1
|
Kundu M, Dey A, Maji PK, Mandal M. Targeting friend leukemia integration 1: A promising approach for prevention and treatment of solid tumors. Int J Biol Macromol 2025; 309:143080. [PMID: 40228766 DOI: 10.1016/j.ijbiomac.2025.143080] [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: 07/07/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Friend leukemia integration 1 (FLI1) is an ETS transcription factor first identified in erythroleukemia. This protein contributes to various cellular functions such as cell growth and proliferation, apoptosis, angiogenesis, etc. FLI1 is also known to be involved in tumorigenesis. The role of this transcription factor as a proto-oncogene, promoting cancer progression, especially Ewing sarcoma, is well reported. Recent research has found the connection of FLI1 with other solid cancers, including breast cancer, prostate cancer, glioma, and lung cancer. The role of this protein in solid cancers is also controversial. FLI1 is found to promote and suppress cancer growth and progression, particularly in Ewing sarcoma and breast cancer. This review article aims to provide a detailed perception of the FLI1-associated mechanisms in various solid cancers for preventive and therapeutic implications. The result of bioinformatic analysis using the cBioportal database (https://www.cbioportal.org/) is also presented in this article to understand the effect of this protein on solid cancers. Moreover, the current status of FLI1 targeting agents for preventing and treating solid cancers has been focused. Several studies established the efficacy of FLI1 inhibitors in solid tumor therapy. A few reports are also available on the effect of FLI1 agonists on solid tumors. This article discussed different FLI1 targeting agents to provide insight into the FLI1 targeting mechanisms required for discovering more potent FLI1 targeting agents and better therapeutic outcomes.
Collapse
Affiliation(s)
- Moumita Kundu
- Department of Pharmaceutical Technology, Brainware University, Barasat, India; Center for Multidisciplinary Research & Innovations, Brainware University, Barasat, India.
| | - Ankita Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Pallab Kumar Maji
- Department of Pharmaceutical Technology, Brainware University, Barasat, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
2
|
Angarola BL, Sharma S, Katiyar N, Kang HG, Nehar-Belaid D, Park S, Gott R, Eryilmaz GN, LaBarge MA, Palucka K, Chuang JH, Korstanje R, Ucar D, Anczukόw O. Comprehensive single-cell aging atlas of healthy mammary tissues reveals shared epigenomic and transcriptomic signatures of aging and cancer. NATURE AGING 2025; 5:122-143. [PMID: 39587369 PMCID: PMC11754115 DOI: 10.1038/s43587-024-00751-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 10/16/2024] [Indexed: 11/27/2024]
Abstract
Aging is the greatest risk factor for breast cancer; however, how age-related cellular and molecular events impact cancer initiation is unknown. In this study, we investigated how aging rewires transcriptomic and epigenomic programs of mouse mammary glands at single-cell resolution, yielding a comprehensive resource for aging and cancer biology. Aged epithelial cells exhibit epigenetic and transcriptional changes in metabolic, pro-inflammatory and cancer-associated genes. Aged stromal cells downregulate fibroblast marker genes and upregulate markers of senescence and cancer-associated fibroblasts. Among immune cells, distinct T cell subsets (Gzmk+, memory CD4+, γδ) and M2-like macrophages expand with age. Spatial transcriptomics reveals co-localization of aged immune and epithelial cells in situ. Lastly, we found transcriptional signatures of aging mammary cells in human breast tumors, suggesting possible links between aging and cancer. Together, these data uncover that epithelial, immune and stromal cells shift in proportions and cell identity, potentially impacting cell plasticity, aged microenvironment and neoplasia risk.
Collapse
Affiliation(s)
| | | | - Neerja Katiyar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Hyeon Gu Kang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - SungHee Park
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Giray N Eryilmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark A LaBarge
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA.
| | - Olga Anczukόw
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA.
| |
Collapse
|
3
|
Wang J, Wang C, Hu A, Yu K, Kuang Y, Gajendran B, Zacksenhaus E, Sample KM, Xiao X, Liu W, Ben-David Y. FLI1 induces erythroleukemia through opposing effects on UBASH3A and UBASH3B expression. BMC Cancer 2024; 24:326. [PMID: 38461240 PMCID: PMC10925000 DOI: 10.1186/s12885-024-12075-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 03/01/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND FLI1 is an oncogenic transcription factor that promotes diverse malignancies through mechanisms that are not fully understood. Herein, FLI1 is shown to regulate the expression of Ubiquitin Associated and SH3 Domain Containing A/B (UBASH3A/B) genes. UBASH3B and UBASH3A are found to act as an oncogene and tumor suppressor, respectively, and their combined effect determines erythroleukemia progression downstream of FLI1. METHODS Promoter analysis combined with luciferase assays and chromatin immunoprecipitation (ChIP) analysis were applied on the UBASH3A/B promoters. RNAseq analysis combined with bioinformatic was used to determine the effect of knocking-down UBASH3A and UBASH3B in leukemic cells. Downstream targets of UBASH3A/B were inhibited in leukemic cells either via lentivirus-shRNAs or small molecule inhibitors. Western blotting and RT-qPCR were used to determine transcription levels, MTT assays to assess proliferation rate, and flow cytometry to examine apoptotic index. RESULTS Knockdown of FLI1 in erythroleukemic cells identified the UBASH3A/B genes as potential downstream targets. Herein, we show that FLI1 directly binds to the UBASH3B promoter, leading to its activation and leukemic cell proliferation. In contrast, FLI1 indirectly inhibits UBASH3A transcription via GATA2, thereby antagonizing leukemic growth. These results suggest oncogenic and tumor suppressor roles for UBASH3B and UBASH3A in erythroleukemia, respectively. Mechanistically, we show that UBASH3B indirectly inhibits AP1 (FOS and JUN) expression, and that its loss leads to inhibition of apoptosis and acceleration of proliferation. UBASH3B also positively regulates the SYK gene expression and its inhibition suppresses leukemia progression. High expression of UBASH3B in diverse tumors was associated with worse prognosis. In contrast, UBASH3A knockdown in erythroleukemic cells increased proliferation; and this was associated with a dramatic induction of the HSP70 gene, HSPA1B. Accordingly, knockdown of HSPA1B in erythroleukemia cells significantly accelerated leukemic cell proliferation. Accordingly, overexpression of UBASH3A in different cancers was predominantly associated with good prognosis. These results suggest for the first time that UBASH3A plays a tumor suppressor role in part through activation of HSPA1B. CONCLUSIONS FLI1 promotes erythroleukemia progression in part by modulating expression of the oncogenic UBASH3B and tumor suppressor UBASH3A.
Collapse
MESH Headings
- Humans
- Leukemia, Erythroblastic, Acute/genetics
- Leukemia, Erythroblastic, Acute/pathology
- Proto-Oncogene Protein c-fli-1/genetics
- Proto-Oncogene Protein c-fli-1/metabolism
- RNA, Small Interfering/genetics
- Genes, Tumor Suppressor
- Gene Expression Regulation
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
- Oncogene Proteins, Fusion/genetics
- RNA-Binding Protein EWS/genetics
- Adaptor Proteins, Signal Transducing/metabolism
Collapse
Affiliation(s)
- Jie Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China
- Natural Products Research Center of Guizhou Province, High Tech Zone, Province Science City, Baiyun District, Guiyang, 550014, China
| | - Chunlin Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China
- Natural Products Research Center of Guizhou Province, High Tech Zone, Province Science City, Baiyun District, Guiyang, 550014, China
| | - Anling Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China
- Natural Products Research Center of Guizhou Province, High Tech Zone, Province Science City, Baiyun District, Guiyang, 550014, China
| | - Kunlin Yu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China
- Natural Products Research Center of Guizhou Province, High Tech Zone, Province Science City, Baiyun District, Guiyang, 550014, China
| | - Yi Kuang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China
- Natural Products Research Center of Guizhou Province, High Tech Zone, Province Science City, Baiyun District, Guiyang, 550014, China
| | - Babu Gajendran
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou Province, Guiyang, 550025, People's Republic of China
| | - Eldad Zacksenhaus
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China
- Natural Products Research Center of Guizhou Province, High Tech Zone, Province Science City, Baiyun District, Guiyang, 550014, China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China.
- Natural Products Research Center of Guizhou Province, High Tech Zone, Province Science City, Baiyun District, Guiyang, 550014, China.
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang-550014, Guizhou, People's Republic of China.
- Natural Products Research Center of Guizhou Province, High Tech Zone, Province Science City, Baiyun District, Guiyang, 550014, China.
| |
Collapse
|
4
|
Pei J, Peng Y, Ma K, Lan C, Zhang T, Li Y, Chen X, Gao H. Integrated analysis reveals FLI1 regulates the tumor immune microenvironment via its cell-type-specific expression and transcriptional regulation of distinct target genes of immune cells in breast cancer. BMC Genomics 2024; 25:250. [PMID: 38448802 PMCID: PMC10916124 DOI: 10.1186/s12864-024-10174-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Immunotherapy is a practical therapeutic approach in breast cancer (BRCA), and the role of FLI1 in immune regulation has gradually been unveiled. However, the specific role of FLI1 in BRCA was conflicted; thus, additional convincing evidence is needed. METHODS We explored the upstream regulation of FLI1 expression via summary data-based Mendelian randomization (SMR) analysis and ncRNA network construction centering on FLI1 using BRCA genome-wide association study (GWAS) summary data with expression quantitative trait loci (eQTLs) and DNA methylation quantitative trait loci (mQTLs) from the blood and a series of in silico analyses, respectively. We illuminated the downstream function of FLI1 in immune regulation by integrating a series of analyses of single-cell RNA sequence data (scRNA-seq). RESULTS We verified a causal pathway from FLI1 methylation to FLI1 gene expression to BRCA onset and demonstrated that FLI1 was downregulated in BRCA. FLI1, a transcription factor, served as myeloid and T cells' communication regulator by targeting immune-related ligands and receptor transcription in BRCA tissues. We constructed a ceRNA network centering on FLI1 that consisted of three LncRNAs (CKMT2-AS1, PSMA3-AS1, and DIO3OS) and a miRNA (hsa-miR-324-5p), and the expression of FLI1 was positively related to a series of immune-related markers, including immune cell infiltration, biomarkers of immune cells, and immune checkpoints. CONCLUSION Low-methylation-induced or ncRNA-mediated downregulation of FLI1 is associated with poor prognosis, and FLI1 might regulate the tumor immune microenvironment via a cell-type-specific target genes manner in BRCA.
Collapse
Affiliation(s)
- Jianying Pei
- National Research Institute for Family Planning, Beijing, 100081, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- Institute of Clinical Medicine, Gansu Provincial Maternity and Child-care Hospital (Gansu Provincial Central Hospital), Lanzhou, 730000, China
| | - Ying Peng
- Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Kexin Ma
- National Research Institute for Family Planning, Beijing, 100081, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Chunyan Lan
- National Research Institute for Family Planning, Beijing, 100081, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Tingting Zhang
- Medical College of Northwest Minzu University, Lanzhou, 730030, China
| | - Yan Li
- Medical College of Northwest Minzu University, Lanzhou, 730030, China
| | - Xiaofang Chen
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.
| | - Huafang Gao
- National Research Institute for Family Planning, Beijing, 100081, China.
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
5
|
Cho U, Cha HJ, Kim HJ, Min SK, Kim HK, Jung HR, Park G, Kim JE. FLI-1 is expressed in a wide variety of hematolymphoid neoplasms: a special concern in the differential diagnosis. Clin Exp Med 2024; 24:18. [PMID: 38280044 PMCID: PMC10821826 DOI: 10.1007/s10238-023-01284-x] [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: 09/19/2023] [Accepted: 11/07/2023] [Indexed: 01/29/2024]
Abstract
Friend Leukemia Virus Integration 1 (FLI-1) is a member of E26 transformation-specific family of transcription factors that participates in hematopoietic and vascular endothelial cell development. Immunohistochemical detection of FLI-1 has been widely used to diagnose vascular tumors or, more evidently, Ewing's sarcoma. However, the expression pattern of FLI-1 in hematolymphoid neoplasms remains unclear. Therefore, in this study, we aimed to investigate the expression of FLI-1 in these tumors, focusing on high-grade lesions, which presents a diagnostic challenge by mimicking Ewing's sarcoma. We evaluated the expression FLI-1 in various types of lymphoid and plasmacytic tumors, including 27 plasmablastic lymphomas, 229 diffuse large B-cell lymphomas, 22 precursor T- or B-lymphoblastic lymphomas, 24 angioimmunoblastic-type nodal T-follicular helper cell lymphomas, 52 peripheral T-cell lymphomas, NOS, 18 Burkitt lymphomas, 18 non-gastric lymphomas of mucosa-associated lymphoid tissue, 38 chronic lymphocytic leukemia/small lymphocytic lymphomas, 15 mantle cell lymphomas, 23 gastric MALT lymphomas, 50 plasma cell myelomas, and 38 follicular lymphomas. We calculated the H-scores of FLI-1 immunostaining, ranging from 0 to 200, and used the scores to analyze the clinicopathological significance of FLI-1 statistically. FLI-1 was expressed to varying degrees in all types of hematological tumors. FLI-1 expression was detected in 84.1% of patients (466/554). FLI-1 was highly expressed in precursor T- or B-lymphoblastic lymphomas. Follicular lymphomas exhibited low FLI-1 expression. In plasmablastic lymphoma, 85.2% of the patients were focally positive for FLI-1. FLI-1 expression did not correlate with clinicopathological variables, such as demographic data or disease stage, in patients with plasmablastic lymphoma and diffuse large B-cell lymphoma. However, FLI-1 overexpression was associated with poorer overall survival in patients with plasmablastic lymphoma. This study demonstrates that FLI-1 is expressed in various hematolymphoid neoplasms. FLI-1 expression can lead to diagnostic confusion, especially in small blue round cell tumors, such as lymphoblastic lymphoma, plasmablastic lymphoma, and plasma cell myeloma, when distinguishing tumors positive for CD99 and CD56 without CD3, CD20, or CD45. Our findings also suggested the possibility of FLI-1 as a potential prognostic biomarker for plasmablastic lymphoma.
Collapse
Affiliation(s)
- Uiju Cho
- Department of Pathology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hee Jeong Cha
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Hyun Jung Kim
- Department of Pathology, Sanggye Paik Hospital, Inje University, Seoul, Republic of Korea
| | - Soo Kee Min
- Department of Pathology, Chung-ang University Gwangmyeong Hospital, Gwangmyeong, Republic of Korea
| | - Hee Kyung Kim
- Department of Pathology, Soonchunhyang University Hospital, Bucheon, Republic of Korea
| | - Hye Ra Jung
- Department of Pathology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Gyeongsin Park
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodaero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Ji Eun Kim
- Department of Pathology, Seoul National University Boramae Hospital, 20 Boramae-Ro 5-Gil, Dongjak-Gu, Seoul, 07061, Republic of Korea.
| |
Collapse
|
6
|
Angarola BL, Sharma S, Katiyar N, Gu Kang H, Nehar-Belaid D, Park S, Gott R, Eryilmaz GN, LaBarge MA, Palucka K, Chuang JH, Korstanje R, Ucar D, Anczukow O. Comprehensive single cell aging atlas of mammary tissues reveals shared epigenomic and transcriptomic signatures of aging and cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563147. [PMID: 37961129 PMCID: PMC10634680 DOI: 10.1101/2023.10.20.563147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Aging is the greatest risk factor for breast cancer; however, how age-related cellular and molecular events impact cancer initiation is unknown. We investigate how aging rewires transcriptomic and epigenomic programs of mouse mammary glands at single cell resolution, yielding a comprehensive resource for aging and cancer biology. Aged epithelial cells exhibit epigenetic and transcriptional changes in metabolic, pro-inflammatory, or cancer-associated genes. Aged stromal cells downregulate fibroblast marker genes and upregulate markers of senescence and cancer-associated fibroblasts. Among immune cells, distinct T cell subsets (Gzmk+, memory CD4+, γδ) and M2-like macrophages expand with age. Spatial transcriptomics reveal co-localization of aged immune and epithelial cells in situ. Lastly, transcriptional signatures of aging mammary cells are found in human breast tumors, suggesting mechanistic links between aging and cancer. Together, these data uncover that epithelial, immune, and stromal cells shift in proportions and cell identity, potentially impacting cell plasticity, aged microenvironment, and neoplasia risk.
Collapse
Affiliation(s)
| | | | - Neerja Katiyar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Hyeon Gu Kang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - SungHee Park
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Giray N Eryilmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark A LaBarge
- Beckman Research Institute at City of Hope, Duarte, CA, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA
| | - Olga Anczukow
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA
| |
Collapse
|
7
|
Salmerón-Bárcenas EG, Mendoza-Catalan MA, Ramírez-Bautista ÁU, Lozano-Santos RA, Torres-Rojas FI, Ávila-López PA, Zacapala-Gómez AE. Identification of Mir-182-3p/FLI-1 Axis as a Key Signaling in Immune Response in Cervical Cancer: A Comprehensive Bioinformatic Analysis. Int J Mol Sci 2023; 24:ijms24076032. [PMID: 37047006 PMCID: PMC10094573 DOI: 10.3390/ijms24076032] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
miRNAs modulate gene expression and play critical functions as oncomiRs or tumor suppressors. The miR-182-3p is important in chemoresistance and cancer progression in breast, lung, osteosarcoma, and ovarian cancer. However, the role of miR-182-3p in cervical cancer (CC) has not been elucidated. AIM To analyze the role of miR-182-3p in CC through a comprehensive bioinformatic analysis. METHODS Gene Expression Omnibus (GEO) databases were used for the expression analysis. The mRNA targets of miR-182-3p were identified using miRDB, TargetScanHuman, and miRPathDB. The prediction of island CpG was performed using the MethPrimer program. The transcription factor binding sites in the FLI-1 promoter were identified using ConSite+, Alibaba2, and ALGGEN-PROMO. The protein-protein interaction (PPI) analysis was performed in STRING 11.5. RESULTS miR-182-3p was significantly overexpressed in CC patients and has potential as a diagnostic. We identified 330 targets of miR-182-3p including FLI-1, which downregulates its expression in CC. Additionally, the aberrant methylation of the FLI-1 promoter and Ap2a transcription factor could be involved in downregulating FLI1 expression. Finally, we found that FLI-1 is a possible key gene in the immune response in CC. CONCLUSIONS The miR-182-3p/FLI-1 axis plays a critical role in immune response in CC.
Collapse
Affiliation(s)
- Eric Genaro Salmerón-Bárcenas
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, CINVESTAV, Ciudad de México 07360, Mexico
| | - Miguel Angel Mendoza-Catalan
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Mexico
| | - Ángela Uray Ramírez-Bautista
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Mexico
| | - Rafael Acxel Lozano-Santos
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Mexico
| | - Francisco Israel Torres-Rojas
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Mexico
| | - Pedro Antonio Ávila-López
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ana Elvira Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39070, Mexico
| |
Collapse
|
8
|
Chen E, Huang J, Chen M, Wu J, Ouyang P, Wang X, Shi D, Liu Z, Zhu W, Sun H, Yang S, Zhang B, Deng W, Qiu H, Xie F. FLI1 regulates radiotherapy resistance in nasopharyngeal carcinoma through TIE1-mediated PI3K/AKT signaling pathway. J Transl Med 2023; 21:134. [PMID: 36814284 PMCID: PMC9945741 DOI: 10.1186/s12967-023-03986-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Radiotherapy resistance is the main cause of treatment failure in nasopharyngeal carcinoma (NPC), which leads to poor prognosis. It is urgent to elucidate the molecular mechanisms underlying radiotherapy resistance. METHODS RNA-seq analysis was applied to five paired progressive disease (PD) and complete response (CR) NPC tissues. Loss-and gain-of-function assays were used for oncogenic function of FLI1 both in vitro and in vivo. RNA-seq analysis, ChIP assays and dual luciferase reporter assays were performed to explore the interaction between FLI1 and TIE1. Gene expression with clinical information from tissue microarray of NPC were analyzed for associations between FLI1/TIE1 expression and NPC prognosis. RESULTS FLI1 is a potential radiosensitivity regulator which was dramatically overexpressed in the patients with PD to radiotherapy compared to those with CR. FLI1 induced radiotherapy resistance and enhanced the ability of DNA damage repair in vitro, and promoted radiotherapy resistance in vivo. Mechanistic investigations showed that FLI1 upregulated the transcription of TIE1 by binding to its promoter, thus activated the PI3K/AKT signaling pathway. A decrease in TIE1 expression restored radiosensitivity of NPC cells. Furthermore, NPC patients with high levels of FLI1 and TIE1 were correlated with poor prognosis. CONCLUSION Our study has revealed that FLI1 regulates radiotherapy resistance of NPC through TIE1-mediated PI3K/AKT signaling pathway, suggesting that targeting the FLI1/TIE1 signaling pathway could be a potential therapeutic strategy to enhance the efficacy of radiotherapy in NPC.
Collapse
Affiliation(s)
- Enni Chen
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Jiajia Huang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Miao Chen
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Jiawei Wu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Puyun Ouyang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Xiaonan Wang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Dingbo Shi
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Zhiqiao Liu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Wancui Zhu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Haohui Sun
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Shanshan Yang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Baoyu Zhang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Wuguo Deng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Huijuan Qiu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Fangyun Xie
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
9
|
Guneri-Sozeri PY, Özden-Yılmaz G, Kisim A, Cakiroglu E, Eray A, Uzuner H, Karakülah G, Pesen-Okvur D, Senturk S, Erkek-Ozhan S. FLI1 and FRA1 transcription factors drive the transcriptional regulatory networks characterizing muscle invasive bladder cancer. Commun Biol 2023; 6:199. [PMID: 36805539 PMCID: PMC9941102 DOI: 10.1038/s42003-023-04561-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Bladder cancer is mostly present in the form of urothelium carcinoma, causing over 150,000 deaths each year. Its histopathological classification as muscle invasive (MIBC) and non-muscle invasive (NMIBC) is the most prominent aspect, affecting the prognosis and progression of this disease. In this study, we defined the active regulatory landscape of MIBC and NMIBC cell lines using H3K27ac ChIP-seq and used an integrative approach to combine our findings with existing data. Our analysis revealed FRA1 and FLI1 as two critical transcription factors differentially regulating MIBC regulatory landscape. We show that FRA1 and FLI1 regulate the genes involved in epithelial cell migration and cell junction organization. Knock-down of FRA1 and FLI1 in MIBC revealed the downregulation of several EMT-related genes such as MAP4K4 and FLOT1. Further, ChIP-SICAP performed for FRA1 and FLI1 enabled us to infer chromatin binding partners of these transcription factors and link this information with their target genes. Finally, we show that knock-down of FRA1 and FLI1 result in significant reduction of invasion capacity of MIBC cells towards muscle microenvironment using IC-CHIP assays. Our results collectively highlight the role of these transcription factors in selection and design of targeted options for treatment of MIBC.
Collapse
Affiliation(s)
- Perihan Yagmur Guneri-Sozeri
- grid.21200.310000 0001 2183 9022Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey ,grid.21200.310000 0001 2183 9022Dokuz Eylül University Izmir International Biomedicine and Genome Institute, Inciralti, 35340 Izmir, Turkey
| | - Gülden Özden-Yılmaz
- grid.21200.310000 0001 2183 9022Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey
| | - Asli Kisim
- grid.419609.30000 0000 9261 240XIzmir Institute of Technology, Urla, 35430 Izmir, Turkey
| | - Ece Cakiroglu
- grid.21200.310000 0001 2183 9022Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey ,grid.21200.310000 0001 2183 9022Dokuz Eylül University Izmir International Biomedicine and Genome Institute, Inciralti, 35340 Izmir, Turkey
| | - Aleyna Eray
- grid.21200.310000 0001 2183 9022Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey ,grid.21200.310000 0001 2183 9022Dokuz Eylül University Izmir International Biomedicine and Genome Institute, Inciralti, 35340 Izmir, Turkey
| | - Hamdiye Uzuner
- grid.21200.310000 0001 2183 9022Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey ,grid.21200.310000 0001 2183 9022Dokuz Eylül University Izmir International Biomedicine and Genome Institute, Inciralti, 35340 Izmir, Turkey
| | - Gökhan Karakülah
- grid.21200.310000 0001 2183 9022Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey ,grid.21200.310000 0001 2183 9022Dokuz Eylül University Izmir International Biomedicine and Genome Institute, Inciralti, 35340 Izmir, Turkey
| | - Devrim Pesen-Okvur
- grid.419609.30000 0000 9261 240XIzmir Institute of Technology, Urla, 35430 Izmir, Turkey
| | - Serif Senturk
- grid.21200.310000 0001 2183 9022Izmir Biomedicine and Genome Center, Inciralti, 35340 Izmir, Turkey ,grid.21200.310000 0001 2183 9022Dokuz Eylül University Izmir International Biomedicine and Genome Institute, Inciralti, 35340 Izmir, Turkey
| | - Serap Erkek-Ozhan
- Izmir Biomedicine and Genome Center, Inciralti, 35340, Izmir, Turkey.
| |
Collapse
|
10
|
Lam L, Tien T, Wildung M, White L, Sellon RK, Fidel JL, Shelden EA. Comparative whole transcriptome analysis of gene expression in three canine soft tissue sarcoma types. PLoS One 2022; 17:e0273705. [PMID: 36099287 PMCID: PMC9469979 DOI: 10.1371/journal.pone.0273705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
Soft tissue sarcomas are pleiotropic tumors of mesenchymal cell origin. These tumors are rare in humans but common in veterinary practice, where they comprise up to 15% of canine skin and subcutaneous cancers. Because they present similar morphologies, primary sites, and growth characteristics, they are treated similarly, generally by surgical resection followed by radiation therapy. Previous studies have examined a variety of genetic changes as potential drivers of tumorigenesis and progression in soft tissue sarcomas as well as their use as markers for soft tissue sarcoma subtypes. However, few studies employing next generation sequencing approaches have been published. Here, we have examined gene expression patterns in canine soft tissue sarcomas using RNA-seq analysis of samples obtained from archived formalin-fixed and paraffin-embedded tumors. We provide a computational framework for using resulting data to categorize tumors, perform cross species comparisons and identify genetic changes associated with tumorigenesis. Functional overrepresentation analysis of differentially expressed genes further implicate both common and tumor-type specific transcription factors as potential mediators of tumorigenesis and aggression. Implications for tumor-type specific therapies are discussed. Our results illustrate the potential utility of this approach for the discovery of new therapeutic approaches to the management of canine soft tissue sarcomas and support the view that both common and tumor-type specific mechanisms drive the development of these tumors.
Collapse
Affiliation(s)
- Lydia Lam
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Tien Tien
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Mark Wildung
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Laura White
- Washington Animal Disease Diagnostic Laboratory, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Rance K. Sellon
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Janean L. Fidel
- Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| | - Eric A. Shelden
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States of America
| |
Collapse
|
11
|
Chen W, Liu Y, Kang S, Lv X, Fu W, Zhang J, Song C. LINC00092 Modulates Oxidative Stress and Glycolysis of Breast Cancer Cells via Pyruvate Carboxylase-Mediated AKT/mTOR Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5215748. [PMID: 35799892 PMCID: PMC9256459 DOI: 10.1155/2022/5215748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/16/2022] [Accepted: 05/24/2022] [Indexed: 11/23/2022]
Abstract
Background The discovery of noncoding RNAs (ncRNAs) offers new options for cancer-targeted therapy. This study is aimed at exploring the regulatory function of LINC00092 on breast cancer (BC) oxidative stress and glycolysis, along with internal mechanism concerning pyruvate carboxylase (PC). Methods Bioinformatics analysis was used to explore LINC00092 (or friend leukemia virus integration 1 (FLI1)) expression on BC progression, as well as oxidative stress and glycolysis in BC. After LINC00092 overexpression or silence, BC cell viability, proliferation, migration, invasion, oxidative stress, glycolysis, and AKT/mTOR pathway were detected. Following 2-DG, SC79, or MK2206 treatment, effects of LINC00092 on BC cells were measured. Moreover, regulatory activity of LINC00092 in PC expression was analyzed. Whether PC participated in the modulation of LINC00092 on BC cell functions was explored. Results LINC00092 was lowly expressed in BC and negatively related to BC progression. FLI1 bound to LINC00092 promoter to positively modulate LINC00092. LINC00092 overexpression inhibited BC cell proliferation, migration, invasion, oxidative stress, glycolysis, and AKT/mTOR pathway and likewise suppressed BC growth in vivo. Silence of LINC00092 had opposite influences. 2-DG partially reversed the LINC00092 silence-resulted increase of BC cell proliferation. SC79 alleviated the function of LINC00092 overexpression on BC cell functions. MK2206 had the contrary influence of SC79. Besides, LINC00092 bound to PC to modulate ubiquitination degradation of PC protein. PC took part in the influences of LINC00092 on BC cell functions. Conclusions LINC0092 modulates oxidative stress and glycolysis of BC cells via the PC-mediated AKT/mTOR pathway, which is possibly a target for BC diagnosis and therapy.
Collapse
Affiliation(s)
- Wei Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
| | - Yushan Liu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
| | - Shaohong Kang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
| | - Xinying Lv
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
| | - Wenfen Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
| | - Jie Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province 350001, China
| | - Chuangui Song
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province 350001, China
| |
Collapse
|
12
|
Bou-Fakhredin R, De Franceschi L, Motta I, Eid AA, Taher AT, Cappellini MD. Redox Balance in β-Thalassemia and Sickle Cell Disease: A Love and Hate Relationship. Antioxidants (Basel) 2022; 11:antiox11050967. [PMID: 35624830 PMCID: PMC9138068 DOI: 10.3390/antiox11050967] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
β-thalassemia and sickle cell disease (SCD) are inherited hemoglobinopathies that result in both quantitative and qualitative variations in the β-globin chain. These in turn lead to instability in the generated hemoglobin (Hb) or to a globin chain imbalance that affects the oxidative environment both intracellularly and extracellularly. While oxidative stress is not among the primary etiologies of β-thalassemia and SCD, it plays a significant role in the pathogenesis of these diseases. Different mechanisms exist behind the development of oxidative stress; the result of which is cytotoxicity, causing the oxidation of cellular components that can eventually lead to cell death and organ damage. In this review, we summarize the mechanisms of oxidative stress development in β-thalassemia and SCD and describe the current and potential antioxidant therapeutic strategies. Finally, we discuss the role of targeted therapy in achieving an optimal redox balance.
Collapse
Affiliation(s)
- Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
| | - Lucia De Franceschi
- Department of Medicine, University of Verona, and Azienda Ospedaliera Universitaria Verona, 37128 Verona, Italy;
| | - Irene Motta
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
- UOC General Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Assaad A. Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Ali T. Taher
- Division of Hematology-Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
- UOC General Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence:
| |
Collapse
|
13
|
Li L, Yu J, Cheng S, Peng Z, Ben-David Y, Luo H. Transcription factor Fli-1 as a new target for antitumor drug development. Int J Biol Macromol 2022; 209:1155-1168. [PMID: 35447268 DOI: 10.1016/j.ijbiomac.2022.04.076] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023]
Abstract
The transcription factor Friend leukemia virus integration 1 (Fli-1) belonging to the E26 Transformation-Specific (ETS) transcription factor family is not only expressed in normal cells such as hematopoietic stem cells and vascular endothelial cells, but also abnormally expressed in various malignant tumors including Ewing sarcoma, Merkel cell sarcoma, small cell lung carcinoma, benign or malignant hemangioma, squamous cell carcinoma, adenocarcinoma, bladder cancer, leukemia, and lymphoma. Fli-1 binds to the promoter or enhancer of the target genes and participates in a variety of physiological and pathological processes of tumor cells, including cell growth, proliferation, differentiation, and apoptosis. The expression of Fli-1 gene is related to the specific biological functions and characteristics of the tissue in which it is located. In tumor research, Fli-1 gene is used as a specific marker for the occurrence, metastasis, efficacy, and prognosis of tumors, thus, a potential new target for tumor diagnosis and treatment. These studies indicated that Fli-1 may be a specific candidate for antitumor drug development. Recent studies identified small molecules regulating Fli-1 thanks to our screened strategy of natural products and their derivatives. Therefore, in this review, the advanced research on Fli-1 as a target for antitumor drug development is analyzed in different cancers. The inhibitors and agonists of Fli-1 that regulate its expression are introduced and their clinical applications in the treatment of cancer, thus providing new therapeutic strategies.
Collapse
Affiliation(s)
- Lanlan Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; College of Pharmacy, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Jia Yu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China
| | - Sha Cheng
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China
| | - Zhilin Peng
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China
| | - Heng Luo
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, P.R. China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Science, Guiyang 550014, P.R. China.
| |
Collapse
|
14
|
Yarmishyn AA, Ishola AA, Chen CY, Verusingam ND, Rengganaten V, Mustapha HA, Chuang HK, Teng YC, Phung VL, Hsu PK, Lin WC, Ma HI, Chiou SH, Wang ML. Circular RNAs Modulate Cancer Hallmark and Molecular Pathways to Support Cancer Progression and Metastasis. Cancers (Basel) 2022; 14:cancers14040862. [PMID: 35205610 PMCID: PMC8869994 DOI: 10.3390/cancers14040862] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Circular RNAs (circRNA) are a type of RNA molecule of circular shape that are now being extensively studied due to the important roles they play in different biological processes. In addition, they were also shown to be implicated in disease such as cancer. Cancer is a complex process which is often defined by a combination of specific processes called cancer hallmarks. In this review, we summarize the literature on circRNAs in cancer and classify them as being implicated in specific cancer hallmarks. Abstract Circular RNAs (circRNAs) are noncoding products of backsplicing of pre-mRNAs which have been established to possess potent biological functions. Dysregulated circRNA expression has been linked to diseases including different types of cancer. Cancer progression is known to result from the dysregulation of several molecular mechanisms responsible for the maintenance of cellular and tissue homeostasis. The dysregulation of these processes is defined as cancer hallmarks, and the molecular pathways implicated in them are regarded as the targets of therapeutic interference. In this review, we summarize the literature on the investigation of circRNAs implicated in cancer hallmark molecular signaling. First, we present general information on the properties of circRNAs, such as their biogenesis and degradation mechanisms, as well as their basic molecular functions. Subsequently, we summarize the roles of circRNAs in the framework of each cancer hallmark and finally discuss the potential as therapeutic targets.
Collapse
Affiliation(s)
- Aliaksandr A. Yarmishyn
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
| | - Afeez Adekunle Ishola
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 112, Taiwan
| | - Chieh-Yu Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 112, Taiwan
| | - Nalini Devi Verusingam
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia
| | - Vimalan Rengganaten
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Postgraduate Programme, Department of Preclinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia
| | - Habeebat Aderonke Mustapha
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hao-Kai Chuang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
| | - Yuan-Chi Teng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
| | - Van Long Phung
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Po-Kuei Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wen-Chang Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan;
| | - Hsin-I Ma
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 112, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Genomic Research Center, Academia Sinica, Taipei 112, Taiwan
| | - Mong-Lien Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; (A.A.Y.); (A.A.I.); (C.-Y.C.); (N.D.V.); (V.R.); (H.A.M.); (H.-K.C.); (Y.-C.T.); (V.L.P.); (S.-H.C.)
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Food Safety and Health Risk Assessment, School of Pharmaceutical Sciences, National Yang-Ming Chiao Tung University, Taipei 112, Taiwan
- Correspondence: ; Tel.: +886-2-5568-1156; Fax: +886-2-2875-7435
| |
Collapse
|
15
|
FLI1 regulates inflammation-associated genes to accelerate leukemogenesis. Cell Signal 2022; 92:110269. [DOI: 10.1016/j.cellsig.2022.110269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 01/03/2023]
|
16
|
Barata T, Vieira V, Rodrigues R, Neves RPD, Rocha M. Reconstruction of tissue-specific genome-scale metabolic models for human cancer stem cells. Comput Biol Med 2021; 142:105177. [PMID: 35026576 DOI: 10.1016/j.compbiomed.2021.105177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023]
Abstract
Cancer Stem Cells (CSCs) contribute to cancer aggressiveness, metastasis, chemo/radio-therapy resistance, and tumor recurrence. Recent studies emphasized the importance of metabolic reprogramming of CSCs for the maintenance and progression of the cancer phenotype through both the fulfillment of the energetic requirements and the supply of substrates fundamental for fast-cell growth, as well as through metabolite-induced epigenetic regulation. Therefore, it is of paramount importance to develop therapeutic strategies tailored to target the metabolism of CSCs. In this work, we built computational Genome-Scale Metabolic Models (GSMMs) for CSCs of different tissues. Flux simulations were then used to predict metabolic phenotypes, identify potential therapeutic targets, and spot already-known Transcription Factors (TFs), miRNAs and antimetabolites that could be used as part of drug repurposing strategies against cancer. Results were in accordance with experimental evidence, provided insights of new metabolic mechanisms for already known agents, and allowed for the identification of potential new targets and compounds that could be interesting for further in vitro and in vivo validation.
Collapse
Affiliation(s)
- Tânia Barata
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Vítor Vieira
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal
| | - Rúben Rodrigues
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal
| | - Ricardo Pires das Neves
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal; IIIUC-Institute of Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal.
| | - Miguel Rocha
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal; Department of Informatics, University of Minho.
| |
Collapse
|
17
|
Modi A, Purohit P, Gadwal A, Ukey S, Roy D, Fernandes S, Banerjee M. In-Silico Analysis of Differentially Expressed Genes and Their Regulating microRNA Involved in Lymph Node Metastasis in Invasive Breast Carcinoma. Cancer Invest 2021; 40:55-72. [PMID: 34396887 DOI: 10.1080/07357907.2021.1969574] [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: 12/24/2022]
Abstract
Axillary nodal metastasis is related to poor prognosis in breast cancer (BC). Key candidate genes in BC lymph node metastasis have been identified from Gene Expression Omnibus datasets and explored through functional enrichment database for annotation, visualization and integrated discovery (DAVID) , protein-protein interaction by Search Tool for the Retrieval of Interacting Genes and proteins (STRING), network visualization (Cytoscape), survival analysis (GEPIA, KM Plotter), and target prediction (miRNet). A total of 102 overlapping differentially expressed genes were found. In-silico survival and expression analyses revealed six candidate hub genes, Desmocollin 3 (DSC3), KRT5, KRT6B, KRT17, KRT81, and SERPINB5, to be significantly associated with nodal metastasis and overall survival, and 83 MicroRNA (miRNAs), which may be potential diagnostic markers and therapeutic targets in BC patients.
Collapse
Affiliation(s)
- Anupama Modi
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Ashita Gadwal
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Shweta Ukey
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Dipayan Roy
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Sujoy Fernandes
- Department of Radiation Oncology, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Mithu Banerjee
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| |
Collapse
|
18
|
Hemida AS, Holah NS. Expression of Friend Leukemia Integration-1 (Fli-1) and the Apoptosis Regulator B Cell Lymphoma-2 (BCL-2) in Gastric Carcinoma; an Immunohistochemical Study. J Immunoassay Immunochem 2021; 43:1954948. [PMID: 34314292 DOI: 10.1080/15321819.2021.1954948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Fli-1 regulates multiple biological functions in different cancers particularly Ewing sarcoma and leukemias. There are controversial reports regarding function and prognostic significance of Fli-1 in epithelial cancer including gastric carcinoma (GC). No previous reports examined relationship between Fli-1 and BCL-2 in GC. This study aimed to investigate the expression of Fli-1 and BCL-2 in GC and expected the prognostic significance of their expression. Study was carried out on 88 gastric specimens (58 GC and 30 chronic gastritis cases). Immunohistochemical staining for Fli-1 and BCL-2 was done. There was significant lower Fli-1 and BCL-2 expression in GC than chronic gastritis. Advanced tumor stage showed significant relation with both low Fli-1 expression and negative BCL-2 expression. There was significant direct correlation between BCL-2 positivity and Fli-1 expression in GC. Cox-regression analysis showed that distant metastasis was the first independent factor affecting patients' OS. Fli-1 could act as tumor-suppressor protein involved in GC carcinogenesis. In addition, both Fli-1 and BCL-2 may be used as good prognostic markers in GC. The direct correlation between BCL-2 and Fli-1 expression could potentiate Fli-1 and BCL-2 as therapeutic targets in GC, acting together to inhibit cellular proliferation.
Collapse
Affiliation(s)
- Aiat Shaban Hemida
- Pathology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| | - Nanis Shawky Holah
- Pathology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| |
Collapse
|
19
|
Xu X, Cheng L, Fan Y, Mao W. Tumor Microenvironment-Associated Immune-Related Genes for the Prognosis of Malignant Pleural Mesothelioma. Front Oncol 2020; 10:544789. [PMID: 33042835 PMCID: PMC7526499 DOI: 10.3389/fonc.2020.544789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare but highly aggressive thoracic malignancy. ESTIMATE algorithm-derived immune scores are commonly used to quantify the immune and stromal components in tumors. Thus, this algorithm may help determine the tumor microenvironment (TME)-related gene expression profile associated with tumor immunity. This study aimed at mining public databases to determine a potential correlation between differentially expressed genes (DEGs) and survival in patients with MPM. We categorized patients from the Gene Expression Omnibus database according to their immune/stromal scores into high- and low-score groups. Functional enrichment analysis and the construction of protein-protein interaction networks showed that the DEGs identified were primarily involved in the TME. Furthermore, we validated these genes in an independent cohort of patients with MPM from The Cancer Genome Atlas database. DEG analysis showed that 29 DEGs were cancer driver genes. Subsequently, 14 TME-related genes, which have been previously neglected, were shown to exhibit significant prognostic potential in MPM. In conclusion, immune/stromal scores are novel predictors of a poor prognosis in patients with MPM. We identified DEGs that are involved in immunity against MPM and may contribute to patient survival. Owing to their potential as prognostic factors for MPM, these 14 TME-related genes need to be studied in detail in the future.
Collapse
Affiliation(s)
- Xiaoling Xu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Medical Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lei Cheng
- Department of Thoracic Radiotherapy, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yun Fan
- Department of Medical Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Weimin Mao
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Thoracic Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
20
|
Miao B, Bauer AS, Hufnagel K, Wu Y, Trajkovic-Arsic M, Pirona AC, Giese N, Taipale J, Siveke JT, Hoheisel JD, Lueong S. The transcription factor FLI1 promotes cancer progression by affecting cell cycle regulation. Int J Cancer 2020; 147:189-201. [PMID: 31846072 DOI: 10.1002/ijc.32831] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/15/2019] [Accepted: 11/29/2019] [Indexed: 12/30/2022]
Abstract
Binding of transcription factors to mutated DNA sequences is a likely regulator of cancer progression. Noncoding regulatory mutations such as those on the core promoter of the gene encoding human telomerase reverse transcriptase have been shown to affect gene expression in cancer. Using a protein microarray of 667 transcription factor DNA-binding domains and subsequent functional assays, we looked for transcription factors that preferentially bind the mutant hTERT promoter and characterized their downstream effects. One of them, friend leukemia integration 1 (FLI1), which belongs to the E26 transforming-specific family of transcription factors, exhibited particularly strong effects with respect to regulating hTERT expression, while the even better binding ELK3 did not. Depletion of FLI1 decreased expression of the genes for cyclin D1 (CCND1) and E2F transcription factor 2 (E2F2) resulting in a G1/S cell cycle arrest and in consequence a reduction of cell proliferation. FLI1 also affected CMTM7, another gene involved in G1/S transition, although by another process that suggests a balanced regulation of the tumor suppressor gene's activity via opposing regulation processes. FLI1 expression was found upregulated and correlated with an increase in CCND1 expression in pancreatic cancer and brain tumors. In non-neoplastic lung cells, however, FLI1 depletion led to rapid progression through the cell cycle. This coincides with the fact that FLI1 is downregulated in lung tumors. Taken together, our data indicate a cell cycle regulatory hub involving FLI1, hTERT, CCND1 and E2F2 in a tissue- and context-dependent manner.
Collapse
Affiliation(s)
- Beiping Miao
- Division of Functional Genome Analysis (B070), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Andrea S Bauer
- Division of Functional Genome Analysis (B070), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katrin Hufnagel
- Infections and Cancer Epidemiology (F022), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yenan Wu
- Division of Functional Genome Analysis (B070), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marija Trajkovic-Arsic
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna C Pirona
- Division of Functional Genome Analysis (B070), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nathalia Giese
- Research Laboratory of the European Pancreas Centre (EPZ) Integrative Oncology Group, University Clinic Heidelberg, Heidelberg, Germany
| | - Jussi Taipale
- Division of Functional Genomics, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Sweden
| | - Jens T Siveke
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg D Hoheisel
- Division of Functional Genome Analysis (B070), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Smiths Lueong
- Division of Functional Genome Analysis (B070), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
21
|
Lumefantrine, an antimalarial drug, reverses radiation and temozolomide resistance in glioblastoma. Proc Natl Acad Sci U S A 2020; 117:12324-12331. [PMID: 32409605 DOI: 10.1073/pnas.1921531117] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive cancer without currently effective therapies. Radiation and temozolomide (radio/TMZ) resistance are major contributors to cancer recurrence and failed GBM therapy. Heat shock proteins (HSPs), through regulation of extracellular matrix (ECM) remodeling and epithelial mesenchymal transition (EMT), provide mechanistic pathways contributing to the development of GBM and radio/TMZ-resistant GBM. The Friend leukemia integration 1 (Fli-1) signaling network has been implicated in oncogenesis in GBM, making it an appealing target for advancing novel therapeutics. Fli-1 is linked to oncogenic transformation with up-regulation in radio/TMZ-resistant GBM, transcriptionally regulating HSPB1. This link led us to search for targeted molecules that inhibit Fli-1. Expression screening for Fli-1 inhibitors identified lumefantrine, an antimalarial drug, as a probable Fli-1 inhibitor. Docking and isothermal calorimetry titration confirmed interaction between lumefantrine and Fli-1. Lumefantrine promoted growth suppression and apoptosis in vitro in parental and radio/TMZ-resistant GBM and inhibited tumor growth without toxicity in vivo in U87MG GBM and radio/TMZ-resistant GBM orthotopic tumor models. These data reveal that lumefantrine, an FDA-approved drug, represents a potential GBM therapeutic that functions through inhibition of the Fli-1/HSPB1/EMT/ECM remodeling protein networks.
Collapse
|
22
|
Wang S, Wang Y, Yu C, Cao Y, Yu Y, Pan Y, Su D, Lu Q, Yang W, Zuo Y, Yang L. Characterization of the relationship between FLI1 and immune infiltrate level in tumour immune microenvironment for breast cancer. J Cell Mol Med 2020; 24:5501-5514. [PMID: 32249526 PMCID: PMC7214163 DOI: 10.1111/jcmm.15205] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/31/2020] [Accepted: 03/06/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common cancer and the leading cause of cancer death among women in the world. Tumour‐infiltrating lymphocytes were defined as the white blood cells left in the vasculature and localized in tumours. Recently, tumour‐infiltrating lymphocytes were found to be associated with good prognosis and response to immunotherapy in tumours. In this study, to examine the influence of FLI1 in immune system in breast cancer, we interrogated the relationship between the FLI1 expression levels with infiltration levels of 28 immune cell types. By splitting the breast cancer samples into high and low expression FLI1 subtypes, we found that the high expression FLI1 subtype was enriched in many immune cell types, and the up‐regulated differentially expressed genes between them were enriched in immune system processes, immune‐related KEGG pathways and biological processes. In addition, many important immune‐related features were found to be positively correlated with the FLI1 expression level. Furthermore, we found that the FLI1 was correlated with the immune‐related genes. Our findings may provide useful help for recognizing the relationship between tumour immune microenvironment and FLI1, and may unravel clinical outcomes and immunotherapy utility for FLI1 in breast cancer.
Collapse
Affiliation(s)
- Shiyuan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yakun Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Chunlu Yu
- Public Health College, Harbin Medical University, Harbin, China
| | - Yiyin Cao
- Public Health College, Harbin Medical University, Harbin, China
| | - Yao Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yi Pan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Dongqing Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Qianzi Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wuritu Yang
- The State key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yongchun Zuo
- The State key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Lei Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
23
|
Rajesh Y, Biswas A, Banik P, Pal I, Das S, Borkar SA, Sardana H, Saha A, Das SK, Emdad L, Fisher PB, Mandal M. Transcriptional regulation of HSPB1 by Friend leukemia integration-1 factor modulates radiation and temozolomide resistance in glioblastoma. Oncotarget 2020; 11:1097-1108. [PMID: 32284788 PMCID: PMC7138161 DOI: 10.18632/oncotarget.27425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 12/21/2019] [Indexed: 02/02/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor and is invariably fatal. Heat shock proteins (HSPs) provide protein signatures/biomarkers for GBM that afford potential as targets for developing anti-GBM drugs. In GBM, elevated expression of hypoxia inducible factors under the influence of Ets family proteins significantly promotes the expression of HSPs. RNAseq analysis identified HSPB1 as a prominent upregulated HSP in GBM and in radiation resistant/temozolomide resistant (radio/TMZR) GBM. Here, we established friend leukemia integration 1 (Fli-1), a member of Ets family to be playing a transcriptional regulatory role on the HSPB1 gene. Fli-1 binds to nucleotide residues GGAA at binding sites 3, 6 and 7 in the 5-kb upstream region of HSPB1. Fli-1 has been linked to oncogenic transformation with upregulation in radio/TMZR GBM. Overexpression of Fli-1 in GBM promotes resistance, whereas Fli-1 knockdown in radio/TMZR GBM cells suppresses resistance. We identify the underlying molecular mechanisms of Fli-1-mediated regulation of HSPB1 that drive extracellular matrix remodeling and epithelial to mesenchymal transition in radio/TMZR GBM cells. This study uncovers Fli-1 as a potential therapeutic target for combating radiation and temozolomide resistance in GBM.
Collapse
Affiliation(s)
- Yetirajam Rajesh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Angana Biswas
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Payel Banik
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Ipsita Pal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Sachin A Borkar
- Department of Neurosurgery & Gamma Knife, All India Institute of Medical Sciences, New Delhi, India
| | - Hardik Sardana
- Department of Neurosurgery & Gamma Knife, All India Institute of Medical Sciences, New Delhi, India
| | - Abhijit Saha
- Radiation Department, UGC DAE Consortium, Jadavpur University, Kolkata, India
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.,VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.,VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.,VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.,VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.,VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.,VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.,Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.,VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| |
Collapse
|
24
|
Ma Y, Xu B, Yu J, Huang L, Zeng X, Shen X, Ren C, Ben-David Y, Luo H. Fli-1 Activation through Targeted Promoter Activity Regulation Using a Novel 3', 5'-diprenylated Chalcone Inhibits Growth and Metastasis of Prostate Cancer Cells. Int J Mol Sci 2020; 21:2216. [PMID: 32210104 PMCID: PMC7139342 DOI: 10.3390/ijms21062216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
The friend leukemia integration 1 (Fli-1) gene is involved in the expression control of key genes in multiple pathogenic/physiological processes, including cell growth, differentiation, and apoptosis; this implies that Fli-1 is a strong candidate for drug development. In our previous study, a 3',5'-diprenylated chalcone, (E)-1-(2-hydroxy-4-methoxy-3,5-diprenyl) phenyl-3-(3-pyridinyl)-propene-1-one (C10), was identified as a novel anti-prostate cancer (PCa) agent. Here, we investigated the molecular mechanisms underlying the anti-cancer effects of C10 on the growth, metastasis, and invasion of PC3 cells in vitro. Our results show that C10 exhibited a strong inhibitory effect on proliferation and metastasis of PC3 cells via several cellular and flow cytometric analyses. Further mechanism studies revealed that C10 likely serves as an Fli-1 agonist for regulating the expression of Fli-1 target genes including phosphatidylinositol 3-kinase (P110), murine double minute2 (MDM2), B-cell lymphoma-2 (Bcl-2), Src homology-2 domain-containing inositol 5-phosphatase 1 (SHIP-1), and globin transcription factor-1 (Gata-1) as well as the phosphorylation of extracellular-regulated protein kinases 1 (ERK1). Further, we confirmed that C10 can regulate the expressions of vascular endothelial growth factor 1 (VEGF-1), transforming growth factor-β2 (TGF-β2), intercellular cell adhesion molecule-1 (ICAM-1), p53, and matrix metalloproteinase 1 (MMP-1) genes associated with tumor apoptosis, migration, and invasion. Thus, C10 exhibits stronger anticancer activity with novel molecular targets and regulatory molecular mechanisms, indicating its great potency for development as a novel targeted anticancer drug.
Collapse
Affiliation(s)
- Youfen Ma
- State key laboratory of functions and applications of medicinal plants, Guizhou medical university, Guiyang 550014, China; (Y.M.); (B.X.); (J.Y.); (X.Z.); (X.S.)
- College of pharmacy, Guizhou Medical University, Guiyang 550029, China
| | - Bixue Xu
- State key laboratory of functions and applications of medicinal plants, Guizhou medical university, Guiyang 550014, China; (Y.M.); (B.X.); (J.Y.); (X.Z.); (X.S.)
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Science, Guiyang 550014, China
| | - Jia Yu
- State key laboratory of functions and applications of medicinal plants, Guizhou medical university, Guiyang 550014, China; (Y.M.); (B.X.); (J.Y.); (X.Z.); (X.S.)
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Science, Guiyang 550014, China
| | - Lirong Huang
- College of Food and Pharmaceutical Engineering, Guizhou Institute of Technology, Guiyang 550003, China;
| | - Xiaoping Zeng
- State key laboratory of functions and applications of medicinal plants, Guizhou medical university, Guiyang 550014, China; (Y.M.); (B.X.); (J.Y.); (X.Z.); (X.S.)
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Science, Guiyang 550014, China
| | - Xiangchun Shen
- State key laboratory of functions and applications of medicinal plants, Guizhou medical university, Guiyang 550014, China; (Y.M.); (B.X.); (J.Y.); (X.Z.); (X.S.)
- College of pharmacy, Guizhou Medical University, Guiyang 550029, China
| | - Chunyan Ren
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Yaacov Ben-David
- State key laboratory of functions and applications of medicinal plants, Guizhou medical university, Guiyang 550014, China; (Y.M.); (B.X.); (J.Y.); (X.Z.); (X.S.)
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Science, Guiyang 550014, China
| | - Heng Luo
- State key laboratory of functions and applications of medicinal plants, Guizhou medical university, Guiyang 550014, China; (Y.M.); (B.X.); (J.Y.); (X.Z.); (X.S.)
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Science, Guiyang 550014, China
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
25
|
Identifying FL11 subtype by characterizing tumor immune microenvironment in prostate adenocarcinoma via Chou's 5-steps rule. Genomics 2020; 112:1500-1515. [DOI: 10.1016/j.ygeno.2019.08.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/03/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022]
|
26
|
Del Portillo A, Komissarova EV, Bokhari A, Hills C, de Gonzalez AK, Kongkarnka S, Remotti HE, Sepulveda JL, Sepulveda AR. Downregulation of Friend Leukemia Integration 1 ( FLI1) follows the stepwise progression to gastric adenocarcinoma. Oncotarget 2019; 10:3852-3864. [PMID: 31231464 PMCID: PMC6570468 DOI: 10.18632/oncotarget.26974] [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: 11/29/2018] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
Gastric adenocarcinoma (GC) is a leading cause of cancer-related deaths worldwide. The transcription factor gene Friend Leukemia Integration 1 (FLI1) is methylated and downregulated in human GC tissues. Using human GC samples, we determined which cells downregulate FLI1, when FLI1 downregulation occurs, if FLI1 downregulation correlates with clinical-pathologic characteristics, and whether FLI1 plays a role in invasion and/or proliferation of cultured cells. We analyzed stomach tissues from 98 patients [8 normal mucosa, 8 intestinal metaplasia (IM), 7 dysplasia, 91 GC] by immunohistochemistry for FLI1. Epithelial cells from normal, IM, and low-grade dysplasia (LGD) showed strong nuclear FLI1 staining. GC epithelial cells showed significantly less nuclear FLI1 staining as compared to normal epithelium, IM and LGD (P=1.2×10-5, P=1.4×10-6 and P=0.006, respectively). FLI1 expression did not correlate with tumor stage or differentiation, but was associated with patient survival, depending on tumor differentiation. We tested the functional role of FLI1 by assaying proliferation and invasion in cultured GC cells. Lentiviral-transduced FLI1 overexpression in GC AGS cells inhibited invasion by 73.5% (P = 0.001) and proliferation by 31.5% (P = 0.002), as compared to controls. Our results support a combined role for FLI1 as a suppressor of invasiveness and proliferation in gastric adenocarcinoma, specifically in the transition from pre-cancer lesions and dysplasia to invasive adenocarcinoma, and suggest that FLI1 may be a prognostic biomarker of survival in gastric cancers.
Collapse
Affiliation(s)
- Armando Del Portillo
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Elena V Komissarova
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Aqiba Bokhari
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Caitlin Hills
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne Koehne de Gonzalez
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarawut Kongkarnka
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Helen E Remotti
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jorge L Sepulveda
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Antonia R Sepulveda
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
27
|
Yan X, Yu Y, Li L, Chen N, Song W, He H, Dong J, Liu X, Cui J. Friend leukemia virus integration 1 is a predictor of poor prognosis of breast cancer and promotes metastasis and cancer stem cell properties of breast cancer cells. Cancer Med 2018; 7:3548-3560. [PMID: 29869379 PMCID: PMC6089157 DOI: 10.1002/cam4.1589] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/22/2022] Open
Abstract
Breast cancer is the most common cancer in women worldwide; despite the developments in diagnosis and therapy, recurrence and metastasis remain the main causes of death among patients with breast cancer. This study aimed to identify a promising biomarker for this disease. The study clarified (1) the association between Friend leukemia virus integration 1 (FLI-1) and various molecular subtypes and (2) the prognostic value of FLI-1 in breast cancer. To the best of our knowledge, this study is the first to report that FLI-1 is a predictor of poor prognosis in patients with breast cancer and overexpressed in the triple negative breast cancer (TNBC) subtype. To further verify the effect of FLI-1 in promoting the metastasis of TNBC, we performed a series of functional experiments in vitro and orthotopic xenograft experiments in the mammary fat pad of nude mice. FLI-1, as a transcription factor, bound to the promoters of key EMT-related genes (CDH1 and VIM), and regulated their expressions at the transcriptional level, thus induced epithelial-mesenchymal transition (EMT). The overexpression of FLI-1 significantly upregulated the expression of mesenchymal markers. After the modulation of FLI-1, the changes in mammary stem cell markers (ALDH1A1 and CD133) and the capacity to form mammospheres were consistent with those of the EMT-related markers. The orthotopic xenograft models further confirmed that the attenuation of stem cell traits after silencing FLI-1 decreased the ability of tumorigenesis. These results indicate that FLI-1 is a useful predictor of poor prognosis in patients with breast cancer. Furthermore, the preliminary exploration of metastatic mechanism in the patients with TNBC will provide a potential target to treat breast cancer in the near future.
Collapse
Affiliation(s)
- Xu Yan
- Pathological Diagnostic Center, The First Hospital of Jilin University, Changchun, Jilin, China.,Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Yu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lingyu Li
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Naifei Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Song
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hua He
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jie Dong
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiangliang Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
28
|
Tsai HP, Tsai TH, Hsieh YJ, Chen YT, Lee CL, Tsai YC, She TC, Lin CL, Chai CY, Kwan AL. Overexpression of Fli-1 in astrocytoma is associated with poor prognosis. Oncotarget 2018; 8:29174-29186. [PMID: 28418872 PMCID: PMC5438722 DOI: 10.18632/oncotarget.16303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 02/20/2017] [Indexed: 02/07/2023] Open
Abstract
Background Astrocytoma, a common and highly malignant type of brain tumor, is associated with poor overall survival despite advances in surgical treatment, radiotherapy, and chemotherapy. The nuclear transcription factor Fli-1 has been shown to increase cellular proliferation and tumorigenesis in many types of cancer; however, previous reports have not described a correlation between clinical outcomes and Fli-1 in astrocytoma patients. The present study aimed to elucidate the clinical role of Fli-1 in astrocytoma. Results High-level of Fli-1 protein expression was significantly association with World Health Organization (WHO) high grade and poor prognosis. A multivariate analysis revealed that the WHO grade and Fli-1 protein expression were independent factor of prognostic factors of patients with astrocytoma. In addition, Fli-1 silencing inhibited proliferation, migration, and invasion and led to the downregulation of Ki-67, VEGF, and cyclin D1 expression in the astrocytoma cells. Materials and methods Fli-1 protein expression in astrocytoma tissue samples were detected via immunohistochemistry, and potential correlations between clinical parameters and Fli-1 expression were assessed in patients with astrocytoma. Additionally, proliferation, invasion, and migration assays of astrocytoma cell lines were conducted to evaluate the effects of short interfering RNA (siRNA) on these processes; in addition, these cells were subjected to western blotting to detect the expression levels of Fli-1, Ki-67, VEGF, and Cyclin D1. Conclusion Fli-1 shows promise as a potential prognostic biomarker and therapeutic molecular target for astrocytoma patients.
Collapse
Affiliation(s)
- Hung-Pei Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tai-Hsin Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ya-Ju Hsieh
- Department of Medical Imaging and Radiological Sciences, Kaohsiung, Taiwan
| | - Yi-Ting Chen
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Ling Lee
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yi-Cheng Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ting-Chang She
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Lung Lin
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
29
|
Liu T, Yao Y, Zhang G, Wang Y, Deng B, Song J, Li X, Han F, Xiao X, Yang J, Xia L, Li YJ, Plachynta M, Zhang M, Yan C, Mu S, Luo H, Zacksenhaus E, Hao X, Ben-David Y. A screen for Fli-1 transcriptional modulators identifies PKC agonists that induce erythroid to megakaryocytic differentiation and suppress leukemogenesis. Oncotarget 2017; 8:16728-16743. [PMID: 28052010 PMCID: PMC5369997 DOI: 10.18632/oncotarget.14377] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/07/2016] [Indexed: 11/25/2022] Open
Abstract
The ETS-related transcription factor Fli-1 affects many developmental programs including erythroid and megakaryocytic differentiation, and is frequently de-regulated in cancer. Fli-1 was initially isolated following retrovirus insertional mutagenesis screens for leukemic initiator genes, and accordingly, inhibition of this transcription factor can suppress leukemia through induction of erythroid differentiation. To search for modulators of Fli-1, we hereby performed repurposing drug screens with compounds isolated from Chinese medicinal plants. We identified agents that can transcriptionally activate or inhibit a Fli-1 reporter. Remarkably, agents that increased Fli-1 transcriptional activity conferred a strong anti-cancer activity upon Fli-1-expressing leukemic cells in culture. As opposed to drugs that suppress Fli1 activity and lead to erythroid differentiation, growth suppression by these new Fli-1 transactivating compounds involved erythroid to megakaryocytic conversion (EMC). The identified compounds are structurally related to diterpene family of small molecules, which are known agonists of protein kinase C (PKC). In accordance, these PKC agonists (PKCAs) induced PKC phosphorylation leading to activation of the mitogen-activated protein kinase (MAPK) pathway, increased cell attachment and EMC, whereas pharmacological inhibition of PKC or MAPK diminished the effect of our PKCAs. Moreover, in a mouse model of leukemia initiated by Fli-1 activation, the PKCA compounds exhibited strong anti-cancer activity, which was accompanied by increased presence of CD41/CD61 positive megakaryocytic cells in leukemic spleens. Thus, PKC agonists offer a novel approach to combat Fli-1-induced leukemia, and possibly other cancers,by inducing EMC in part through over-activation of the PKC-MAPK-Fli-1 pathway.
Collapse
Affiliation(s)
- Tangjingjun Liu
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Yao Yao
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Gang Zhang
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China
| | - Ye Wang
- College of Ecology, Lishui University, Zhejiang, China
| | - Bin Deng
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China
| | - Jialei Song
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,The Laboratory of Cell Biochemistry and Topogenic Regulation, College of Bioengineering and Faculty of Sciences, Chongqing University, Chongqing, China
| | - Xiaogang Li
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China
| | - Fei Han
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China
| | - Xiao Xiao
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Jue Yang
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China
| | - Lei Xia
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,School of Pharmaceutical Sciences, Guizhou University, Guizhou, China
| | - You-Jun Li
- Department of Anatomy, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Maksym Plachynta
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China
| | - Mu Zhang
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China
| | - Chen Yan
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China
| | - Shuzhen Mu
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Heng Luo
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Eldad Zacksenhaus
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Division of Advanced Diagnostics, Toronto General Research Institute-University Health Network, Toronto, Ontario, Canada
| | - Xiaojiang Hao
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,School of Pharmaceutical Sciences, Guizhou University, Guizhou, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Yaacov Ben-David
- Department of Biology and Chemistry, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guizhou, China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| |
Collapse
|
30
|
Sizemore GM, Pitarresi JR, Balakrishnan S, Ostrowski MC. The ETS family of oncogenic transcription factors in solid tumours. Nat Rev Cancer 2017; 17:337-351. [PMID: 28450705 DOI: 10.1038/nrc.2017.20] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Findings over the past decade have identified aberrant activation of the ETS transcription factor family throughout all stages of tumorigenesis. Specifically in solid tumours, gene rearrangement and amplification, feed-forward growth factor signalling loops, formation of gain-of-function co-regulatory complexes and novel cis-acting mutations in ETS target gene promoters can result in increased ETS activity. In turn, pro-oncogenic ETS signalling enhances tumorigenesis through a broad mechanistic toolbox that includes lineage specification and self-renewal, DNA damage and genome instability, epigenetics and metabolism. This Review discusses these different mechanisms of ETS activation and subsequent oncogenic implications, as well as the clinical utility of ETS factors.
Collapse
Affiliation(s)
- Gina M Sizemore
- The Comprehensive Cancer Center, The Ohio State University
- Department of Cancer Biology and Genetics, The Ohio State University, 598 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, Ohio 43210, USA
| | - Jason R Pitarresi
- The Comprehensive Cancer Center, The Ohio State University
- Department of Cancer Biology and Genetics, The Ohio State University, 598 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, Ohio 43210, USA
| | - Subhasree Balakrishnan
- The Comprehensive Cancer Center, The Ohio State University
- Department of Cancer Biology and Genetics, The Ohio State University, 598 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, Ohio 43210, USA
| | - Michael C Ostrowski
- The Comprehensive Cancer Center, The Ohio State University
- Department of Cancer Biology and Genetics, The Ohio State University, 598 Biomedical Research Tower, 460 W. 12th Avenue, Columbus, Ohio 43210, USA
| |
Collapse
|
31
|
Takahashi T, Asano Y, Sugawara K, Yamashita T, Nakamura K, Saigusa R, Ichimura Y, Toyama T, Taniguchi T, Akamata K, Noda S, Yoshizaki A, Tsuruta D, Trojanowska M, Sato S. Epithelial Fli1 deficiency drives systemic autoimmunity and fibrosis: Possible roles in scleroderma. J Exp Med 2017; 214:1129-1151. [PMID: 28232470 PMCID: PMC5379967 DOI: 10.1084/jem.20160247] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 11/08/2016] [Accepted: 01/17/2017] [Indexed: 01/06/2023] Open
Abstract
Systemic sclerosis (SSc), or scleroderma, is a multisystem autoimmune disorder characterized by vasculopathy and fibrosis in the skin and internal organs, most frequently in the esophagus and lungs. Hitherto, studies on SSc pathogenesis centered on immune cells, vascular cells, and fibroblasts. Although dysregulated keratinocytes in SSc have been recently reported, the contribution of epithelial cells to pathogenesis remains unexplored. In this study, we demonstrated the induction of SSc-like molecular phenotype in keratinocytes by gene silencing of transcription factor Friend leukemia virus integration 1 (Fli1), the deficiency of which is implicated in SSc pathogenesis. Keratin 14-expressing epithelial cell-specific Fli1 knockout mice spontaneously developed dermal and esophageal fibrosis with epithelial activation. Furthermore, they developed remarkable autoimmunity with interstitial lung disease derived from thymic defects with down-regulation of autoimmune regulator (Aire). Importantly, Fli1 directly regulated Aire expression in epithelial cells. Collectively, epithelial Fli1 deficiency might be involved in the systemic autoimmunity and selective organ fibrosis in SSc. This study uncovers unidentified roles of dysregulated epithelial cells in SSc pathogenesis.
Collapse
Affiliation(s)
- Takehiro Takahashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Koji Sugawara
- Department of Dermatology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka 545-8585, Japan
| | - Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kouki Nakamura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ryosuke Saigusa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yohei Ichimura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tetsuo Toyama
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takashi Taniguchi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kaname Akamata
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Shinji Noda
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Daisuke Tsuruta
- Department of Dermatology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka 545-8585, Japan
| | - Maria Trojanowska
- Arthritis Center, Rheumatology, Boston University School of Medicine, Boston, MA 02118
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
32
|
Zacksenhaus E, Liu J, Jiang Z, Yao Y, Xia L, Shrestha M, Ben-David Y. Transcription Factors in Breast Cancer—Lessons From Recent Genomic Analyses and Therapeutic Implications. CHROMATIN PROTEINS AND TRANSCRIPTION FACTORS AS THERAPEUTIC TARGETS 2017; 107:223-273. [DOI: 10.1016/bs.apcsb.2016.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Lee E, Moon A. Identification of Biomarkers for Breast Cancer Using Databases. J Cancer Prev 2016; 21:235-242. [PMID: 28053957 PMCID: PMC5207607 DOI: 10.15430/jcp.2016.21.4.235] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is one of the major causes of cancer death in women. Many studies have sought to identify specific molecules involved in breast cancer and understand their characteristics. Many biomarkers which are easily measurable, dependable, and inexpensive, with a high sensitivity and specificity have been identified. The rapidly increasing technology development and availability of epigenetic informations play critical roles in cancer. The accumulated data have been collected, stored, and analyzed in various types of databases. It is important to acknowledge useful and available data and retrieve them from databases. Nowadays, many researches utilize the databases, including The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), Surveillance, Epidemiology and End Results (SEER), and Embase, to find useful informations on biomarkers for breast cancer. This review summarizes the current databases which have been utilized for identification of biomarkers for breast cancer. The information provided by this review would be beneficial to seeking appropriate strategies for diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Eunhye Lee
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Aree Moon
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul, Korea
| |
Collapse
|
34
|
Jung HH, Lee SH, Kim JY, Ahn JS, Park YH, Im YH. Statins affect ETS1-overexpressing triple-negative breast cancer cells by restoring DUSP4 deficiency. Sci Rep 2016; 6:33035. [PMID: 27604655 PMCID: PMC5015082 DOI: 10.1038/srep33035] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 08/17/2016] [Indexed: 12/13/2022] Open
Abstract
We investigated the molecular mechanisms underlying statin-induced growth suppression of triple-negative breast cancer (TNBC) that overexpress the transcription factor ets proto-oncogene 1(ets-1) and downregulate dual specific protein phosphatase 4(dusp4) expression. We examined the gene expression of BC cell lines using the nCounter expression assay, MTT viability assay, cell proliferation assay and Western blot to evaluate the effects of simvastatin. Finally, we performed cell viability testing in TNBC cell line-transfected DUSP4. We demonstrated that ETS1 mRNA and protein were overexpressed in TNBC cells compared with other BC cell lines (P = <0.001) and DUSP4 mRNA was downregulated (P = <0.001). MTT viability assay showed that simvastatin had significant antitumor activity (P = 0.002 in 0.1 μM). In addition, simvastatin could restore dusp4 deficiency and suppress ets-1 expression in TNBC. Lastly, we found that si-DUSP4 RNA transfection overcame the antitumor activity of statins. MAPK pathway inhibitor, U0126 and PI3KCA inhibitor LY294002 also decreased levels of ets-1, phosphor-ERK and phosphor-AKT on Western blot assay. Accordingly, our study indicates that simvastatin potentially affects the activity of transcriptional factors such as ets-1 and dusp4 through the MAPK pathway. In conclusion, statins might be potential candidates for TNBC therapy reducing ets-1 expression via overexpression of dusp4.
Collapse
Affiliation(s)
- Hae Hyun Jung
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Soo-Hyeon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Ji-Yeon Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Yeon Hee Park
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.,Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.,Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea
| | - Young-Hyuck Im
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.,Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
35
|
Tsui KH, Lin YH, Chung LC, Chuang ST, Feng TH, Chiang KC, Chang PL, Yeh CJ, Juang HH. Prostate-derived ets factor represses tumorigenesis and modulates epithelial-to-mesenchymal transition in bladder carcinoma cells. Cancer Lett 2016; 375:142-151. [PMID: 26965996 DOI: 10.1016/j.canlet.2016.02.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/27/2016] [Accepted: 02/29/2016] [Indexed: 12/27/2022]
Abstract
Prostate-derived Ets (E-twenty six) factor (PDEF), an epithelium-specific member of the Ets family of transcription factors, has been shown to play a role in suppressing the development of many epithelium-derived cancers such as prostate and breast cancer. It is not clear, however, whether PDEF is involved in the development or progression of bladder cancer. In a comparison between normal urothelium and bladder tumor tissue, we identified significant decreases of PDEF in the tumor tissue. Further, the immunohistochemistry assays indicated a significantly higher immunostaining of PDEF in low-grade bladder tumors. Additionally, the highly differentiated transitional-cell bladder carcinoma RT-4 cells expressed significantly more PDEF levels than the bladder carcinoma HT1376 and the T24 cells. Ectopic overexpression of PDEF attenuated proliferation, invasion, and tumorigenesis of bladder carcinoma cells in vitro and in vivo. PDEF enhanced the expression levels of mammary serine protease inhibitor (MASPIN), N-myc downstream regulated gene 1 (NDRG1), KAI1, and B-cell translocation gene 2 (BTG2). PDEF modulated epithelial-mesenchymal-transition (EMT) by upregulating E-cadherin expression and downregulating the expression of N-cadherin, SNAIL, SLUG, and vimentin, leading to lower migration and invasion abilities of bladder carcinoma cells. Filamentous actin (F-actin) polarization and remodeling were observed in PDEF-knockdown RT-4 cells. Our results suggest that PDEF gene expression is associated with the extent of bladder neoplasia and PDEF modulated the expressions of EMT-related genes. The induction of BTG2, NDRG1, MASPIN, and KAI1 gene expressions by PDEF may explain the inhibitory functions of PDEF on the proliferation, invasion, and tumorigenesis in bladder carcinoma cells.
Collapse
Affiliation(s)
- Ke-Hung Tsui
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan; Department of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Li-Chuan Chung
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Sung-Ting Chuang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Kun-Chun Chiang
- Zebafish Center, General Surgery Department, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Phei-Lang Chang
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan; Department of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Chi-Ju Yeh
- Department of Pathology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan; Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan.
| |
Collapse
|
36
|
High-definition CpG methylation of novel genes in gastric carcinogenesis identified by next-generation sequencing. Mod Pathol 2016; 29:182-93. [PMID: 26769141 DOI: 10.1038/modpathol.2015.144] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/01/2015] [Indexed: 12/12/2022]
Abstract
Gastric cancers are the most frequent gastric malignancy and usually arise in the sequence of Helicobacter pylori-associated chronic gastritis. CpG methylation is a central mechanism of epigenetic gene regulation affecting cancer-related genes, and occurs early in gastric carcinogenesis. DNA samples from non-metaplastic gastric mucosa with variable levels of gastritis (non-metaplastic mucosa), intestinal metaplasia, or gastric cancer were screened with methylation arrays for CpG methylation of cancer-related genes and 30 gene targets were further characterized by high-definition bisulfite next-generation sequencing. In addition, data from The Cancer Genome Atlas were analyzed for correlation of methylation with gene expression. Overall, 13 genes had significantly increased CpG methylation in gastric cancer vs non-metaplastic mucosa (BRINP1, CDH11, CHFR, EPHA5, EPHA7, FGF2, FLI1, GALR1, HS3ST2, PDGFRA, SEZ6L, SGCE, and SNRPN). Further, most of these genes had corresponding reduced expression levels in gastric cancer compared with intestinal metaplasia, including novel hypermethylated genes in gastric cancer (FLI1, GALR1, SGCE, and SNRPN), suggesting that they may regulate neoplastic transformation from non-malignant intestinal metaplasia to cancer. Our data suggest a tumor-suppressor role for FLI1 in gastric cancer, consistent with recently reported data in breast cancer. For the genes with strongest methylation/expression correlation, namely FLI1, the expression was lowest in microsatellite-unstable tumors compared with other gastric cancer molecular subtypes. Importantly, reduced expression of hypermethylated BRINP1 and SGCE was significantly associated with favorable survival in gastric cancer. In summary, we report novel methylation gene targets that may have functional roles in discrete stages of gastric carcinogenesis and may serve as biomarkers for diagnosis and prognosis of gastric cancer.
Collapse
|
37
|
Hou C, Tsodikov OV. Structural Basis for Dimerization and DNA Binding of Transcription Factor FLI1. Biochemistry 2015; 54:7365-74. [PMID: 26618620 DOI: 10.1021/acs.biochem.5b01121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
FLI1 (Friend leukemia integration 1) is a metazoan transcription factor that is upregulated in a number of cancers. In addition, rearrangements of the fli1 gene cause sarcomas, leukemias, and lymphomas. These rearrangements encode oncogenic transcription factors, in which the DNA binding domain (DBD or ETS domain) of FLI1 on the C-terminal side is fused to a part of an another protein on the N-terminal side. Such abnormal cancer cell-specific fusions retain the DNA binding properties of FLI1 and acquire non-native protein-protein or protein-nucleic acid interactions of the substituted region. As a result, these fusions trigger oncogenic transcriptional reprogramming of the host cell. Interactions of FLI1 fusions with other proteins and with itself play a critical role in the oncogenic regulatory functions, and they are currently under intense scrutiny, mechanistically and as potential novel anticancer drug targets. We report elusive crystal structures of the FLI1 DBD, alone and in complex with cognate DNA containing a GGAA recognition sequence. Both structures reveal a previously unrecognized dimer of this domain, consistent with its dimerization in solution. The homodimerization interface is helix-swapped and dominated by hydrophobic interactions, including those between two interlocking Phe362 residues. A mutation of Phe362 to an alanine disrupted the propensity of this domain to dimerize without perturbing its structure or the DNA binding function, consistent with the structural observations. We propose that FLI1 DBD dimerization plays a role in transcriptional activation and repression by FLI1 and its fusions at promoters containing multiple FLI1 binding sites.
Collapse
Affiliation(s)
- Caixia Hou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky , 789 South Limestone Street, Lexington, Kentucky 40536-0596, United States
| | - Oleg V Tsodikov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky , 789 South Limestone Street, Lexington, Kentucky 40536-0596, United States
| |
Collapse
|
38
|
Tang SW, Bilke S, Cao L, Murai J, Sousa FG, Yamade M, Rajapakse V, Varma S, Helman LJ, Khan J, Meltzer PS, Pommier Y. SLFN11 Is a Transcriptional Target of EWS-FLI1 and a Determinant of Drug Response in Ewing Sarcoma. Clin Cancer Res 2015; 21:4184-93. [PMID: 25779942 DOI: 10.1158/1078-0432.ccr-14-2112] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 03/04/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE SLFN11 was identified as a critical determinant of response to DNA-targeted therapies by analyzing gene expression and drug sensitivity of NCI-60 and CCLE datasets. However, how SLFN11 is regulated in cancer cells remained unknown. Ewing sarcoma, which is characterized by the chimeric transcription factor EWS-FLI1, has notably high SLFN11 expression, leading us to investigate whether EWS-FLI1 drives SLFN11 expression and the role of SLFN11 in the drug response of Ewing sarcoma cells. EXPERIMENTAL DESIGN Binding sites of EWS-FLI1 on the SLFN11 promoter were analyzed by chromatin immunoprecipitation sequencing and promoter-luciferase reporter analyses. The relationship between SLFN11 and EWS-FLI1 were further examined in EWS-FLI1-knockdown or -overexpressing cells and in clinical tumor samples. RESULTS EWS-FLI1 binds near the transcription start site of SLFN11 promoter and acts as a positive regulator of SLFN11 expression in Ewing sarcoma cells. EWS-FLI1-mediated SLFN11 expression is responsible for high sensitivity of Ewing sarcoma to camptothecin and combinations of PARP inhibitors with temozolomide. Importantly, Ewing sarcoma patients with higher SLFN11 expression showed better tumor-free survival rate. The correlated expression between SLFN11 and FLI1 extends to leukemia, pediatric, colon, breast, and prostate cancers. In addition, expression of other ETS members correlates with SLFN11 in NCI-60 and CCLE datasets, and molecular experiments demonstrate that ETS1 acts as a positive regulator for SLFN11 expression in breast cancer cells. CONCLUSIONS Our results imply the emerging relevance of SLFN11 as an ETS transcription factor response gene and for therapeutic response to topoisomerase I inhibitors and temozolomide-PARP inhibitor combinations in ETS-activated cancers.
Collapse
Affiliation(s)
- Sai-Wen Tang
- Laboratory of Molecular Pharmacology, Developmental Therapeutics Branch, NCI, NIH, Bethesda, Maryland
| | - Sven Bilke
- Genetics Branch, NCI, NIH, Bethesda, Maryland
| | - Liang Cao
- Genetics Branch, NCI, NIH, Bethesda, Maryland
| | - Junko Murai
- Laboratory of Molecular Pharmacology, Developmental Therapeutics Branch, NCI, NIH, Bethesda, Maryland
| | - Fabricio G Sousa
- Laboratory of Molecular Pharmacology, Developmental Therapeutics Branch, NCI, NIH, Bethesda, Maryland. CETROGEN, PPGFARM, UFMS, Campo Grande, Brazil
| | - Mihoko Yamade
- Laboratory of Molecular Pharmacology, Developmental Therapeutics Branch, NCI, NIH, Bethesda, Maryland
| | - Vinodh Rajapakse
- Laboratory of Molecular Pharmacology, Developmental Therapeutics Branch, NCI, NIH, Bethesda, Maryland
| | - Sudhir Varma
- Laboratory of Molecular Pharmacology, Developmental Therapeutics Branch, NCI, NIH, Bethesda, Maryland
| | - Lee J Helman
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Javed Khan
- Genetics Branch, NCI, NIH, Bethesda, Maryland
| | | | - Yves Pommier
- Laboratory of Molecular Pharmacology, Developmental Therapeutics Branch, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|