1
|
Orang A, Dredge BK, Liu CY, Bracken JM, Chen CH, Sourdin L, Whitfield HJ, Lumb R, Boyle ST, Davis MJ, Samuel MS, Gregory PA, Khew-Goodall Y, Goodall GJ, Pillman KA, Bracken CP. Basonuclin-2 regulates extracellular matrix production and degradation. Life Sci Alliance 2023; 6:e202301984. [PMID: 37536977 PMCID: PMC10400885 DOI: 10.26508/lsa.202301984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023] Open
Abstract
Epithelial-mesenchymal transition is essential for tissue patterning and organization. It involves both regulation of cell motility and alterations in the composition and organization of the ECM-a complex environment of proteoglycans and fibrous proteins essential for tissue homeostasis, signaling in response to chemical and biomechanical stimuli, and is often dysregulated under conditions such as cancer, fibrosis, and chronic wounds. Here, we demonstrate that basonuclin-2 (BNC2), a mesenchymal-expressed gene, that is, strongly associated with cancer and developmental defects across genome-wide association studies, is a novel regulator of ECM composition and degradation. We find that at endogenous levels, BNC2 controls the expression of specific collagens, matrix metalloproteases, and other matrisomal components in breast cancer cells, and in fibroblasts that are primarily responsible for the production and processing of the ECM within the tumour microenvironment. In so doing, BNC2 modulates the motile and invasive properties of cancers, which likely explains the association of high BNC2 expression with increasing cancer grade and poor patient prognosis.
Collapse
Affiliation(s)
- Ayla Orang
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
| | - B Kate Dredge
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
| | - Chi Yau Liu
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
| | - Julie M Bracken
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
| | - Chun-Hsien Chen
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
| | - Laura Sourdin
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
| | - Holly J Whitfield
- Division of Bioinformatics, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Rachael Lumb
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
| | - Sarah T Boyle
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
| | - Melissa J Davis
- South Australian ImmunogGENomics Cancer Institute, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Division of Bioinformatics, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
- Department of Clinical Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
- Fraser Institute, University of Queensland, Wooloongabba, Australia
| | - Michael S Samuel
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
- Department of Medicine and School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
- Department of Medicine and School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Gregory J Goodall
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
- Department of Medicine and School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Katherine A Pillman
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
- Department of Medicine and School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Cameron P Bracken
- Centre for Cancer Biology, An Alliance of SA Pathology and University of South Australia, Adelaide, Australia
- Department of Medicine and School of Biological Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
2
|
Jiang Q, Wang H, Yuan D, Qian X, Ma X, Yan M, Xing W. Circular_0086414 induces SPARC like 1 ( SPARCL1) production to inhibit esophageal cancer cell proliferation, invasion and glycolysis and induce cell apoptosis by sponging miR-1290. Bioengineered 2022; 13:12099-12114. [PMID: 35549806 PMCID: PMC9275914 DOI: 10.1080/21655979.2022.2073114] [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] [Indexed: 11/28/2022] Open
Abstract
Circular RNA (circRNA) plays an important role in cancer progression. Here, we investigated the function of circ_0086414 in the malignant progression of esophageal cancer (EC). RNA expression of circ_0086414, microRNA-1290 (miR-1290), and SPARC like 1 (SPARCL1) was detected by quantitative real-time polymerase chain reaction. The protein levels of N-cadherin, E-cadherin, and SPARCL1 were checked by Western blotting analysis. Cell proliferation was investigated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 5-Ethynyl-29-deoxyuridine (EdU), and cell colony formation assays. Cell invasion and apoptosis were analyzed by transwell invasion assay and flow cytometry analysis, respectively. Glycolysis was evaluated by analyzing glucose consumption and lactate production. In an xenograft mouse model, the effect of circ_0086414 on tumor tumorigenesis was investigated. The interactions among circ_0086414, miR-1290, and SPARCL1 were identified by dual-luciferase reporter and RNA pull-down assays. Results showed that circ_0086414 and SPARCL1 expression were significantly downregulated, while miR-1290 was upregulated in EC tissues and cells. EC patients with low circ_0086414 expression had a poor prognosis. Increasing circ_0086414 expression led to decreased EC cell proliferation, invasion and glycolysis and increased cell apoptosis, accompanied by a decrease of N-cadherin expression and an increase of E-cadherin expression. Also, the enforced expression of circ_0086414 delayed tumor tumorigenesis. Besides, circ_0086414 acted as a miR-1290 sponge and regulated EC cell processes by binding to the miRNA. MiR-1290 also participated in EC malignant progression through SPARCL1. Further, circ_0086414 stimulated SPARCL1 production by negatively regulating miR-1290. Thus, circ_0086414 inhibited EC cell malignancy through the miR-1290/SPARCL1 pathway, providing a reliable target for the therapy of EC.
Collapse
Affiliation(s)
- Qingfeng Jiang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haoran Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongfeng Yuan
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Qian
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaochao Ma
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ming Yan
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenqun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Li R, Xia Y, Chen X, Li X, Huang G, Peng X, Liu K, Zhang C, Li M, Lin Y, Dong J, Ji L, Lai Y. Identification of a three-miRNA panel in serum for bladder cancer diagnosis by a diagnostic test. Transl Cancer Res 2022; 11:1005-1016. [PMID: 35706801 PMCID: PMC9189164 DOI: 10.21037/tcr-21-2611] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/20/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Bladder cancer (BC) is the tenth most common cancer in the world. Serum microRNA (miRNA) profiles previously have been reported as non-invasive biomarkers in cancer screening. The non-invasive and reliable diagnostic biomarkers are urgently needed for detecting BC, while cystoscopy is invasive. Our study aimed to identify candidate miRNAs in serum as potential diagnostic biomarkers for BC detection. METHODS This study was including the screening stage, training stage, and validation stage with 137 BC patients and 127 healthy controls (HCs). We identified the expression of 28 serum miRNAs from 5 BC pools and 3 HC pools in the initial screening stage. The other 112 BC patients and 112 HCs were randomly divided into training stage with 30 BC patients and 30 HCs and validation stages with 82 BC patients and 82 HCs. These HCs matched BC patients based on age and gender with P value >0.05. Identified dysregulated miRNAs were further confirmed in the training stage, and validation stages by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The diagnostic value of miRNAs was assessed by receiver operating characteristic (ROC) curves and the area under the ROC curve (AUC). Target genes of 3 candidate miRNAs were predicted by bioinformatic analysis. RESULTS Five miRNAs (miR-106a-5p, miR-145-5p, miR-132-3p, miR-7-5p and miR-148b-3p) in serum were obviously dysregulated in BC patients compared to HCs. The ability to diagnose BC of 3 candidate miRNAs was estimated by AUC, with miR-132-3p (AUC =0.781; sensitivity =68.29%, specificity =81.71%), miR-7-5p (AUC =0.778; sensitivity =59.76%, specificity =84.15%) and miR-148b-3p (AUC =0.837; sensitivity =81.71%, specificity =71.95%). Combined application of these candidate miRNAs with parallel test could improve the diagnostic value (AUC =0.922; sensitivity =90.24%, specificity =81.71%). BNC2, GAS7, and NTRK2, considered as target genes of the three-miRNA panel, may play an important role in the process of BC development. CONCLUSIONS A three-miRNA panel in serum was identified for BC diagnosis in our study, which HCs were used for differential diagnosis. The three-miRNA panel (miR-132-3p, miR-7-5p, and miR-148b-3p) might be performed as a non-invasive and convenient diagnostic tool for BC screening and diagnosis.
Collapse
Affiliation(s)
- Rongkang Li
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Yong Xia
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xuan Chen
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Xinji Li
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Guocheng Huang
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Xiqi Peng
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Kaihao Liu
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Chunduo Zhang
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
| | - Mingyang Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yu Lin
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jing Dong
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ling Ji
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yongqing Lai
- Department of Urology, Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| |
Collapse
|
4
|
Winters BR, De Sarkar N, Arora S, Bolouri H, Jana S, Vakar-Lopez F, Cheng HH, Schweizer MT, Yu EY, Grivas P, Lee JK, Kollath L, Holt SK, McFerrin L, Ha G, Nelson PS, Montgomery RB, Wright JL, Lam HM, Hsieh AC. Genomic distinctions between metastatic lower and upper tract urothelial carcinoma revealed through rapid autopsy. JCI Insight 2019; 5:128728. [PMID: 31145100 DOI: 10.1172/jci.insight.128728] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Little is known about the genomic differences between metastatic urothelial carcinoma (LTUC) and upper tract urothelial carcinoma (UTUC). We compare genomic features of primary and metastatic UTUC and LTUC tumors in a cohort of patients with end stage disease. METHODS We performed whole exome sequencing on matched primary and metastatic tumor samples (N=37) from 7 patients with metastatic UC collected via rapid autopsy. Inter- and intra-patient mutational burden, mutational signatures, predicted deleterious mutations, and somatic copy alterations (sCNV) were analyzed. RESULTS We investigated 3 patients with UTUC (3 primary samples, 13 metastases) and 4 patients with LTUC (4 primary samples, 17 metastases). We found that sSNV burden was higher in metastatic LTUC compared to UTUC. Moreover, the APOBEC mutational signature was pervasive in metastatic LTUC and less so in UTUC. Despite a lower overall sSNV burden, UTUC displayed greater inter- and intra-individual genomic distances at the copy number level between primary and metastatic tumors than LTUC. Our data also indicate that metastatic UTUC lesions can arise from small clonal populations present in the primary cancer. Importantly, putative druggable mutations were found across patients with the majority shared across all metastases within a patient. CONCLUSIONS Metastatic UTUC demonstrated a lower overall mutational burden but greater structural variability compared to LTUC. Our findings suggest that metastatic UTUC displays a greater spectrum of copy number divergence from LTUC. Importantly, we identified druggable lesions shared across metastatic samples, which demonstrate a level of targetable homogeneity within individual patients.
Collapse
Affiliation(s)
| | - Navonil De Sarkar
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA.,Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Sonali Arora
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Hamid Bolouri
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Sujata Jana
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Funda Vakar-Lopez
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Heather H Cheng
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Michael T Schweizer
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Evan Y Yu
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Petros Grivas
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA
| | - John K Lee
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA.,Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | - Lisa McFerrin
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Gavin Ha
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peter S Nelson
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA.,Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Robert B Montgomery
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jonathan L Wright
- Department of Urology and.,Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Hung-Ming Lam
- Department of Urology and.,Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Andrew C Hsieh
- Department of Medicine, Division of Oncology, University of Washington School of Medicine, Seattle, Washington, USA.,Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
5
|
Hsu HC, Lapke N, Chen SJ, Lu YJ, Jhou RS, Yeh CY, Tsai WS, Hung HY, Hsieh JCH, Yang TS, Thiam TK, You JF. PTPRT and PTPRD Deleterious Mutations and Deletion Predict Bevacizumab Resistance in Metastatic Colorectal Cancer Patients. Cancers (Basel) 2018; 10:cancers10090314. [PMID: 30200630 PMCID: PMC6162606 DOI: 10.3390/cancers10090314] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/25/2018] [Accepted: 09/03/2018] [Indexed: 12/22/2022] Open
Abstract
Background: Bevacizumab-based regimens are used as standard treatments for colorectal cancer. Unfortunately, there are no established predictive markers for bevacizumab response. Methods: Tumor samples from 36 metastatic colorectal cancer patients treated with bevacizumab plus chemotherapy were analyzed by next-generation sequencing of all coding exons of more than 400 genes. Single gene and signaling pathway analyses were performed to correlate genomic data with response. Results: Among the genes most frequently mutated in our cohort, only mutations in PTPRT, a phosphatase involved in JAK/STAT signaling, were associated with response status, with deleterious mutations being enriched in non-responders. Pathway analysis revealed that deleterious mutations in genes of the JAK/STAT pathway, namely in PTPRT and the related gene PTPRD, correlated with resistance. Mutations in RTK/PI3K/RAS, Wnt and TGFβ pathways did not associate with response. Lack of response was observed in all patients with deleterious mutations or copy number loss of PTPRT/PTPRD (n = 10), compared to only 30.8% (n = 8) of patients without such alterations (relative risk, 3.25; 95% CI, 1.83–5.79, p = 0.0003). Similarly, PTPRT/PTPRD deleterious alterations were associated with shorter progression-free survival, an association that was retained in multivariate analysis (HR, 3.33; 95% CI, 1.47–7.54; p = 0.0038). Conclusion: Deleterious alterations in PTPRT/PTPRD are potential biomarkers for bevacizumab resistance.
Collapse
Affiliation(s)
- Hung-Chih Hsu
- Division of Hematology/Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan City 333, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan.
| | - Nina Lapke
- ACT Genomics, Neihu Dist., Taipei City 114, Taiwan.
| | - Shu-Jen Chen
- ACT Genomics, Neihu Dist., Taipei City 114, Taiwan.
| | - Yen-Jung Lu
- ACT Genomics, Neihu Dist., Taipei City 114, Taiwan.
| | | | - Chien-Yuh Yeh
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan.
- ACT Genomics, Neihu Dist., Taipei City 114, Taiwan.
| | - Wen-Sy Tsai
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan.
- Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 333, Taiwan.
| | - Hsin-Yuan Hung
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan.
- Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 333, Taiwan.
| | - Jason Chia-Hsun Hsieh
- Division of Hematology/Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan City 333, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan.
| | - Tsai-Sheng Yang
- Division of Hematology/Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan City 333, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan.
| | | | - Jeng-Fu You
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan.
- Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City 333, Taiwan.
| |
Collapse
|
6
|
Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2017; 96:98-134. [PMID: 29031806 DOI: 10.1016/j.biocel.2017.10.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Aberrant protein phosphorylation is one of the hallmarks of cancer cells, and in many cases a prerequisite to sustain tumor development and progression. Like protein kinases, protein phosphatases are key regulators of cell signaling. However, their contribution to aberrant signaling in cancer cells is overall less well appreciated, and therefore, their clinical potential remains largely unexploited. In this review, we provide an overview of tumor suppressive protein phosphatases in human cancer. Along their mechanisms of inactivation in defined cancer contexts, we give an overview of their functional roles in diverse signaling pathways that contribute to their tumor suppressive abilities. Finally, we discuss their emerging roles as predictive or prognostic markers, their potential as synthetic lethality targets, and the current feasibility of their reactivation with pharmacologic compounds as promising new cancer therapies. We conclude that their inclusion in clinical practice has obvious potential to significantly improve therapeutic outcome in various ways, and should now definitely be pushed forward.
Collapse
Affiliation(s)
- Bob Meeusen
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium.
| |
Collapse
|
7
|
Cesaratto L, Grisard E, Coan M, Zandonà L, De Mattia E, Poletto E, Cecchin E, Puglisi F, Canzonieri V, Mucignat MT, Zucchetto A, Stocco G, Colombatti A, Nicoloso MS, Spizzo R. BNC2 is a putative tumor suppressor gene in high-grade serous ovarian carcinoma and impacts cell survival after oxidative stress. Cell Death Dis 2016; 7:e2374. [PMID: 27899818 PMCID: PMC5059877 DOI: 10.1038/cddis.2016.278] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 07/04/2016] [Accepted: 07/29/2016] [Indexed: 12/21/2022]
Abstract
Rs3814113 is the single-nucleotide polymorphism (SNP) showing the strongest association with high-grade serous ovarian carcinoma (HGSOC) incidence and is located in an intergenic region about 44 kb downstream of basonuclin 2 (BNC2) gene. Lifetime number of ovulations is associated with increased risk to develop HGSOC, probably because of cell damage of extrauterine Müllerian epithelium by ovulation-induced oxidative stress. However, the impact of low-penetrance HGSOC risk alleles (e.g. rs3814113) on the damage induced by oxidative stress remains unclear. Therefore, the purpose of this study was to investigate whether rs3814113 genetic interval regulates BNC2 expression and whether BNC2 expression levels impact on cell survival after oxidative stress. To do this, we analyzed gene expression levels of BNC2 first in HGSOC data sets and then in an isogenic cell line that we engineered to carry a 5 kb deletion around rs3814113. Finally, we silenced BNC2 and measured surviving cells after hydrogen peroxide (H2O2) treatment to simulate oxidative stress after ovulation. In this paper, we describe that BNC2 expression levels are reduced in HGSOC samples compared with control samples, and that BNC2 expression levels decrease following oxidative stress and ovulation in vitro and in vivo, respectively. Moreover, deletion of 5 kb surrounding rs3814113 decreases BNC2 expression levels in an isogenic cell line, and silencing of BNC2 expression levels increases cell survival after H2O2 treatment. Altogether, our findings suggest that the intergenic region located around rs3814113 regulates BNC2 expression, which in turn affects cell survival after oxidative stress response. Indeed, HGSOC samples present lower BNC2 expression levels that probably, in the initial phases of oncogenic transformation, conferred resistance to oxidative stress and ultimately reduced the clearance of cells with oxidative-induced damages.
Collapse
Affiliation(s)
- Laura Cesaratto
- Division of Experimental Oncology2, Department of Translational Research, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Eleonora Grisard
- Division of Experimental Oncology2, Department of Translational Research, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Michela Coan
- Division of Experimental Oncology2, Department of Translational Research, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
- Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Luigi Zandonà
- Division of Experimental Oncology2, Department of Translational Research, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Elena De Mattia
- Division of Experimental and Clinical Pharmacology, Department of Translational Research Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Elena Poletto
- Department of Oncology, University Hospital of Udine, Udine, Italy
| | - Erika Cecchin
- Division of Experimental and Clinical Pharmacology, Department of Translational Research Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Fabio Puglisi
- Department of Oncology, University Hospital of Udine, Udine, Italy
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - Vincenzo Canzonieri
- Division of Pathology, Department of Translational Research, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Maria Teresa Mucignat
- Division of Experimental Oncology2, Department of Translational Research, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Antonella Zucchetto
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Gabriele Stocco
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Alfonso Colombatti
- Division of Experimental Oncology2, Department of Translational Research, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Milena S Nicoloso
- Division of Experimental Oncology2, Department of Translational Research, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| | - Riccardo Spizzo
- Division of Experimental Oncology2, Department of Translational Research, Centro di Riferimento Oncologico (CRO Aviano), National Cancer Institute, Aviano, Italy
| |
Collapse
|