1
|
Wang Y, Gong Y, Li X, Long W, Zhang J, Wu J, Dong Y. Targeting the ZNF-148/miR-335/SOD2 signaling cascade triggers oxidative stress-mediated pyroptosis and suppresses breast cancer progression. Cancer Med 2023; 12:21308-21320. [PMID: 37909239 PMCID: PMC10726847 DOI: 10.1002/cam4.6673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 09/24/2023] [Accepted: 10/20/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND The implication of zinc finger protein 148 (ZNF-148) in pathophysiology of most human cancers has been reported; however, the biological functions of ZNF-148 in breast cancer remain unclear. This study sought to elucidate the potential molecular mechanism of ZNF-148 on breast cancer pathology. METHODS ZNF148 expression was tested in breast cancer tissues and cells. Then, cells were transfected with ZNF-148 overexpression or downregulation vector, and the cell proliferation, pyroptosis, apoptosis, and reactive oxygen species (ROS) production were analyzed by MTT, western blot, flow cytometry, and immunofluorescence staining, respectively. Tumor-bearing nude mouse was used to evaluate tumorigenesis of ZNF-148. Mechanisms underpinning ZNF-148 were examined using bioinformatics and luciferase assays. RESULTS We found that ZNF-148 was upregulated in breast cancer tissues and cell lines. Knockdown of ZNF-148 suppressed malignant phenotypes, including cell proliferation, epithelial-mesenchymal transition, and tumorigenesis in vitro and in vivo, while ZNF-148 overexpression had the opposite effects. Further experiments showed that ZNF-148 deficiency promoted ROS production and triggered both apoptotic and pyroptotic cell death, which were restored by cotreating cells with ROS scavengers. A luciferase reporter assay revealed that miR-335 was the downstream target of ZNF-148 and that overexpressed ZNF-148 increased superoxide dismutase 2 (SOD2) expression by sponging miR-335. In parallel, both miR-335 downregulation and SOD2 overexpression abrogated the antitumor effects of ZNF-148 deficiency on proliferation and pyroptosis in breast cancer cells. CONCLUSIONS Our findings indicated that ZNF-148 promotes breast cancer progression by triggering miR-335/SOD2/ROS-mediated pyroptotic cell death and aid the identification of potential therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Yanmei Wang
- Department of Breast SurgeryFirst affiliated hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| | - Yansi Gong
- Department of Breast SurgeryFirst affiliated hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| | - Xuesha Li
- Department of Breast SurgeryFirst affiliated hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| | - Weizhao Long
- Department of Breast SurgeryFirst affiliated hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| | - Jiayu Zhang
- Department of Breast SurgeryFirst affiliated hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| | - Jiefang Wu
- School of MedicineYunnan UniversityKunmingPeople's Republic of China
| | - Yilong Dong
- School of MedicineYunnan UniversityKunmingPeople's Republic of China
| |
Collapse
|
2
|
Kim M, Singh M, Lee BK, Hibbs M, Richardson K, Ellies L, Wintle L, Stuart LM, Wang JY, Voon DC, Blancafort P, Wang J, Kim J, Leedman PJ, Woo AJ. A MYC-ZNF148-ID1/3 regulatory axis modulating cancer stem cell traits in aggressive breast cancer. Oncogenesis 2022; 11:60. [PMID: 36207293 PMCID: PMC9546828 DOI: 10.1038/s41389-022-00435-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
The MYC proto-oncogene (MYC) is one of the most frequently overexpressed genes in breast cancer that drives cancer stem cell-like traits, resulting in aggressive disease progression and poor prognosis. In this study, we identified zinc finger transcription factor 148 (ZNF148, also called Zfp148 and ZBP-89) as a direct target of MYC. ZNF148 suppressed cell proliferation and migration and was transcriptionally repressed by MYC in breast cancer. Depletion of ZNF148 by short hairpin RNA (shRNA) and CRISPR/Cas9 increased triple-negative breast cancer (TNBC) cell proliferation and migration. Global transcriptome and chromatin occupancy analyses of ZNF148 revealed a central role in inhibiting cancer cell de-differentiation and migration. Mechanistically, we identified the Inhibitor of DNA binding 1 and 3 (ID1, ID3), drivers of cancer stemness and plasticity, as previously uncharacterized targets of transcriptional repression by ZNF148. Silencing of ZNF148 increased the stemness and tumorigenicity in TNBC cells. These findings uncover a previously unknown tumor suppressor role for ZNF148, and a transcriptional regulatory circuitry encompassing MYC, ZNF148, and ID1/3 in driving cancer stem cell traits in aggressive breast cancer.
Collapse
Affiliation(s)
- Mijeong Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Manjot Singh
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, 6027, Australia
| | - Bum-Kyu Lee
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY, 12144, USA
| | - Moira Hibbs
- RPH Research Centre, Royal Perth Hospital, Perth, WA, 6000, Australia
| | - Kirsty Richardson
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
| | - Lesley Ellies
- Division of Pharmacology and Toxicology, School of Biomedical Sciences, The University of Western Australia, Perth, WA, 6000, Australia
| | - Larissa Wintle
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
| | - Lisa M Stuart
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
| | - Jenny Y Wang
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Dominic C Voon
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, 920-1192, Japan
- Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Pilar Blancafort
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
- School of Human Sciences, The University of Western Australia, Perth, WA, 6000, Australia
- The Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Jianlong Wang
- Department of Medicine, Columbia Center for Human Development, Columbia Stem Cell Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Peter J Leedman
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia.
| | - Andrew J Woo
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia.
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, 6027, Australia.
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, 6000, Australia.
| |
Collapse
|
3
|
Emfinger CH, de Klerk E, Schueler KL, Rabaglia ME, Stapleton DS, Simonett SP, Mitok KA, Wang Z, Liu X, Paulo JA, Yu Q, Cardone RL, Foster HR, Lewandowski SL, Perales JC, Kendziorski CM, Gygi SP, Kibbey RG, Keller MP, Hebrok M, Merrins MJ, Attie AD. β Cell-specific deletion of Zfp148 improves nutrient-stimulated β cell Ca2+ responses. JCI Insight 2022; 7:e154198. [PMID: 35603790 PMCID: PMC9220824 DOI: 10.1172/jci.insight.154198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 04/20/2022] [Indexed: 12/05/2022] Open
Abstract
Insulin secretion from pancreatic β cells is essential for glucose homeostasis. An insufficient response to the demand for insulin results in diabetes. We previously showed that β cell-specific deletion of Zfp148 (β-Zfp148KO) improves glucose tolerance and insulin secretion in mice. Here, we performed Ca2+ imaging of islets from β‑Zfp148KO and control mice fed both a chow and a Western-style diet. β-Zfp148KO islets demonstrated improved sensitivity and sustained Ca2+ oscillations in response to elevated glucose levels. β-Zfp148KO islets also exhibited elevated sensitivity to amino acid-induced Ca2+ influx under low glucose conditions, suggesting enhanced mitochondrial phosphoenolpyruvate-dependent (PEP-dependent), ATP-sensitive K+ channel closure, independent of glycolysis. RNA-Seq and proteomics of β-Zfp148KO islets revealed altered levels of enzymes involved in amino acid metabolism (specifically, SLC3A2, SLC7A8, GLS, GLS2, PSPH, PHGDH, and PSAT1) and intermediary metabolism (namely, GOT1 and PCK2), consistent with altered PEP cycling. In agreement with this, β-Zfp148KO islets displayed enhanced insulin secretion in response to l-glutamine and activation of glutamate dehydrogenase. Understanding pathways controlled by ZFP148 may provide promising strategies for improving β cell function that are robust to the metabolic challenge imposed by a Western diet.
Collapse
Affiliation(s)
| | | | - Kathryn L. Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mary E. Rabaglia
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Donnie S. Stapleton
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Shane P. Simonett
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kelly A. Mitok
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ziyue Wang
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Xinyue Liu
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Qing Yu
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca L. Cardone
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, Connecticut, USA
| | - Hannah R. Foster
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sophie L. Lewandowski
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - José C. Perales
- Department of Physiological Sciences, School of Medicine, University of Barcelona, L’Hospitalet del Llobregat, Barcelona, Spain
| | - Christina M. Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard G. Kibbey
- Department of Internal Medicine (Endocrinology), Yale University, New Haven, Connecticut, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, USA
| | - Mark P. Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Matthew J. Merrins
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Alan D. Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
4
|
Zhang MF, Li QL, Yang YF, Cao Y, Zhang CZ. FMNL1 Exhibits Pro-Metastatic Activity via CXCR2 in Clear Cell Renal Cell Carcinoma. Front Oncol 2020; 10:564614. [PMID: 33324547 PMCID: PMC7726248 DOI: 10.3389/fonc.2020.564614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022] Open
Abstract
Formin-like (FMNL) proteins are responsible for cytoskeletal remodeling and have been implicated in the progression and spread of human cancers. Yet the clinical significance and biological function of FMNL1 in clear cell renal cell carcinoma (ccRCC) remain unclear. In this study, the expression of FMNL1 in ccRCC and its clinical value were determined by tissue microarray-based IHC and statistical analyses. The role of FMNL1 in ccRCC metastasis and the underlying mechanism were investigated via in vitro and in vivo models using gene regulation detection, ChIP, Luciferase reporter assays, and rescue experiments. We show that FMNL1 is upregulated in ccRCC and exhibits pro-metastatic activity via induction of CXCR2. High expression of FMNL1 is significantly correlated with advanced tumor stage, higher pathological tumor grade, tumor metastasis, and unfavorable prognosis in two independent cohorts containing over 800 patients with ccRCC. The upregulation of FMNL1 in ccRCC is mediated by the loss of GATA3. Ectopic expression of FMNL1 promotes, whereas FMNL1 depletion inhibits cell migration in vitro and tumor metastasis in vivo. The FMNL1-enhanced cell mobility is markedly attenuated by the knockdown of CXCR2. Further studies demonstrate that FMNL1 increases the expression of CXCR2 via HDAC1. In clinical samples, FMNL1 expression is positively associated with CXCR2, and is negatively connected to GATA3 expression. Collectively, our data suggest FMNL1 serve as a potential prognostic factor and function as an oncogene. The axis of GATA3/FMNL1/CXCR2 may present a promising therapeutic target for tumor metastasis in ccRCC.
Collapse
Affiliation(s)
- Mei-Fang Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qiu-Li Li
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yu-Feng Yang
- Department of Pathology, Dongguan Third People's Hospital, Dongguan, China
| | - Yun Cao
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chris Zhiyi Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
5
|
Evelönn EA, Landfors M, Haider Z, Köhn L, Ljungberg B, Roos G, Degerman S. DNA methylation associates with survival in non-metastatic clear cell renal cell carcinoma. BMC Cancer 2019; 19:65. [PMID: 30642274 PMCID: PMC6332661 DOI: 10.1186/s12885-019-5291-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the most common subtype among renal cancer and is associated with poor prognosis if metastasized. Up to one third of patients with local disease at diagnosis will develop metastasis after nephrectomy, and there is a need for new molecular markers to identify patients with high risk of tumor progression. In the present study, we performed genome-wide promoter DNA methylation analysis at diagnosis to identify DNA methylation profiles associated with risk for progress. METHOD Diagnostic tissue samples from 115 ccRCC patients were analysed by Illumina HumanMethylation450K arrays and methylation status of 155,931 promoter associated CpGs were related to genetic aberrations, gene expression and clinicopathological parameters. RESULTS The ccRCC samples separated into two clusters (cluster A/B) based on genome-wide promoter methylation status. The samples in these clusters differed in tumor diameter (p < 0.001), TNM stage (p < 0.001), morphological grade (p < 0.001), and patients outcome (5 year cancer specific survival (pCSS5yr) p < 0.001 and cumulative incidence of progress (pCIP5yr) p < 0.001. An integrated genomic and epigenomic analysis in the ccRCCs, revealed significant correlations between the total number of genetic aberrations and total number of hypermethylated CpGs (R = 0.435, p < 0.001), and predicted mitotic age (R = 0.407, p < 0.001). We identified a promoter methylation classifier (PMC) panel consisting of 172 differently methylated CpGs accompanying progress of disease. Classifying non-metastatic patients using the PMC panel showed that PMC high tumors had a worse prognosis compared with the PMC low tumors (pCIP5yr 38% vs. 8%, p = 0.001), which was confirmed in non-metastatic ccRCCs in the publically available TCGA-KIRC dataset (pCIP5yr 39% vs. 16%, p < 0.001). CONCLUSION DNA methylation analysis at diagnosis in ccRCC has the potential to improve outcome-prediction in non-metastatic patients at diagnosis.
Collapse
Affiliation(s)
- Emma Andersson Evelönn
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| | - Mattias Landfors
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| | - Zahra Haider
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| | - Linda Köhn
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and andrology, Umeå University, Umeå, Sweden
| | - Göran Roos
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| | - Sofie Degerman
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| |
Collapse
|
6
|
Guo XB, Huang B, Pan YH, Su SG, Li Y. ESCO2 inhibits tumor metastasis via transcriptionally repressing MMP2 in colorectal cancer. Cancer Manag Res 2018; 10:6157-6166. [PMID: 30538563 PMCID: PMC6257866 DOI: 10.2147/cmar.s181265] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background Establishment of cohesion 1 homolog 2 (ESCO2) plays important roles in the regulation of cohesion and genomic stability and has been implicated in human cancers. Yet, its clinical significance and biological function in colorectal cancer (CRC) are unknown. Methods The expression of ESCO2 was examined by quantitative real-time PCR, Western blot, and immunohistochemistry. The role of ESCO2 in the tumor metastasis of CRC and the related mechanisms were investigated using in vitro and in vivo models. Results In this study, we show that low expression of ESCO2 in CRC was closely correlated with lymphatic and distant metastasis. Patients with low ESCO2 expression experienced shorter overall survival and disease-free survival in two independent cohorts containing a total of 587 CRC cases. ESCO2 overexpression suppressed, whereas ESCO2 knockdown promoted cell migration in vitro and tumor metastasis in vivo via modulation of epithelial–mesenchymal transition (EMT) process. Mechanistically, ESCO2 inhibited the transcriptional activity of MMP2 promoter to downregulate its expression. Reexpression of MMP2 partially attenuated the ESCO2-mediated malignant phenotypes. Conclusion Collectively, our data suggest that ESCO2 serves as a potential prognostic factor and exerts antimetastatic activity in CRC.
Collapse
Affiliation(s)
- Xiong-Bo Guo
- Department of General Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Huang
- Department of General Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Ying-Hua Pan
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shu-Guang Su
- Department of Pathology, The Affiliated Hexian Memorial Hospital of Southern Medical University, Guangzhou, China,
| | - Yan Li
- Department of Pathology, The Affiliated Hexian Memorial Hospital of Southern Medical University, Guangzhou, China,
| |
Collapse
|
7
|
Wang QS, Chen C, Zhan J, Fang XF, Chen GG, Yang SL, Chen RW, Tong F, Hu JL. Peritumoral overexpression of ZBP-89 is associated with unfavorable disease-free survival rates in patients with hepatocellular carcinoma following hepatectomy. Oncol Lett 2018; 15:7828-7836. [PMID: 29731904 PMCID: PMC5920541 DOI: 10.3892/ol.2018.8353] [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: 08/29/2017] [Accepted: 02/09/2018] [Indexed: 11/05/2022] Open
Abstract
Previous studies have revealed that the peritumoral environment has a profound influence on tumor initiation and progression. Zinc-binding protein-89 (ZBP-89) has been observed to be involved with tumor development, recurrence, and metastasis. High intratumoral expression of ZBP-89 has been associated with improved prognosis in several tumor types. However, the prognostic values of peritumoral expression of ZBP-89 remain to be elucidated in patients with hepatocellular carcinoma (HCC) following curative resection. In the present study, peritumoral ZBP-89 expression was examined using immunohistochemistry in 102 HCC patients who had received curative hepatectomy. Expression of ZBP-89 protein was positive in 66.3% of the peritumoral samples from 102 HCC patients. HCC patients with high peritumoral ZBP-89 expression exhibited significantly shorter disease-free survival (DFS) times (P=0.012) than those patients with low peritumoral ZBP-89 expression. Additionally, high ZBP-89 expression in peritumoral HCC tissue was positively associated with the presence of liver cirrhosis. Univariate and multivariate Cox proportional hazard regression analyses demonstrated that albumin levels ≤35 g/l, multiple tumors, tumor sizes ≥5 cm, and macroscopic vascular invasion may serve as independent prognostic factors for overall survival (OS) [hazard ratio (HR)=2.031; P=0.014] in patients with HCC. The multivariate Cox regression model identified that high ZBP-89 expression, multiple tumors and macroscopic vascular invasion were independent prognostic factors for shorter DFS durations. High expression of ZBP-89 in peritumoral HCC tissues was associated with a shorter DFS in HCC patients following curative hepatectomy. Additionally, high ZBP-89 expression in peritumoral HCC tissue was positively associated with the presence of liver cirrhosis in HCC patients, indicating that cirrhosis accompanied by high ZBP-89 expression may be a contributing factor to the poor prognosis of patients with HCC. Therefore, peritumoral ZBP-89 expression may be a good prognostic marker to predict DFS time in HCC patients following curative hepatectomy and may provide novel insights into the molecular mechanisms of HCC initiation.
Collapse
Affiliation(s)
- Qiu-Shuang Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Chen Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing Zhan
- Department of Gastroenterology and Hepatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xie-Fan Fang
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - George G Chen
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, SAR, P.R. China
| | - Sheng-Li Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ren-Wang Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Fan Tong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jian-Li Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
8
|
Nilton A, Sayin VI, Zou ZV, Sayin SI, Bondjers C, Gul N, Agren P, Fogelstrand P, Nilsson O, Bergo MO, Lindahl P. Targeting Zfp148 activates p53 and reduces tumor initiation in the gut. Oncotarget 2018; 7:56183-56192. [PMID: 27487143 PMCID: PMC5302905 DOI: 10.18632/oncotarget.10899] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/13/2016] [Indexed: 12/30/2022] Open
Abstract
The transcription factor Zinc finger protein 148 (Zfp148, ZBP-89, BFCOL, BERF1, htβ) interacts physically with the tumor suppressor p53, but the significance of this interaction is not known. We recently showed that knockout of Zfp148 in mice leads to ectopic activation of p53 in some tissues and cultured fibroblasts, suggesting that Zfp148 represses p53 activity. Here we hypothesize that targeting Zfp148 would unleash p53 activity and protect against cancer development, and test this idea in the APCMin/+ mouse model of intestinal adenomas. Loss of one copy of Zfp148 markedly reduced tumor numbers and tumor-associated intestinal bleedings, and improved survival. Furthermore, after activation of β-catenin-the initiating event in colorectal cancer-Zfp148 deficiency activated p53 and induced apoptosis in intestinal explants of APCMin/+ mice. The anti-tumor effect of targeting Zfp148 depended on p53, as Zfp148 deficiency did not affect tumor numbers in APCMin/+ mice lacking one or both copies of Trp53. The results suggest that Zfp148 controls the fate of newly transformed intestinal tumor cells by repressing p53 and that targeting Zfp148 might be useful in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Anna Nilton
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Volkan I Sayin
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden.,Department of Biochemistry, Institute of Biomedicine, Gothenburg, Sweden
| | - Zhiyuan V Zou
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Sama I Sayin
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Cecilia Bondjers
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Nadia Gul
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Pia Agren
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Per Fogelstrand
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Ola Nilsson
- Sahlgrenska Cancer Center, Institute of Biomedicine, Department of Pathology and Genetics, Gothenburg, Sweden
| | - Martin O Bergo
- Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Per Lindahl
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden.,Department of Biochemistry, Institute of Biomedicine, Gothenburg, Sweden
| |
Collapse
|
9
|
Fouani L, Menezes SV, Paulson M, Richardson DR, Kovacevic Z. Metals and metastasis: Exploiting the role of metals in cancer metastasis to develop novel anti-metastatic agents. Pharmacol Res 2017; 115:275-287. [DOI: 10.1016/j.phrs.2016.12.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 01/06/2023]
|
10
|
Essien BE, Sundaresan S, Ocadiz-Ruiz R, Chavis A, Tsao AC, Tessier AJ, Hayes MM, Photenhauer A, Saqui-Salces M, Kang AJ, Shah YM, Győrffy B, Merchant JL. Transcription Factor ZBP-89 Drives a Feedforward Loop of β-Catenin Expression in Colorectal Cancer. Cancer Res 2016; 76:6877-6887. [PMID: 27758879 DOI: 10.1158/0008-5472.can-15-3150] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 09/12/2016] [Accepted: 09/28/2016] [Indexed: 01/24/2023]
Abstract
In colorectal cancer, APC-mediated induction of unregulated cell growth involves posttranslational mechanisms that prevent proteasomal degradation of proto-oncogene β-catenin (CTNNB1) and its eventual translocation to the nucleus. However, about 10% of colorectal tumors also exhibit increased CTNNB1 mRNA. Here, we show in colorectal cancer that increased expression of ZNF148, the gene coding for transcription factor ZBP-89, correlated with reduced patient survival. Tissue arrays showed that ZBP-89 protein was overexpressed in the early stages of colorectal cancer. Conditional deletion of Zfp148 in a mouse model of Apc-mediated intestinal polyps demonstrated that ZBP-89 was required for polyp formation due to induction of Ctnnb1 gene expression. Chromatin immunoprecipitation (ChIP) and EMSA identified a ZBP-89-binding site in the proximal promoter of CTNNB1 Reciprocally, siRNA-mediated reduction of CTNNB1 expression also decreased ZBP-89 protein. ChIP identified TCF DNA binding sites in the ZNF148 promoter through which Wnt signaling regulates ZNF148 gene expression. Suppression of either ZNF148 or CTNNB1 reduced colony formation in WNT-dependent, but not WNT-independent cell lines. Therefore, the increase in intracellular β-catenin protein initiated by APC mutations is sustained by ZBP-89-mediated feedforward induction of CTNNB1 mRNA. Cancer Res; 76(23); 6877-87. ©2016 AACR.
Collapse
Affiliation(s)
- Bryan E Essien
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Sinju Sundaresan
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Ramon Ocadiz-Ruiz
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Aaron Chavis
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Amy C Tsao
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Arthur J Tessier
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Michael M Hayes
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Amanda Photenhauer
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Milena Saqui-Salces
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Anthony J Kang
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Balazs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Budapest, Hungary
| | - Juanita L Merchant
- Department of Internal Medicine-Gastroenterology, University of Michigan, Ann Arbor, Michigan. .,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
11
|
Liu GH, Chen J, Ji ZG, Zhou L. Expression of Neural Wiskott-Aldrich Syndrome Protein in Clear Cell Renal Cell Carcinoma and Its Correlation with Clinicopathological Features. Urol Int 2014; 95:79-85. [PMID: 25115631 DOI: 10.1159/000365595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/01/2014] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Neural Wiskott-Aldrich syndrome protein (N-WASP) expression is associated with tumor cell invasion and migration. However, its expression status in clear cell renal cell carcinoma (CCRCC) remains unclear. We examined the level of N-WASP in CCRCC and its association with clinicopathological features characteristic. MATERIALS AND METHODS 73 CCRCC patients who underwent radical nephrectomy or partial nephrectomy were enrolled. Immunohistochemical staining for N-WASP was performed on tissue microarrays constructed from tumor and para-tumor tissue obtained from these patients. The difference in N-WASP expression between tumor tissue and adjacent normal renal tissue was examined. Correlations between N-WASP expression in the tumor and clinicopathological parameters were analyzed and the relationship between N-WASP expression and overall survival also assessed. Uni- and multivariate survival analyses were performed. RESULTS N-WASP expression was significantly reduced in tumor tissues and was significantly related to the histological grade of CCRCC. A higher level of N-WASP expression in the tumor was associated with relatively poor survival in CCRCC patients. The level of N-WASP expression, age at time of surgery, and histological grade were all responsible for clinical outcome in CCRCC patients. N-WASP was an independent predictor for overall survival. CONCLUSIONS N-WASP was downregulated in CCRCC and could serve as a prognostic biomarker for predicting clinical outcome of CCRCC.
Collapse
Affiliation(s)
- Guang-Hua Liu
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | | | | | | |
Collapse
|
12
|
Yan SM, Wu HN, He F, Hu XP, Zhang ZY, Huang MY, Wu X, Huang CY, Li Y. High expression of zinc-binding protein-89 predicts decreased survival in esophageal squamous cell cancer. Ann Thorac Surg 2014; 97:1966-73. [PMID: 24698505 DOI: 10.1016/j.athoracsur.2014.01.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/17/2014] [Accepted: 01/28/2014] [Indexed: 02/05/2023]
Abstract
BACKGROUND Zinc-binding protein-89 (ZBP-89), a Krüppel-type four-zinc finger transcription factor, is associated with many cellular functions, including cell growth, differentiation, and apoptosis. It has been reported to be involved in several human cancers. However, ZBP-89 expression pattern and its clinical significance have not yet been investigated in esophageal squamous cell cancer. METHODS In this study, immunostaining was performed to detect ZBP-89 expression in esophageal squamous cell cancer, and then the correlations between ZBP-89 expression and both clinicopathologic variables and overall survival were analyzed. RESULTS Compared with adjacent normal tissues, ZBP-89 expression was significantly upregulated in esophageal squamous cell cancer tissues. Increased ZBP-89 expression was associated with N category (p = 0.009) and TNM stage (p = 0.023). Patients with high expression of ZBP-89 demonstrated shortened overall survival compared with those with low expression of ZBP-89 (mean overall survival, 56.961 months versus 76.029 months; p < 0.001). Multivariate Cox regression analysis indicated that ZBP-89 expression had a significant, independent predictive value for survival of esophageal squamous cell cancer (relative risk, 1.581; p = 0.024). CONCLUSIONS Our data show that increased expression of ZBP-89 is associated with poor prognosis for esophageal squamous cell cancer patients and may act as a novel, useful, and independent prognostic indicator for esophageal squamous cell cancer. Further studies are warranted.
Collapse
Affiliation(s)
- Shu-Mei Yan
- State Key Laboratory of Oncology in South China and Department of Pathology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Hui-Ni Wu
- School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Fan He
- Department of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiao-peng Hu
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhi-yi Zhang
- State Key Laboratory of Oncology in South China and Department of Pathology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Ma-Yan Huang
- State Key Laboratory of Oncology in South China and Department of Pathology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Xiao Wu
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, People's Republic of China
| | - Chun-yu Huang
- Department of Endoscopy, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yong Li
- State Key Laboratory of Oncology in South China and Department of Pathology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China.
| |
Collapse
|
13
|
Li Y, Tan BB, Zhao Q, Fan LQ, Liu Y, Wang D. Regulatory mechanism of ZNF139 in multi-drug resistance of gastric cancer cells. Mol Biol Rep 2014; 41:3603-10. [PMID: 24515389 DOI: 10.1007/s11033-014-3224-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 01/29/2014] [Indexed: 11/26/2022]
Abstract
Our previous study found increased zinc finger protein 139 (ZNF139) expression in gastric cancer (GC) cells. Purpose of the study is to further clarify the role and mechanism of ZNF139 in multi-drug resistance (MDR) of GC cells. MTT assay, RT-PCR, Western blotting were employed to detect susceptibility of GC cells to chemotherapeutic agents (5-FU, L-OHP) in vitro, and expressions of ZNF139 and MDR associated genes MDR1/P-gp, MRP1, Bcl-2, Bax were also detected. siRNA specific to ZNF139 was transfected into MKN28 cells, then chemosensitivity of GC cells as well as changes of ZNF139 and MDR associated genes were detected. It's found the inhibition rate of 5-FU, L-OHP to well-differentiated GC tissues and cell line was lower than that in the poorly differentiated tissues and cell line; expressions of ZNF139 and MDR1/P-gp, MRP1 and Bcl-2 in well-differentiated GC tissues and cell line MKN28 were higher, while Bax expression was lower. After ZNF139-siRNA was transfected into MKN28, ZNF139 expression in GC cells was inhibited by 90%; inhibition rate of 5-FU, L-OHP to tumor cells increased, and expressions of MDR1/P-gp, MRP1 and Bcl-2 were down-regulated, while Bax was up-regulated. ZNF139 was involved in GC MDR by promoting expressions of MDR1/P-gp, MRP1 and Bcl-2 and inhibiting Bax simultaneously.
Collapse
Affiliation(s)
- Yong Li
- Department of General Surgery, The Fourth Affiliated Hospital, Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China,
| | | | | | | | | | | |
Collapse
|