1
|
Maharati A, Tolue Ghasaban F, Akhlaghipour I, Taghehchian N, Zangouei AS, Moghbeli M. MicroRNA-495: a therapeutic and diagnostic tumor marker. J Mol Histol 2023; 54:559-578. [PMID: 37759132 DOI: 10.1007/s10735-023-10159-0] [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: 10/11/2022] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Therapeutic and diagnostic progresses have significantly reduced the mortality rate among cancer patients during the last decade. However, there is still a high rate of mortality among cancer patients. One of the important reasons involved in the high mortality rate is the late diagnosis in advanced tumor stages that causes the failure of therapeutic strategies in these patients. Therefore, investigating the molecular mechanisms involved in tumor progression has an important role in introducing the efficient early detection markers. MicroRNAs (miRNAs) as stable factors in body fluids are always considered as non-invasive diagnostic and prognostic markers. In the present review, we investigated the role of miR-495 in tumor progression. It has been reported that miR-495 has mainly a tumor suppressor function through the regulation of transcription factors and tyrosine kinases as well as cellular processes such as multidrug resistance, chromatin remodeling, and signaling pathways. This review can be an effective step towards introducing the miR-495 as a non-invasive diagnostic/prognostic marker as well as a suitable target in tumor therapy.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Tolue Ghasaban
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Wang S, Liu R. Insights into the pleiotropic roles of ZNF703 in cancer. Heliyon 2023; 9:e20140. [PMID: 37810156 PMCID: PMC10559930 DOI: 10.1016/j.heliyon.2023.e20140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Zinc finger proteins (ZNFs) belong to the NET/NLZ protein family. In physiological functions, ZNF703 play significant roles in embryonic development, especially in the nervous system. As an transcription factors with zinc finger domains, abnormal regulation of the ZNF703 protein is associated with enhanced proliferation, invasion, and metastasis as well as drug resistance in many tumors, although mechanisms of action vary depending on the specific tumor microenvironment. ZNF703 lacks a nuclear localization sequence despite its function requiring nuclear DNA binding. The purpose of this review is to summarize the architecture of ZNF703, its roles in tumorigenesis, and tumor progression, as well as future oncology therapeutic prospects, which have implications for understanding tumor susceptibility and progression.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Obstetrics and Gynaecology, Tianjin Central Hospital of Gynecology Obstetrics, No. 156 Nan Kai San Ma Lu, Tianjin, 300000, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, 300000, China
- Department of Obstetrics and Gynaecology, Nankai University Maternity Hospital, Tianjin, 300000, China
| | - Rong Liu
- Department of Obstetrics and Gynecology, Tianjin First Center Hospital, Tianjin, China
| |
Collapse
|
3
|
Chuang TD, Ton N, Rysling S, Quintanilla D, Boos D, Gao J, McSwiggin H, Yan W, Khorram O. The Influence of Race/Ethnicity on the Transcriptomic Landscape of Uterine Fibroids. Int J Mol Sci 2023; 24:13441. [PMID: 37686244 PMCID: PMC10487975 DOI: 10.3390/ijms241713441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
The objective of this study was to determine if the aberrant expression of select genes could form the basis for the racial disparity in fibroid characteristics. The next-generation RNA sequencing results were analyzed as fold change [leiomyomas/paired myometrium, also known as differential expression (DF)], comparing specimens from White (n = 7) and Black (n = 12) patients. The analysis indicated that 95 genes were minimally changed in tumors from White (DF ≈ 1) but were significantly altered by more than 1.5-fold (up or down) in Black patients. Twenty-one novel genes were selected for confirmation in 69 paired fibroids by qRT-PCR. Among these 21, coding of transcripts for the differential expression of FRAT2, SOX4, TNFRSF19, ACP7, GRIP1, IRS4, PLEKHG4B, PGR, COL24A1, KRT17, MMP17, SLN, CCDC177, FUT2, MYO5B, MYOG, ZNF703, CDC25A, and CDCA7 was significantly higher, while the expression of DAB2 and CAV2 was significantly lower in tumors from Black or Hispanic patients compared with tumors from White patients. Western blot analysis revealed a greater differential expression of PGR-A and total progesterone (PGR-A and PGR-B) in tumors from Black compared with tumors from White patients. Collectively, we identified a set of genes uniquely expressed in a race/ethnicity-dependent manner, which could form the underlying mechanisms for the racial disparity in fibroids and their associated symptoms.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Nhu Ton
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Shawn Rysling
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Derek Quintanilla
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Drake Boos
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Jianjun Gao
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Hayden McSwiggin
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Udu-Ituma S, Adélaïde J, Le TK, Omabe K, Finetti P, Paris C, Guille A, Bertucci F, Birnbaum D, Rocchi P, Chaffanet M. ZNF703 mRNA-Targeting Antisense Oligonucleotide Blocks Cell Proliferation and Induces Apoptosis in Breast Cancer Cell Lines. Pharmaceutics 2023; 15:1930. [PMID: 37514116 PMCID: PMC10384502 DOI: 10.3390/pharmaceutics15071930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
The luminal B molecular subtype of breast cancers (BC) accounts for more than a third of BCs and is associated with aggressive clinical behavior and poor prognosis. The use of endocrine therapy in BC treatment has significantly contributed to the decrease in the number of deaths in recent years. However, most BC patients with prolonged exposure to estrogen receptor (ER) selective modulators such as tamoxifen develop resistance and become non-responsive over time. Recent studies have implicated overexpression of the ZNF703 gene in BC resistance to endocrine drugs, thereby highlighting ZNF703 inhibition as an attractive modality in BC treatment, especially luminal B BCs. However, there is no known inhibitor of ZNF703 due to its nuclear association and non-enzymatic activity. Here, we have developed an antisense oligonucleotide (ASO) against ZNF703 mRNA and shown that it downregulates ZNF703 protein expression. ZNF703 inhibition decreased cell proliferation and induced apoptosis. Combined with cisplatin, the anti-cancer effects of ZNF703-ASO9 were improved. Moreover, our work shows that ASO technology may be used to increase the number of targetable cancer genes.
Collapse
Affiliation(s)
- Sandra Udu-Ituma
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
- Department of Biology, Alex Ekwueme Federal University Ndufu-Alike Ikwo, Abakaliki P.M.B. 1010, Ebonyi State, Nigeria
- European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - José Adélaïde
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Thi Khanh Le
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
- European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Kenneth Omabe
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Pascal Finetti
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Clément Paris
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Arnaud Guille
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - François Bertucci
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Daniel Birnbaum
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Palma Rocchi
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
- European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Max Chaffanet
- Equipe Labellisée Ligue Nationale Contre le Cancer, Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| |
Collapse
|
5
|
Mohamed AH, Harb OA, Shorbagy SE, Balata R, Amin MF, Abd-Elaziz O. Prognostic Roles of ZNF703 and SMAD4 Expression in Patients with Papillary Thyroid Cancer and Association with Nodal Metastasis. Indian J Surg Oncol 2022; 13:169-177. [PMID: 35462659 PMCID: PMC8986956 DOI: 10.1007/s13193-022-01519-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 02/03/2022] [Indexed: 11/24/2022] Open
Abstract
It is important to detect novel predictive biomarkers of cervical lymph node metastasis (CLNM) in papillary thyroid carcinoma (PTC) to help the surgeons in making early decision of performing central lymph node dissection and aggressive management strategies in selected high-risk patients, thus improving their prognosis. Zinc finger protein 703 (ZNF703) is a member of the neutrophil extracellular trap (NET) transcription factors family which has roles in proliferation and invasion of cancer cells. SMAD4 is a protein that has a role in cellular processes including cell proliferation, invasion, and metastasis through many genes' transcription. In this study, we aimed to assess the expression of ZNF703 and SMAD4 in PTC and evaluated the correlation between its expression, clinicopathological features of PTC cases, and prognostic parameters of patients to evaluate their roles in PTC progression. This is a retrospective study which included 40 cases with PTC. For immunohistochemistry, tissues stained their paraffin blocks with ZNF703 and SMAD4. We followed patients to detect disease progression and recurrence. Positive ZNF703 expression and negative SMAD4 expression were associated with higher incidence of CLNM, advanced stage and large tumor size, higher incidence of disease progression, recurrence, unfavorable PFS, and unfavorable OS rates. The higher ZNF703 expression and the lower SMAD4 expression were significantly increased in PTC patients with cervical LNM compared with those without. ZNF703 over expression and downregulation SMAD4 expression was significantly increased in PTC patients. Elevated expression of ZNF703 in tumor tissue with CLNM can be considered a predictive factor for the development of metastasis.
Collapse
Affiliation(s)
| | - Ola A. Harb
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Shereen El Shorbagy
- Department of Medical Oncology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rawda Balata
- Department of Clinical Oncology& Nuclear Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed Farouk Amin
- Department of General Surgery, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Osama Abd-Elaziz
- Department of General Surgery, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
6
|
Rea J, Carissimo A, Trisciuoglio D, Illi B, Picard D, Remke M, Laneve P, Caffarelli E. Identification and Functional Characterization of Novel MYC-Regulated Long Noncoding RNAs in Group 3 Medulloblastoma. Cancers (Basel) 2021; 13:cancers13153853. [PMID: 34359754 PMCID: PMC8345409 DOI: 10.3390/cancers13153853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Medulloblastoma is the most common malignant pediatric brain tumor, which accounts for approximately 20% of all childhood brain tumors. To date, no pharmacological approaches are decisive in the treatment of this cancer, while the secondary effects of conventional therapies as chemotherapy, radiotherapy or surgical interventions heavily affect the quality of life of patients. This requires the rapid development of alternative molecular therapies, which are the future challenge of personalized medicine. In this context, we addressed our research towards the most aggressive form of Medulloblastoma to identify novel genes responsible for its onset and/or progression. We discovered three newly implicated genes, for which we highlighted a contribution in the control of cancer cell features. Deepening into the Medulloblastoma biology, this study represents a further step forward for the development of molecular therapies in the era of precision oncology. Abstract The impact of protein-coding genes on cancer onset and progression is a well-established paradigm in molecular oncology. Nevertheless, unveiling the contribution of the noncoding genes—including long noncoding RNAs (lncRNAs)—to tumorigenesis represents a great challenge for personalized medicine, since they (i) constitute the majority of the human genome, (ii) are essential and flexible regulators of gene expression and (iii) present all types of genomic alterations described for protein-coding genes. LncRNAs have been increasingly associated with cancer, their highly tissue- and cancer type-specific expression making them attractive candidates as both biomarkers and therapeutic targets. Medulloblastoma is one of the most common malignant pediatric brain tumors. Group 3 is the most aggressive subgroup, showing the highest rate of metastasis at diagnosis. Transcriptomics and reverse genetics approaches were combined to identify lncRNAs implicated in Group 3 Medulloblastoma biology. Here we present the first collection of lncRNAs dependent on the activity of the MYC oncogene, the major driver gene of Group 3 Medulloblastoma. We assessed the expression profile of selected lncRNAs in Group 3 primary tumors and functionally characterized these species. Overall, our data demonstrate the direct involvement of three lncRNAs in Medulloblastoma cancer cell phenotypes.
Collapse
Affiliation(s)
- Jessica Rea
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy;
| | | | - Daniela Trisciuoglio
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy; (D.T.); (B.I.)
| | - Barbara Illi
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy; (D.T.); (B.I.)
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, 40225 Düsseldorf, Germany; (D.P.); (M.R.)
- Department of Neuropathology, Faculty of Medicine, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Consortium Neuro-Oncogenomics Cancer Research (DKTK), Partner Site Essen/Düsseldorf, 40225 Düsseldorf, Germany
| | - Marc Remke
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, 40225 Düsseldorf, Germany; (D.P.); (M.R.)
- Department of Neuropathology, Faculty of Medicine, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Consortium Neuro-Oncogenomics Cancer Research (DKTK), Partner Site Essen/Düsseldorf, 40225 Düsseldorf, Germany
| | - Pietro Laneve
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy; (D.T.); (B.I.)
- Correspondence: (P.L.); (E.C.); Tel.: +39-06-49912205 (P.L.); +39-06-49912201 (E.C.)
| | - Elisa Caffarelli
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy; (D.T.); (B.I.)
- Correspondence: (P.L.); (E.C.); Tel.: +39-06-49912205 (P.L.); +39-06-49912201 (E.C.)
| |
Collapse
|
7
|
Gu Y, Wan C, Zhou G, Zhu J, Shi Z, Zhuang Z. TYMSOS drives the proliferation, migration, and invasion of gastric cancer cells by regulating ZNF703 via sponging miR-4739. Cell Biol Int 2021; 45:1710-1719. [PMID: 33847425 DOI: 10.1002/cbin.11610] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is a kind of malignancy originating from the epithelium of gastric mucosa. Long noncoding RNAs (lncRNAs) are tightly related to the GC progression. Herein, our research was meant to investigate a novel lncRNA thymidylate synthetase opposite strand (TYMSOS) in GC. Quantitative real-time polymerase chain reaction was used to analyze TYMSOS expression in GC cells. 5-Ethynyl-2'-deoxyuridine, flow cytometry analysis, and transwell assay detected the influence of TYMSOS on GC cell proliferation, apoptosis, migration, and invasion. Subcellular fractionation and fluorescent in situ hybridization assays determined the cellular localization of TYMSOS in GC cells. Bioinformatics programs, RNA-binding protein immunoprecipitation, RNA pull-down, and luciferase reporter assays measured the molecular interplays of TYMSOS in GC cells. In brief, TYMSOS was highly expressed in GC cells, and TYMSOS silence inhibited GC cell proliferation, migration, and invasion while elevating cell apoptosis. Functionally, TYMSOS functioned as a competing endogenous RNA to posttranscriptionally modulate GC progression. TYMSOS interacted with miR-4739 to regulate its target gene zinc finger protein 703. Collectively, our study proved the tumor-promoting role of TYMSOS in GC cells, which might offer the utility value for GC treatment.
Collapse
Affiliation(s)
- Yulan Gu
- Department of Oncology, Changshu Second People's Hospital, Changshu, Jiangsu, China
| | - Chuandan Wan
- Department of Medical Laboratory Science and Education, Changshu Medical Laboratory, Changshu, Jiangsu, China
| | - Guoqiang Zhou
- Department of General Surgery, Changshu Second People's Hospital, Changshu, Jiangsu, China
| | - Jinlian Zhu
- Department of Oncology, Changshu Second People's Hospital, Changshu, Jiangsu, China
| | - Zhiliang Shi
- Department of General Surgery, Changshu Second People's Hospital, Changshu, Jiangsu, China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
8
|
Guo J, Luo C, Yang Y, Dong J, Guo Z, Yang J, Lian H, Ye C, Liu M. MiR-491-5p, as a Tumor Suppressor, Prevents Migration and Invasion of Breast Cancer by Targeting ZNF-703 to Regulate AKT/mTOR Pathway. Cancer Manag Res 2021; 13:403-413. [PMID: 33488122 PMCID: PMC7816048 DOI: 10.2147/cmar.s279747] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Background Large amounts of microRNAs (miRNAs) have been reported to be aberrantly expressed in malignant cancers. MiR-491-5p makes a significant contribution to the inhibition of multiple cancer processes. However, the specific mechanism and function of miR-491-5p and in breast cancer (BC) is still not fully elucidated. Methods MiR-491-5p and ZNF-703 expressions or gene transfection effects were identified by RT-qPCR or Western blot in BC tissues or cells. And ZNF-703 expression was monitored through immunohistochemistry method. Cellular function was also confirmed using Transwell assay. Besides, AKT/mTOR pathway-related proteins were analyzed using Western blotting analysis. Moreover, the interplay between miR-491-5p and ZNF-703 was verified through dual-luciferase reporter assay. Results miR-491-5p was lowly expressed, ZNF-703 was highly expressed in BC, and miR-491-5p with low expression and ZNF-703 with high expression were associated with poor prognosis of BC patients. Results of cellular function revealed that overexpression of miR-491-5p markedly suppressed BC cell migration and invasion, and knockdown of miR-491-5p had the opposite effect. Besides, mechanism research disclosed that miR-491-5p directly could bind to ZNF-703 and downregulate ZNF-703. Moreover, we proved that ZNF-703 could prominently reverse the influences of miR-491-5p on the migration and invasion of BC cells. More importantly, the data revealed that miR-491-5p repressed AKT/mTOR pathway by ZNF-703 in BC cells. Conclusion MiR-491-5p prominently suppresses the metastasis of BC cells through ZNF-703 to regulate AKT/mTOR pathway, indicating that miR-491-5p and ZNF-703 might be served as the potential therapeutic targets for BC.
Collapse
Affiliation(s)
- Jingyun Guo
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Can Luo
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yuqin Yang
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jianyu Dong
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Zhaoze Guo
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jinlamao Yang
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Huining Lian
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Changsheng Ye
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Minfeng Liu
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| |
Collapse
|
9
|
Klæstad E, Sawicka JE, Engstrøm MJ, Ytterhus B, Valla M, Bofin AM. ZNF703 gene copy number and protein expression in breast cancer; associations with proliferation, prognosis and luminal subtypes. Breast Cancer Res Treat 2021; 186:65-77. [PMID: 33389351 DOI: 10.1007/s10549-020-06035-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/25/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE Amplification of 8p12 is frequent in breast cancer and associated with poor prognosis in luminal subtypes. ZNF703 has been identified as the driver gene of proliferation in the A1 amplicon situated in 8p12. In this study, the aims were to investigate associations between ZNF703 copy number alterations and molecular subtypes, proliferation and prognosis, and using immunohistochemistry, examine associations between ZNF703 copy number and ZNF703 protein expression. METHODS Copy number alterations in 702 primary breast tumours and corresponding lymph node metastases were examined using fluorescence in situ hybridization with probes for ZNF703 and centromere 8. In addition, protein expression was studied in 869 tumours from the same cohort. Associations between copy number alterations and protein expression and tumour characteristics were assessed using Pearson chi square test. The prognostic impact of ZNF703 copy number increase and protein expression was assessed estimating cumulative incidence of breast cancer death and hazard ratios. RESULTS We found mean ZNF703 copy number ≥ 6 in 7% of tumours, most frequently in Luminal B subtypes. We found a positive association between increased copy number, and high proliferation, high histological grade, and poor prognosis. Luminal A tumours with high copy number had high histological grade and poor prognosis (borderline significant). We found positive nuclear staining in 76% of primary tumours. There was an association between copy number status and protein expression, but no association between protein expression and prognosis. CONCLUSIONS In breast cancer, high ZNF703 copy number is associated with increased proliferation, Luminal B subtypes and poor prognosis.
Collapse
Affiliation(s)
- Elise Klæstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons Gate, 7030, Trondheim, Norway.
| | | | - Monica Jernberg Engstrøm
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons Gate, 7030, Trondheim, Norway.,Department of Breast and Endocrine Surgery, St. Olav's Hospital, Trondheim University Hospital, 7006, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons Gate, 7030, Trondheim, Norway
| | - Marit Valla
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons Gate, 7030, Trondheim, Norway.,Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, 7006, Trondheim, Norway
| | - Anna Mary Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons Gate, 7030, Trondheim, Norway
| |
Collapse
|
10
|
Wang S, Wang C, Hu Y, Li X, Jin S, Liu O, Gou R, Zhuang Y, Guo Q, Nie X, Zhu L, Liu J, Lin B. ZNF703 promotes tumor progression in ovarian cancer by interacting with HE4 and epigenetically regulating PEA15. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:264. [PMID: 33246486 PMCID: PMC7693506 DOI: 10.1186/s13046-020-01770-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
Background It is known that the transcription factor zinc finger protein 703 (ZNF703) plays an important role in physiological functions and the occurrence and development of various tumors. However, the role and mechanism of ZNF703 in ovarian cancer are unclear. Materials and methods Immunohistochemistry was used to analyze the expression of ZNF703 in ovarian cancer patients and to assess the effect of ZNF703 expression on the survival and prognosis of ovarian cancer patients. ZNF703 overexpression and suppression expression experiments were used to evaluate the effect of ZNF703 on malignant biological behavior of ovarian cancer cells in vitro. Detecting the interaction between HE4 and ZNF703 by immunofluorescence colocalization and coprecipitation, and nuclear translocation. Chromatin immunoprecipitation-sequencing (ChIP-Seq), dual luciferase reporter assay, ChIP-PCR, in vivo model were applied to study the molecular mechanism of ZNF703 affecting the development of ovarian cancer. Results ZNF703 was highly expressed in ovarian cancer tissues, and its expression level is related to the prognosis of ovarian cancer patients. In vivo and in vitro experiments confirmed that ZNF703 overexpression/inhibition expression will promoted/inhibited the malignant biological behavior of ovarian cancer. Mechanically, ZNF703 interacted with HE4, and HE4 promoted nuclear translocation of ZNF703. ChIP-Seq identified multiple regulatory targets of ZNF703, of which ZNF703 directly binds to the enhancer region of PEA15 to promote the transcription of PEA15 and thereby promoted the proliferation of cancer cells. Conclusion The results showed that ZNF703 as an oncogene played an important role in the epigenetic modification of ovarian cancer proliferation, and suggested that ZNF703 as a transcription factor may become a prognostic factor and a potential therapeutic target for ovarian cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-020-01770-0.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Caixia Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuexin Hu
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Xiao Li
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Shan Jin
- Department of Obstetrics and Gynaecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Ouxuan Liu
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Rui Gou
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Yuan Zhuang
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Qian Guo
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Xin Nie
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Liancheng Zhu
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Juanjuan Liu
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China
| | - Bei Lin
- Department of Obstetrics and Gynaecology, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China. .,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shenyang, Liaoning, China.
| |
Collapse
|
11
|
Wang Y, Wu Z, Li Y, Zheng Z, Yan J, Tian S, Han L. Long Non-Coding RNA H19 Promotes Proliferation, Migration and Invasion and Inhibits Apoptosis of Breast Cancer Cells by Targeting miR-491-5p/ZNF703 Axis. Cancer Manag Res 2020; 12:9247-9258. [PMID: 33061615 PMCID: PMC7532042 DOI: 10.2147/cmar.s246009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 08/25/2020] [Indexed: 12/24/2022] Open
Abstract
Background Breast cancer is one of the most common cancers worldwide. Long non-coding RNAs and microRNAs act as important regulators in human cancers. This study aims to explore the molecular mechanism among H19, miR-491-5p and zinc finger 703 (ZNF703) in breast cancer. Materials and Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to detect the expression of H19, miR-491-5p and ZNF703. Cell Counting Kit 8 (CCK-8) assay was performed to evaluate cell proliferation. Cell apoptosis was assessed by flow cytometry assay. The number of migrated and invaded cells was counted by transwell assay. Dual luciferase reporter assay was carried out to test luciferase activity. Protein level of ZNF703 was measured by Western blot assay. Results H19 was highly expressed in breast tissues and cells. H19 knockdown inhibited proliferation, induced apoptosis and blocked migration and invasion. Moreover, H19 bound to miR-491-5p and negatively regulated miR-491-5p expression. MiR-491-5p inhibition abrogated the activities of proliferation, apoptosis, migration and invasion affected by H19 knockdown. Furthermore, miR-491-5p directly targeted ZNF703 and inversely modulated ZNF703 expression. ZNF703 up-regulation rescued the effects of miR-491-5p overexpression on proliferation, apoptosis, migration and invasion. In addition, H19 knockdown reduced ZNF703 expression by targeting miR-491-5p/ZNF703 axis. Conclusion H19 promoted proliferation, migration and invasion and retarded apoptosis of breast cancer cells via sponging miR-491-5p to down-regulate ZNF703 expression.
Collapse
Affiliation(s)
- Yongkun Wang
- Department of Thyroid Surgery, Liaocheng People's Hospital (Clinical Hospital of Shandong First Medical University) Liaocheng, Shandong, People's Republic of China
| | - Zhen Wu
- Department of Thyroid Surgery, Liaocheng People's Hospital (Clinical Hospital of Shandong First Medical University) Liaocheng, Shandong, People's Republic of China
| | - Yingxue Li
- Department of Pathology, Liaocheng People's Hospital (Clinical Hospital of Shandong First Medical University) Liaocheng, Shandong, People's Republic of China
| | - Zheng Zheng
- Department of Pathology, Liaocheng People's Hospital (Clinical Hospital of Shandong First Medical University) Liaocheng, Shandong, People's Republic of China
| | - Jinqiang Yan
- Department of Pathology, Liaocheng People's Hospital (Clinical Hospital of Shandong First Medical University) Liaocheng, Shandong, People's Republic of China
| | - Shuyan Tian
- Department of Pathology, Liaocheng People's Hospital (Clinical Hospital of Shandong First Medical University) Liaocheng, Shandong, People's Republic of China
| | - Lin Han
- Department of Pathology, Liaocheng People's Hospital (Clinical Hospital of Shandong First Medical University) Liaocheng, Shandong, People's Republic of China
| |
Collapse
|
12
|
Huh SJ, Oh SY, Lee S, Lee JH, Kim SH, Pak MK, Kim HJ. Mutational analysis of extranodal marginal zone lymphoma using next generation sequencing. Oncol Lett 2020; 20:205. [PMID: 32963611 PMCID: PMC7491050 DOI: 10.3892/ol.2020.12068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
Extranodal marginal zone lymphoma is a type of low-grade B-cell lymphoma that can be classified as a mucosal-associated lymphoid tissue (MALT) lymphoma. Recently, second-generation or next-generation sequencing (NGS), which allows simultaneous sequencing of hundreds to billions of DNA strands, has been a focus of attention and is rapidly being adopted in various fields. In the present study, paraffin-embedded tissue samples of gastric MALT lymphoma (n=1) and small intestine MALT lymphoma (n=4) were selected, and DNA was extracted from the tissue samples. After performing quality control, NGS was performed using HemaSCAN™, a custom panel of 426 genes, including essential blood cancer genes. NGS revealed single nucleotide variations (SNVs), short insertions and deletions (InDels) and copy number variations (CNVs). These genomic variants were reported as annotated, known or novel variants. An annotated variant, an erb-b2 receptor tyrosine kinase 2 gene amplification, was observed in one patient. Known and novel variants, including SNVs of SET binding protein 6 (SETBP6), Runt-related transcription factor 1 and Kelch-like ECH-associated protein 1 genes, InDel of the marker of proliferation Ki-67 gene, and CNVs of the zinc finger protein 703 and NOTCH1 genes, were observed in ≥2 patients. Additionally, InDels with frameshift mutations were identified in the B-cell lymphoma/leukemia 10, DEAD-box helicase 3 X-linked, forkhead box O3 and mucin 2, oligomeric mucus/gel-forming genes in one patient. Since few NGS studies have been performed on MALT lymphoma, the current results were unable to determine if the different mutations that were identified are ‘actionable’ (that is, potentially responsive to a targeted therapy) Further studies are required to determine the associations between genetic mutations and the development of MALT lymphoma.
Collapse
Affiliation(s)
- Seok Jae Huh
- Department of Internal Medicine, Dong-A University College of Medicine, Seo-gu, Busan 49201, Republic of Korea
| | - Sung Yong Oh
- Department of Internal Medicine, Dong-A University College of Medicine, Seo-gu, Busan 49201, Republic of Korea
| | - Suee Lee
- Department of Internal Medicine, Dong-A University College of Medicine, Seo-gu, Busan 49201, Republic of Korea
| | - Ji Hyun Lee
- Department of Internal Medicine, Dong-A University College of Medicine, Seo-gu, Busan 49201, Republic of Korea
| | - Sung Hyun Kim
- Department of Internal Medicine, Dong-A University College of Medicine, Seo-gu, Busan 49201, Republic of Korea
| | - Min Kyung Pak
- Department of Pathology, Dong-A University College of Medicine, Seo-gu, Busan 49201, Republic of Korea
| | - Hyo-Jin Kim
- Department of Internal Medicine, Dong-A University College of Medicine, Seo-gu, Busan 49201, Republic of Korea
| |
Collapse
|
13
|
Hu Z, Bi G, Sui Q, Bian Y, Du Y, Liang J, Li M, Zhan C, Lin Z, Wang Q. Analyses of multi-omics differences between patients with high and low PD1/PDL1 expression in lung squamous cell carcinoma. Int Immunopharmacol 2020; 88:106910. [PMID: 32829091 DOI: 10.1016/j.intimp.2020.106910] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Immunotherapy has achieved excellent results in patients with lung squamous cell carcinoma. However, in which population it can exert the greatest effect is still unknown. Some studies have suggested that its effect is related to the expression level of PD1. Analyzing the relationship between PD1 expression level and genetic differences in lung squamous cell carcinoma patients will be helpful in understanding the underlying causes of this immunotherapy effect and provide a reference for clinical practice. METHODS In this study, we used RNA-seq, miRNA-seq, methylation array, mutation profiles, and copy number variation data from the TCGA database and RNA-seq data from the GEO database to analyze the distinctive genomic patterns associated with PD1 and PDL1 expression. RNA-seq data from 44 LUSC patients who underwent surgery at Zhongshan Hospital were also included in the study. RESULTS After grouping LUSC patients according to the expression levels of PD1 and PDL1, we found no significant difference in survival between the two groups. However, 178 genes, including IL-21, KLRC3, and KLRC4, were significantly upregulated in both the TCGA and GEO databases in the high expression group, and there was a precise correlation between gene expression and epigenetic changes in the two groups. At the same time, the overall level of somatic mutations was not significantly different between the two groups. It is worth noting that the gene enrichment results showed that the differential pathways were mainly enriched in immune regulation, especially T cell-related immune activities. CONCLUSION We found that the differences in gene expression between the two groups were related to immunity, which may affect the effectiveness of immunotherapy. We hope our results can provide a reference for further research and help in finding other targets to improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yajing Du
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Zongwu Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
14
|
Xu G, Zhang Y, Li N, Wu Y, Zhang J, Xu R, Ming H. LBX2-AS1 up-regulated by NFIC boosts cell proliferation, migration and invasion in gastric cancer through targeting miR-491-5p/ZNF703. Cancer Cell Int 2020; 20:136. [PMID: 32351330 PMCID: PMC7183605 DOI: 10.1186/s12935-020-01207-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/06/2020] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The crucial role of long non-coding RNAs (lncRNAs) has been certified in human cancers. The lncRNAs with abnormal expressions could act as tumor inhibitors or oncogenes in the advancement of tumors. LBX2-AS1 was once reported to accelerate esophageal squamous cell carcinoma. Nonetheless, its function in gastric cancer (GC) remained a riddle. METHODS RT-qPCR was used to examine the expression of NFIC/LBX2-AS1/miR-491-5p/ZNF703 in GC cell lines. The functions of LBX2-AS1 in GC were appraised by colony formation, EdU, flow cytometry analysis, transwell and wound healing assays. Luciferase reporter, ChIP and RNA pull down assays were utilized to evaluate the interactions among genes. RESULTS LBX2-AS1 was up-regulated in GC cell lines. Knockdown of LBX2-AS1 repressed the proliferative, migratory, and invasive abilities of GC cells. Moreover, LBX2-AS1 was transcriptionally activated by NFIC. And LBX2-AS1 could bind with miR-491-5p. Besides, miR-491-5p depletion or ZNF703 upregulation could counteract the repressing effects of LBX2-AS1 silence on GC progression. CONCLUSION In a word, LBX2-AS1 up-regulated by NFIC promoted GC progression via targeting miR-491-5p/ZNF703, implying LBX2-AS1 was an underlying treatment target for GC patients.
Collapse
Affiliation(s)
- Gang Xu
- Oncology Department, The 960th Hospital of the PLA, No. 20 Zhanbei Road, Zibo, 255300 Shandong China
| | - Yan Zhang
- Oncology Department, The 960th Hospital of the PLA, No. 20 Zhanbei Road, Zibo, 255300 Shandong China
| | - Na Li
- Oncology Department, The 960th Hospital of the PLA, No. 20 Zhanbei Road, Zibo, 255300 Shandong China
| | - Yanling Wu
- Oncology Department, The 960th Hospital of the PLA, No. 20 Zhanbei Road, Zibo, 255300 Shandong China
| | - Jinbiao Zhang
- Oncology Department, The 960th Hospital of the PLA, No. 20 Zhanbei Road, Zibo, 255300 Shandong China
| | - Rui Xu
- Oncology Department, The 960th Hospital of the PLA, No. 20 Zhanbei Road, Zibo, 255300 Shandong China
| | - Hui Ming
- Oncology Department, The 960th Hospital of the PLA, No. 20 Zhanbei Road, Zibo, 255300 Shandong China
| |
Collapse
|
15
|
Huan C, Xiaoxu C, Xifang R. Zinc Finger Protein 521, Negatively Regulated by MicroRNA-204-5p, Promotes Proliferation, Motility and Invasion of Gastric Cancer Cells. Technol Cancer Res Treat 2020; 18:1533033819874783. [PMID: 31526099 PMCID: PMC6749787 DOI: 10.1177/1533033819874783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE This study aims to investigate the expression, role, and detailed mechanism of microRNA-204-5p and zinc finger protein 521 in gastric cancer. METHODS Immunohistochemistry was adopted to detect the expressions of zinc finger protein 521 in 82 cases of gastric cancer tissues. Western blot was used to detect the expressions of zinc finger protein 521 in gastric cancer cells and adjacent cells. Moreover, the correlation between zinc finger protein 521 and the prognosis of patients were also evaluated. Cell Counting Kit 8 assay and colony formation assay were performed to figure out the impact of zinc finger protein 521 on the proliferation of gastric cancer cells. By conducting flow cytometry, the effect of zinc finger protein 521 on the apoptosis of gastric cancer cells was determined. The scratch wound healing assay and transwell invasion assay were carried out to determine the effect of zinc finger protein 521 on regulating the motility and invasion of gastric cancer cells. Ultimately, the targeting relationship and interaction between microRNA-204-5p and zinc finger protein 521 were verified by real-time polymerase chain reaction, Western blot, and dual luciferase reporter gene assay. RESULTS Compared with adjacent cells, zinc finger protein 521 was highly expressed in gastric cancer cells, which was related to TNM stage (P = .0388), tumor size (P = .0168), and local lymph node metastasis (P = .0024). Overexpressed zinc finger protein 521 can promote the proliferation, migration, and invasion of gastric cancer cells and inhibit the apoptosis. Zinc finger protein 521 is a target gene of microRNA-106-5p, and there was a negative correlation between the expression of zinc finger protein 521 and microRNA-204-5p. CONCLUSION Zinc finger protein 521 can arrest the apoptosis and enhance the proliferation, migration, and invasion of gastric cancer cells via regulating microRNA-204-5p. Our study may provide novel clues for the treatment of patients with gastric cancer.
Collapse
Affiliation(s)
- Chen Huan
- Department of Gastroenterology, The First People's Hospital of Yichang, Yichang, Hubei, China.,Department of Gastroenterology, The People's Hospital of Three Gorges University, Yichang, Hubei, China
| | - Cai Xiaoxu
- Department of Gastroenterology, The People's Hospital of Three Gorges University, Yichang, Hubei, China.,Department of Oncology, The First People's Hospital of Yichang, Yichang, Hubei, China
| | - Ren Xifang
- Department of Gastroenterology, The First People's Hospital of Yichang, Yichang, Hubei, China.,Department of Gastroenterology, The People's Hospital of Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
16
|
Yang X, Liu G, Li W, Zang L, Li D, Wang Q, Yu F, Xiang X. Silencing of zinc finger protein 703 inhibits medullary thyroid carcinoma cell proliferation in vitro and in vivo. Oncol Lett 2019; 19:943-951. [PMID: 31897207 PMCID: PMC6924197 DOI: 10.3892/ol.2019.11153] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 10/18/2019] [Indexed: 12/19/2022] Open
Abstract
Zinc finger protein 703 (ZNF703) is a new member of the zinc finger protein family of transcription factors that plays an important role during embryogenesis in metazoans. The overexpression of ZNF703 contributes to tumorigenesis and progression of a number of malignancies by activating the Akt/mammalian target of rapamycin (mTOR) signaling pathway. This pathway is activated in medullary thyroid cancer (MTC), but its mechanism of action is not yet fully understood. The aim of the present study was to examine the role of ZNF703 and its association with Akt/mTOR activation in MTC. The present study used the phosphorylation of Akt1 protein at serine 473 (pAkt473) as an indicator of signaling activation. Immunohistochemistry (IHC) staining and western blot analyses were performed in order to examine the expression of ZNF703 in 34 cases of MTC and 12 cases of corresponding normal thyroid tissues. ZNF703 expression in MTC was significantly higher compared with the corresponding normal thyroid tissues (P<0.05). Furthermore, expression of ZNF703 was associated with tumor size, lymph node metastasis and advanced stage of disease. IHC also demonstrated that the level of ZNF703 was positively correlated with p-Akt473 in the 34 cases of MTC. The human MTC cell line TT was selected for further investigation as TT cells exhibit Akt/mTOR activation. The biological effects of silencing ZNF703 in TT cells on proliferation and apoptosis, both in vitro and in vivo were investigated in the present study. ZNF703 silencing inhibited the proliferation of TT cells in vitro and inhibited xenograft tumor growth in vivo. These effects were accompanied by the substantial decrease of pAkt473 and the induction of p53 protein. These results demonstrate that ZNF703 may play a relevant role in MTC due to its association with the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaolin Yang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Geling Liu
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Weijuan Li
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Luyang Zang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Ding Li
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Qianqian Wang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Fang Yu
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Xiuxiu Xiang
- Department of Endocrinology (Section I), Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
17
|
Orhan C, Bakır B, Dalay N, Buyru N. ZNF703 is an important player in head and neck cancer. Clin Otolaryngol 2019; 44:1080-1086. [DOI: 10.1111/coa.13450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/10/2019] [Accepted: 09/29/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Ceren Orhan
- Department of Medical Biology Cerrahpasa Medical Faculty Istanbul University Istanbul Turkey
| | - Burak Bakır
- Department of Medical Biology Cerrahpasa Medical Faculty Istanbul University Istanbul Turkey
| | - Nejat Dalay
- Department of Medical Biology Cerrahpasa Medical Faculty Istanbul University Istanbul Turkey
| | - Nur Buyru
- Department of Medical Biology Cerrahpasa Medical Faculty Istanbul University Istanbul Turkey
| |
Collapse
|
18
|
Akbari Asl E, Fallah Mehrabadi J, Afshar D, Noorbazargan H, Tahmasebi H, Rahimi A. Apoptotic Effects of Linum album Extracts on AGS Human Gastric Adenocarcinoma Cells and ZNF703 Oncogene Expression. Asian Pac J Cancer Prev 2018; 19:2911-2916. [PMID: 30362321 PMCID: PMC6291028 DOI: 10.22034/apjcp.2018.19.10.2911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Introduction: Linum album is a medicinal plant endemic in Iran that is very important pharmaceutically. The present
study concerns the effect of different extracts of L. album on ZNF703 gene expression and apoptosis in human gastric
carcinoma AGS cells. Method and material: Hydro alchoholic L. album extracts from various plant sources were
produced by Maceration. AGS cells were treated with different concentrations (200, 400, 600, 800 and 1000 μg/ml)
and the cytotoxicity potency was assessed after 24 h by MTT assay. Then, quantitative real time PCR was conducted
for ZNF703 gene expression in AGS cells. Also, cell apoptosis/necrosis was assessed with the aid of Annexin V/PI
staining and quantification by flow cytometry. Results: L. album extracts exerted dose-dependent toxicity in the AGS
cell line. The mRNA levels of ZNF703 gene expression were significantly decreased with rhizome, fruit at fruiting,
leaf and stem at anthesis (P<0.001), and leaf and stem at fruiting extracts as compared to the controls (P<0.01). Also,
the number of apoptotic cells was increased from 2.70% (statistically significant; p<0.05) in untreated AGS cells to 44%,
following treatment with the leaf and stem at anthesis example. Discussion: Our findings revealed that the L. album
extracts can induce apoptosis and might modulate cytotoxicity by down regulating ZNF703 gene expression in AGS
cells. Therefore, this extract could be a good candidate for inhibiting cancer cell growth, especially that of gastric
cancer. In addition, ZNF703 may have potential as a therapeutic target.
Collapse
Affiliation(s)
- Elham Akbari Asl
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | | | | | | | | | | |
Collapse
|
19
|
ZNF703 is Overexpressed in Papillary Thyroid Carcinoma Tissues and Mediates K1 Cell Proliferation. Pathol Oncol Res 2018; 26:355-364. [DOI: 10.1007/s12253-018-0494-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022]
|
20
|
Zhai J, Yang Z, Cai X, Yao G, An Y, Wang W, Fan Y, Zeng C, Liu K. ZNF280B promotes the growth of gastric cancer in vitro and in vivo. Oncol Lett 2018; 15:5819-5824. [PMID: 29556309 DOI: 10.3892/ol.2018.8060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/07/2016] [Indexed: 12/26/2022] Open
Abstract
Zinc finger protein 280B (ZNF280B) mediates pro-survival and pro-growth functions in prostate cancer. However, in gastric cancer, its clinical significance remains poorly characterized. In the present study, the expression levels of ZNF280B in 60 patients with gastric cancer were examined using immunohistochemistry. The association between ZNF280B expression and clinicopathological features was assessed. Positive ZNF280B staining was demonstrated for 38 (63.3%) samples out of 60 gastric cancer cases in immunohistochemical analysis. ZNF280B expression was significantly associated with tumor size (P=0.017) and TNM stage (P=0.001). Furthermore, the proliferation index in the positive ZNF280B expression group was significantly higher (38.8±6.2) compared with that of the negative ZNF280B expression group (16.9±8.9; P<0.01). These results suggest that ZNF280B expression may be associated with the proliferation of gastric cancer cells. The role of ZNF280B in the growth of gastric cancer cells (MGC-803) was also investigated in vitro and in vivo by enhancing the expression of ZNF280B. A colony formation assay indicated that the number of colonies in the MGC-803 cells with enhanced ZNF280B (146±5.8) was significantly higher than that of the MGC-803 control group (97±5.1) and the negative control group (101±6.5; P<0.05). An MTT assay demonstrated that ZNF280B significantly promoted the proliferation of MGC-803 cells at days 3 and 4 (P<0.05). It was observed that the overexpression of ZNF280B may promote the growth of gastric cancer in vivo in xenograft studies. These findings indicate that ZNF280B may be a novel therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Jingming Zhai
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Zheng Yang
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaodong Cai
- Department of Neurology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Guoliang Yao
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Yanhui An
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Wei Wang
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Yonggang Fan
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Chao Zeng
- Department of Pathology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Kefeng Liu
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
21
|
Li K, Wang J, Han J, Lan Y, Xie C, Pan S, Liu L. Overexpression of ZNF703 facilitates tumorigenesis and predicts unfavorable prognosis in patients with cholangiocarcinoma. Oncotarget 2018; 7:76108-76117. [PMID: 27764785 PMCID: PMC5342799 DOI: 10.18632/oncotarget.12627] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/27/2016] [Indexed: 01/03/2023] Open
Abstract
Background NET (NocA/Nlz, Elbow, Tlp-1) family members have recently emerged as important players in the development of human cancers. Zinc finger protein 703 (ZNF703), locating on chromosome 8 (8p11.23), a member of the NET/Nlz family of zinc finger transcription factors, had been demonstrated to be a much novel oncogene of several malignancies. This study aimed to investigate the expression of ZNF703 in cholangiocarcinoma (CCA) and attempted to elucidate its biological effects in CCA progression. Methods The correlation between ZNF703 expression and clinicopathological characteristics of CCA was evaluated through analyzing 85 cases. The biological effects of ZNF703 were investigated both in vitro and in vivo in which proliferation, migration, and invasive potential were mainly explored. Statistical software SPSS 16.0 was used for statistical analyses. Results ZNF703 was overexpressed in CCA tissues with subcellular localizations mainly in the nucleus and partly in the cytoplasm or membrane. High expression of ZNF703 was related to tumor location (P=0.002), pathological grading (P=0.024), depth of invasion (P=0.002), distant metastasis (P=0. 011) and AJCC stage (P=0.008). Both in vitro and in vivo studies demonstrated that ZNF703 could potently promote proliferation, migration and invasion throughout the progression of CCA. Conclusion ZNF703 can potently facilitate tumor growth and metastasis in many respects throughout the progression of CCA, which may act as an oncogene in CCA and can be considered as a novel potential therapeutic target.
Collapse
Affiliation(s)
- Keyu Li
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Jiabei Wang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Jihua Han
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Yaliang Lan
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Changming Xie
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Shangha Pan
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Lianxin Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| |
Collapse
|
22
|
Liao X, Lu Y, Yang J, Kuang T, Jiang L, Wang Y, Kang H, Jiang B, Zhou X, He S. Transcription factor Sp1 is necessary and functional in regulating expression of oncogene ZNF703. Genes Genomics 2017. [DOI: 10.1007/s13258-017-0577-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
23
|
Li J, Chen Y, Chen Z, He A, Xie H, Zhang Q, Cai Z, Liu Y, Huang W. SPRY4-IT1: A novel oncogenic long non-coding RNA in human cancers. Tumour Biol 2017; 39:1010428317711406. [PMID: 28651500 DOI: 10.1177/1010428317711406] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNAs are classified as a kind of RNA, which are longer than 200 nucleotides in length and cannot be translated into proteins. Multiple studies have demonstrated that long non-coding RNAs are involved in various cellular processes, including proliferation, differentiation, cell death, and metastasis. Among numerous long non-coding RNAs, we focus on Sprouty4-Intron 1 (SPRY4-IT1), a well-known long non-coding RNA that is overexpressed in various kinds of tumor tissues and cell lines. Accumulating evidences show that SPRY4-IT1 was dysregulated in various cancers, including melanoma, breast cancer, esophageal squamous cell carcinoma, non-small cell lung cancer, gastric cancer, colon cancer, and hepatocellular carcinoma, and amplification of SPRY4-IT1 was associated with different clinicopathological features of cancer patients. Importantly, SPRY4-IT1 exerts important roles in tumor progression and metastasis. However, detailed molecular mechanisms of SPRY4-IT1 in cancer progression and metastasis were poorly understood. In this review, we have focused on the characteristics of SPRY4-IT1 and illustrated the biological function and mechanism of SPRY4-IT1 in cancer development.
Collapse
Affiliation(s)
- Jianfa Li
- 1 Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
- 2 Shantou University Medical College, Shantou, People's Republic of China
| | - Yincong Chen
- 2 Shantou University Medical College, Shantou, People's Republic of China
| | - Zhicong Chen
- 1 Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
- 2 Shantou University Medical College, Shantou, People's Republic of China
| | - Anbang He
- 2 Shantou University Medical College, Shantou, People's Republic of China
- 3 Anhui Medical University, Hefei, People's Republic of China
| | - Haibiao Xie
- 1 Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
- 2 Shantou University Medical College, Shantou, People's Republic of China
| | - Qiaoxiao Zhang
- 1 Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
| | - Zhiming Cai
- 1 Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
- 2 Shantou University Medical College, Shantou, People's Republic of China
- 3 Anhui Medical University, Hefei, People's Republic of China
| | - Yuchen Liu
- 1 Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
| | - Weiren Huang
- 1 Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
- 2 Shantou University Medical College, Shantou, People's Republic of China
- 3 Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
24
|
Evolution of the NET (NocA, Nlz, Elbow, TLP-1) protein family in metazoans: insights from expression data and phylogenetic analysis. Sci Rep 2016; 6:38383. [PMID: 27929068 PMCID: PMC5144077 DOI: 10.1038/srep38383] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 10/24/2016] [Indexed: 02/03/2023] Open
Abstract
The NET (for NocA, Nlz, Elbow, TLP-1) protein family is a group of conserved zinc finger proteins linked to embryonic development and recently associated with breast cancer. The members of this family act as transcriptional repressors interacting with both class I histone deacetylases and Groucho/TLE co-repressors. In Drosophila, the NET family members Elbow and NocA are vital for the development of tracheae, eyes, wings and legs, whereas in vertebrates ZNF703 and ZNF503 are important for the development of the nervous system, eyes and limbs. Despite the relevance of this protein family in embryogenesis and cancer, many aspects of its origin and evolution remain unknown. Here, we show that NET family members are present and expressed in multiple metazoan lineages, from cnidarians to vertebrates. We identified several protein domains conserved in all metazoan species or in specific taxonomic groups. Our phylogenetic analysis suggests that the NET family emerged in the last common ancestor of cnidarians and bilaterians and that several rounds of independent events of gene duplication occurred throughout evolution. Overall, we provide novel data on the expression and evolutionary history of the NET family that can be relevant to understanding its biological role in both normal conditions and disease.
Collapse
|
25
|
Baykara O, Dalay N, Kaynak K, Buyru N. ZNF703 Overexpression may act as an oncogene in non-small cell lung cancer. Cancer Med 2016; 5:2873-2878. [PMID: 27650486 PMCID: PMC5083741 DOI: 10.1002/cam4.847] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/17/2016] [Accepted: 07/08/2016] [Indexed: 12/03/2022] Open
Abstract
Despite therapeutic advances, lung cancer remains one of the most common causes of cancer‐related deaths worldwide. The ZNF703 gene has been identified as the driver of the 8p11‐12 region and its amplification or overexpression has been associated with several types of cancers. It has also been shown that ZNF703 overexpression can activate the Akt/mTOR signaling pathway. The aim of our study was to investigate the role of the ZNF703 gene in association with Akt/mTOR activation in non‐small cell lung cancer (NSCLC). Expression levels in tumors and matched noncancerous tissue samples from 47 patients were analyzed by qRT‐PCR and the Akt phosphorylation levels were investigated by Western blotting. Our results show that ZNF703 is up‐regulated in 63.4% of NSCLC tumor samples. Althogh the correlation did not reach a statistically significant level Akt phosphorylation was increased in tumor tissues expressing high levels of ZNF703. The role of the ZNF703 gene has not been investigated in NSCLC. Our data show that ZNF703 may contribute to tumor development in NSCLC by activating the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Onur Baykara
- Department of Medical Biology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Nejat Dalay
- Istanbul University Oncology Institute, Istanbul, Turkey
| | - Kamil Kaynak
- Department of Chest Surgery, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Nur Buyru
- Department of Medical Biology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey.
| |
Collapse
|
26
|
Li C, Li H, Zhang T, Li J, Ma F, Li M, Sui Z, Chang J. ZFX is a Strong Predictor of Poor Prognosis in Renal Cell Carcinoma. Med Sci Monit 2015; 21:3380-5. [PMID: 26540164 PMCID: PMC4638281 DOI: 10.12659/msm.894708] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background This study was designed to assay the expression of zinc finger protein X-linked (ZFX) in renal cell carcinoma (RCC) tissues and evaluate the correlation between ZFX expression and prognosis of RCC patients. Material/Methods The expressions of ZFX mRNA in 53 RCC tissues and 51 normal tissues were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Immunohistochemistry (IHC) technology was used to measure the expression of ZFX protein. Then chi-square test was conducted to verify the association between ZFX expression and clinical parameters. Next, we explored the overall survival rate of RCC patients with Kaplan-Meier analysis. Finally, the correlation between ZFX expression and the prognosis of RCC patients was evaluated by Cox regression analysis. Results The qRT-PCR result showed that the ZFX was significantly up-regulated in RCC tissues. As for the IHC consequence, the positive rate of ZFX expression in RCC specimens was 79.2%, while that in the normal control tissues was only 17.6%. Chi-square test showed that ZFX expression shared no close relationship with age, sex, or smoking (P>0.05), but was tightly associated with TNM stage, tumor size, and lymph node metastasis (P<0.05). Kaplan-Meier analysis showed that patients with ZFX positive expression had higher mortality than those with negative expression (P<0.05). Cox regression analysis revealed that ZFX expression had tight correlation with prognosis of RCC patients (HR=4.997, P=0.045, 95%CI=1.033–24.180). Conclusions Our findings show that ZFX could be considered as a predictor for prognosis of RCC patients.
Collapse
Affiliation(s)
- Changying Li
- Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Hongjie Li
- Institute of Basic Medicine, Hebei United University, Tangshan, Hebei, China (mainland)
| | - Ting Zhang
- Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Jianmin Li
- Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Fuling Ma
- Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Mei Li
- Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Zhifang Sui
- Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Jiwu Chang
- Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| |
Collapse
|
27
|
Jen J, Lin LL, Chen HT, Liao SY, Lo FY, Tang YA, Su WC, Salgia R, Hsu CL, Huang HC, Juan HF, Wang YC. Oncoprotein ZNF322A transcriptionally deregulates alpha-adducin, cyclin D1 and p53 to promote tumor growth and metastasis in lung cancer. Oncogene 2015; 35:2357-69. [PMID: 26279304 PMCID: PMC4865475 DOI: 10.1038/onc.2015.296] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 06/08/2015] [Accepted: 07/05/2015] [Indexed: 12/24/2022]
Abstract
ZNF322A encoding a classical Cys2His2 zinc finger transcription factor was previously revealed as a potential oncogene in lung cancer patients. However, the oncogenic role of ZNF322A and its underlying mechanism in lung tumorigenesis remain elusive. Here we show ZNF322A protein overexpression in 123 Asian and 74 Caucasian lung cancer patients. Multivariate Cox regression analysis indicated that ZNF322A was an independent risk factor for a poor outcome in lung cancer, corroborating the Kaplan–Meier results that patients with ZNF322A protein overexpression had significantly poorer overall survival than other patients. Overexpression of ZNF322A promoted cell proliferation and soft agar growth by prolonging cell cycle in S phase in multiple lung cell lines, including the immortalized lung cell BEAS-2B. In addition, ZNF322A overexpression enhanced cell migration and invasion, whereas knockdown of ZNF322A reduced cell growth, invasion and metastasis abilities in vitro and in vivo. Quantitative proteomic analysis revealed potential ZNF322A-regulated downstream targets, including alpha-adducin (ADD1), cyclin D1 (CCND1), and p53. Using luciferase promoter activity assay combined with site-directed mutagenesis and sequential chromatin immunoprecipitation-PCR assay, we found that ZNF322A could form a complex with c-Jun and cooperatively activate ADD1 and CCND1 but repress p53 gene transcription by recruiting differential chromatin modifiers, such as histone deacetylase 3, in an AP-1 element dependent manner. Reconstitution experiments indicated that CCND1 and p53 were important to ZNF322A-mediated promotion of cell proliferation, whereas ADD1 was necessary for ZNF322A-mediated cell migration and invasion. Our results provide compelling evidence that ZNF322A overexpression transcriptionally dysregulates genes involved in cell growth and motility therefore contributes to lung tumorigenesis and poor prognosis.
Collapse
Affiliation(s)
- J Jen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - L-L Lin
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - H-T Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - S-Y Liao
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - F-Y Lo
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Y-A Tang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - W-C Su
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - R Salgia
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - C-L Hsu
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - H-C Huang
- Institute of Biomedical Informatics and Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, Taiwan
| | - H-F Juan
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Y-C Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
28
|
Elevated ZNF703 Protein Expression Is an Independent Unfavorable Prognostic Factor for Survival of the Patients with Head and Neck Squamous Cell Carcinoma. DISEASE MARKERS 2015; 2015:640263. [PMID: 26063961 PMCID: PMC4429199 DOI: 10.1155/2015/640263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/15/2015] [Indexed: 01/26/2023]
Abstract
AIM Data from The Cancer Genome Atlas (TCGA) show that the ZNF703 gene amplifies and overexpresses in head and neck squamous cell carcinomas (HNSCC). However, the clinical relevance of this observation in HNSCC is unclear. The purpose of this study was to clarify the expression of ZNF703 protein and its prognostic effect on HNSCC. METHODS Two hundred ten HNSCC patients from Sun Yat-Sen University Cancer Center with complete survival follow-up were included in this study. Tumor samples from primary sites were collected. The expression of the ZNF703 protein was tested by immunohistochemistry (IHC). RESULTS The high expression of ZNF703 in HNSCC tumor tissues was significantly higher than that of the matched noncancerous tissues (48.6% versus 11.6%, P < 0.001). ZNF703 overexpression was correlated with tumor position (laryngeal carcinoma) and recurrence (all P < 0.05). Multivariate analysis revealed that ZNF703 protein overexpression was an independent prognostic factor (P = 0.022, hazard ratio = 1.635, 95% CI 1.073-2.493) in HNSCC patients. CONCLUSION ZNF703 overexpression is associated with adverse prognosis in HNSCC, which might be a novel biomarker of HNSCC.
Collapse
|
29
|
Zong D, Yin L, Zhong Q, Guo WJ, Xu JH, Jiang N, Lin ZR, Li MZ, Han P, Xu L, He X, Zeng MS. ZNF488 Enhances the Invasion and Tumorigenesis in Nasopharyngeal Carcinoma Via the Wnt Signaling Pathway Involving Epithelial Mesenchymal Transition. Cancer Res Treat 2015; 48:334-44. [PMID: 25779368 PMCID: PMC4720103 DOI: 10.4143/crt.2014.311] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/05/2014] [Indexed: 02/06/2023] Open
Abstract
Purpose The purpose of this study was to investigate the function of Zinc finger protein 488 (ZNF488) in nasopharyngeal carcinoma (NPC). Materials and Methods The endogenous expression of ZNF488 in NPC tissues, normal nasopharyngeal epithelium tissues and NPC cell lines were detected by quantitative reverse transcription polymerase chain reaction. ZNF488 over-expressing and knock-down NPC cell line models were established through retroviral vector pMSCV mediated over-expression and small interfering RNA (siRNA) mediated knock-down. The invasion and migration capacities were evaluated by wound healing and transwell invasion assays in ZNF488 over-expressing and control cell lines. Soft-agar colony formation and a xenograft experiment were performed to study tumorigenic ability in vitro and in vivo. Immunofluorescence and western blotting analysis were used to examine protein changes followed by ZNF488 over-expression. Microarray analysis was performed to explore gene expression profilings, while luciferase reporter assay to evaluate the transcriptive activity of Tcf/Lef. Results ZNF488 was over-expressed in NPC tissues compared with normal tissues, especially higher in 5-8F and S18, which are well-established high metastatic NPC clones. Functional studies indicate that over-expression of ZNF488 provokes invasion, whereas knock-down of ZNF488 alleviates invasive capability. Moreover, over-expression of ZNF488 promotes NPC tumor growth both in vitro and in vivo. Our data further show that over-expression of ZNF488 induces epithelial mesenchymal transition (EMT) by activating the WNT/β-catenin signaling pathway. Conclusion Our data strongly suggest that ZNF488 acts as an oncogene, promoting invasion and tumorigenesis by activating the Wnt/β-catenin pathway to induce EMT in NPC.
Collapse
Affiliation(s)
- Dan Zong
- Jiangsu Cancer Hospital, Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Li Yin
- Jiangsu Cancer Hospital, Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wen-Jie Guo
- Jiangsu Cancer Hospital, Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Jian-Hua Xu
- Jiangsu Cancer Hospital, Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Ning Jiang
- Jiangsu Cancer Hospital, Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Zhi-Rui Lin
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Man-Zhi Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ping Han
- Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lin Xu
- Jiangsu Cancer Hospital, Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Xia He
- Jiangsu Cancer Hospital, Nanjing Medical University, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
30
|
Baykara O, Bakir B, Buyru N, Kaynak K, Dalay N. Amplification of chromosome 8 genes in lung cancer. J Cancer 2015; 6:270-5. [PMID: 25663945 PMCID: PMC4317763 DOI: 10.7150/jca.10638] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/18/2014] [Indexed: 12/25/2022] Open
Abstract
Chromosomal alterations are frequent events in lung carcinogenesis and usually display regions of focal amplification containing several overexpressed oncogenes. Although gains and losses of chromosomal loci have been reported copy number changes of the individual genes have not been analyzed in lung cancer. In this study 22 genes were analyzed by MLPA in tumors and matched normal tissue samples from 82 patients with non-small cell lung cancer. Gene amplifications were observed in 84% of the samples. Chromosome 8 was found to harbor the most frequent copy number alterations. The most frequently amplified genes were ZNF703, PRDM14 and MYC on chromosome 8 and the BIRC5 gene on chromosome 17. The frequency of deletions were much lower and the most frequently deleted gene was ADAM9. Amplification of the ZNF703, PRDM14 and MYC genes were highly correlated suggesting that the genes displaying high copy number changes on chromosome 8 collaborate during lung carcinogenesis.
Collapse
Affiliation(s)
- Onur Baykara
- 1. Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University, Turkey
| | - Burak Bakir
- 1. Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University, Turkey
| | - Nur Buyru
- 1. Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University, Turkey
| | - Kamil Kaynak
- 2. Department of Chest Surgery, Cerrahpasa Medical Faculty, Istanbul University, Turkey
| | - Nejat Dalay
- 3. Department of Basic Oncology, I.U. Oncology Institute, Istanbul University, Turkey
| |
Collapse
|