taylor
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Aaron L Sarver
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
16
|
Laissue P. The forkhead-box family of transcription factors: key molecular players in colorectal cancer pathogenesis. Mol Cancer 2019; 18:5. [PMID: 30621735 PMCID: PMC6325735 DOI: 10.1186/s12943-019-0938-x] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/01/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly occurring cancer worldwide and the fourth most frequent cause of death having an oncological origin. It has been found that transcription factors (TF) dysregulation, leading to the significant expression modifications of genes, is a widely distributed phenomenon regarding human malignant neoplasias. These changes are key determinants regarding tumour’s behaviour as they contribute to cell differentiation/proliferation, migration and metastasis, as well as resistance to chemotherapeutic agents. The forkhead box (FOX) transcription factor family consists of an evolutionarily conserved group of transcriptional regulators engaged in numerous functions during development and adult life. Their dysfunction has been associated with human diseases. Several FOX gene subgroup transcriptional disturbances, affecting numerous complex molecular cascades, have been linked to a wide range of cancer types highlighting their potential usefulness as molecular biomarkers. At least 14 FOX subgroups have been related to CRC pathogenesis, thereby underlining their role for diagnosis, prognosis and treatment purposes. This manuscript aims to provide, for the first time, a comprehensive review of FOX genes’ roles during CRC pathogenesis. The molecular and functional characteristics of most relevant FOX molecules (FOXO, FOXM1, FOXP3) have been described within the context of CRC biology, including their usefulness regarding diagnosis and prognosis. Potential CRC therapeutics (including genome-editing approaches) involving FOX regulation have also been included. Taken together, the information provided here should enable a better understanding of FOX genes’ function in CRC pathogenesis for basic science researchers and clinicians.
Collapse
Affiliation(s)
- Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 N° 63C-69, Bogotá, Colombia.
| |
Collapse
|
17
|
Leng X, Ma J, Liu Y, Shen S, Yu H, Zheng J, Liu X, Liu L, Chen J, Zhao L, Ruan X, Xue Y. Mechanism of piR-DQ590027/MIR17HG regulating the permeability of glioma conditioned normal BBB. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:246. [PMID: 30305135 PMCID: PMC6180493 DOI: 10.1186/s13046-018-0886-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
Background The blood-brain barrier (BBB) strongly restricts the entry of anti-glioma drugs into tumor tissues and thus decreases chemotherapy efficacy. Malignant gliomas are highly invasive tumours that use the perivascular space for invasion and co-opt existing vessels as satellite tumor form. Because regulation of the effect of noncoding RNA on BBB function is attracting growing attention, we investigated the effects of noncoding RNA on the permeability of glioma conditioned normal BBB and the mechanism involved using PIWI-associated RNA piR-DQ590027 as a starting point. Methods The mRNA levels of MIR17HG, miR-153, miR-377, ZO-1, occludin, and claudin-5 were determined using real-time PCR. Transient cell transfection was performed using Lipofectamine 3000 reagent. TEER and HRP flux were applied to measure the permeability of glioma conditioned normal BBB. Western blotting and immunofluorescence assays were used to measure ZO-1, occludin, and claudin-5 levels. Reporter vector construction and a luciferase reporter assay were performed to detect the binding sites of MIR17HG and piR-DQ590027, MIR17HG and miR-153 (miR-377), and FOXR2 and miR-153 (miR-377). RNA immunoprecipitation was used to test the interaction between miR-153 (miR-377) and its target proteins. Chromatin immunoprecipitation was performed to detect the interaction between the transcription factor FOXR2 and ZO-1, occludin, and claudin-5. Results piR-DQ590027 was expressed at low levels in glioma-conditioned ECs (GECs) of the in vitro glioma conditioned normal BBB model. Overexpression of piR-DQ590027 down-regulated the expressions of ZO-1, occludin, and claudin-5 and increased the permeability of glioma conditioned normal BBB. MIR17HG had high expression in GECs but miR-153 and miR-377 had low expression. piR-DQ590027 bound to and negatively regulated MIR17HG. FOXR2 was a downstream target of miR-153 and miR-377; MIR17HG bound separately to miR-153 and miR-377 and negatively regulated their ability to mediate FOXR2 expression. FOXR2 associated with the promoter regions of ZO-1, occludin, and claudin-5 in GECs to promote their transcription. Conclusion The piR-DQ590027/MIR17HG/miR-153 (miR-377)/FOXR2 pathway plays an important role in regulating glioma conditioned normal BBB permeability and provides a new target for the comprehensive treatment of glioma. Electronic supplementary material The online version of this article (10.1186/s13046-018-0886-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xue Leng
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Shuyuan Shen
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Hai Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.,Liaoning Research Center for Clinical Medicine in Nervous System Disease, Shenyang, 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, 110004, People's Republic of China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Jiajia Chen
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Lini Zhao
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xuelei Ruan
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, 110122, People's Republic of China. .,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
18
|
Li B, Huang W, Cao N, Lou G. Forkhead‐box R2 promotes metastasis and growth by stimulating angiogenesis and activating hedgehog signaling pathway in ovarian cancer. J Cell Biochem 2018; 119:7780-7789. [PMID: 29943836 DOI: 10.1002/jcb.27148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/18/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Bing Li
- Department of Gynecology Harbin Medical University Cancer Hospital Harbin China
| | - Wei Huang
- Department of Gynecology Harbin Medical University Cancer Hospital Harbin China
| | - Ning Cao
- Department of Gynecology Harbin Medical University Cancer Hospital Harbin China
| | - Ge Lou
- Department of Gynecology Harbin Medical University Cancer Hospital Harbin China
| |
Collapse
|
19
|
Wang XH, Cui YX, Wang ZM, Liu J. Down-regulation of FOXR2 inhibits non-small cell lung cancer cell proliferation and invasion through the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2018; 500:229-235. [PMID: 29634928 DOI: 10.1016/j.bbrc.2018.04.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 04/06/2018] [Indexed: 02/08/2023]
Abstract
Forkhead box R2 (FOXR2), a new member of the FOX family, is an important player in a wide range of cellular processes such as proliferation, migration, differentiation and apoptosis. Recently, FOXR2 has been reported to be implicated in cancer development. However, the biological functions of FOXR2 in non-small cell lung cancer (NSCLC) remain unclear. In this study, we investigated the specific role of FOXR2 in NSCLC. The results showed that down-regulation of FOXR2 significantly inhibited NSCLC cell proliferation and invasion in vitro and suppressed NSCLC cell growth and metastasis in vivo. In addition, the decrease in FOXR2 expression markedly reduced the protein levels of β-catenin, cyclinD1 and c-Myc and hence inactivated the Wnt/β-catenin pathway in NSCLC cells. Taken together, we concluded that FOXR2 might be considered as a promising therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Xin-Hua Wang
- Department of Clinical Laboratory, Linyi City Central Hospital, Linyi, 276400, China
| | - Yan-Xiang Cui
- Department of Clinical Laboratory, Traditional Chinese Medical Hospital of Huangdao District, Qingdao, 266000, China
| | - Zhen-Min Wang
- Department of Clinical Laboratory, Linyi City Central Hospital, Linyi, 276400, China
| | - Jian Liu
- Department of Clinical Laboratory, Linyi City Central Hospital, Linyi, 276400, China.
| |
Collapse
|
20
|
Wei AW, Li LF. Long non-coding RNA SOX21-AS1 sponges miR-145 to promote the tumorigenesis of colorectal cancer by targeting MYO6. Biomed Pharmacother 2017; 96:953-959. [PMID: 29217166 DOI: 10.1016/j.biopha.2017.11.145] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 01/03/2023] Open
Abstract
Emerging evidences have proved that long non-coding RNAs (lncRNAs) act as important molecular regulator in the tumor progression, including colorectal cancer (CRC). LncRNA SOX21-AS1 has been verified as oncogenic molecular in other cancers and tumorigenesis. In present study, our team investigates the clinical characteristic and molecular function in CRC carcinogenesis. Results showed that lncRNA SOX21-AS1 expression was significantly over-expressed in CRC tissue samples and cells. The aberrant over-expression of SOX21-AS1 indicated poor prognosis of CRC patients. In vitro and in vivo validation experiments, SOX21-AS1 silencing inhibited the proliferation, invasion, and decreased the tumor growth of CRC cells. Moreover, miR-145 was proved to be the target of SOX21-AS1, besides, myosin VI (MYO6) was found to be one of the targets of miR-145 using bioinformatics prediction programs and rescue confirmation experiments. In summary, our study reveals the tumorigenic effect of lncRNA SOX21-AS1 in CRC cells via targeting miR-145/MYO6, providing a novel insight for CRC carcinogenesis.
Collapse
Affiliation(s)
- An-Wei Wei
- Department of Gastroenterology, Huaihe Hospital of Henan University, 475000, China.
| | - Li-Fang Li
- Department of General Surgery, Huaihe Hospital of Henan University, 475000, China
| |
Collapse
|