1
|
Sun Y, Guo G, Zhang Y, Chen X, Lu Y, Hong R, Xiong J, Li J, Hu X, Wang S, Liu Y, Zhang Z, Yang X, Nan Y, Huang Q. IKBKE promotes the ZEB2-mediated EMT process by phosphorylating HMGA1a in glioblastoma. Cell Signal 2024; 116:111062. [PMID: 38242271 DOI: 10.1016/j.cellsig.2024.111062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
IKBKE (Inhibitor of Nuclear Factor Kappa-B Kinase Subunit Epsilon) is an important oncogenic protein in a variety of tumors, which can promote tumor growth, proliferation, invasion and drug resistance, and plays a critical regulatory role in the occurrence and progression of malignant tumors. HMGA1a (High Mobility Group AT-hook 1a) functions as a cofactor for proper transcriptional regulation and is highly expressed in multiple types of tumors. ZEB2 (Zinc finger E-box Binding homeobox 2) exerts active functions in epithelial mesenchymal transformation (EMT). In our current study, we confirmed that IKBKE can increase the proliferation, invasion and migration of glioblastoma cells. We then found that IKBKE can phosphorylate HMGA1a at Ser 36 and/or Ser 44 sites and inhibit the degradation process of HMGA1a, and regulate the nuclear translocation of HMGA1a. Crucially, we observed that HMGA1a can regulate ZEB2 gene expression by interacting with ZEB2 promoter region. Hence, HMGA1a was found to promote the ZEB2-related metastasis. Consequently, we demonstrated that IKBKE can exert its oncogenic functions via the IKBKE/HMGA1a/ZEB2 signalling axis, and IKBKE may be a prominent biomarker for the treatment of glioblastoma in the future.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Gaochao Guo
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Yu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Xingjie Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Yalin Lu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Rujun Hong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Jinbiao Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Jiabo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Xue Hu
- Department of Clinical Nutrition, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China
| | - Shuaishuai Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Yang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Zhimeng Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Ningbo Hospital of Zhejiang University, Ningbo, Zhejiang 315000, China
| | - Xuejun Yang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Yang Nan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China.
| |
Collapse
|
2
|
Panawan O, Silsirivanit A, Chang C, Putthisen S, Boonnate P, Yokota T, Nishisyama‐Ikeda Y, Detarya M, Sawanyawisuth K, Kaewkong W, Muisuk K, Luang S, Vaeteewoottacharn K, Kariya R, Yano H, Komohara Y, Ohta K, Okada S, Wongkham S, Araki N. Establishment and characterization of a novel cancer stem-like cell of cholangiocarcinoma. Cancer Sci 2023; 114:3230-3246. [PMID: 37026527 PMCID: PMC10394157 DOI: 10.1111/cas.15812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/22/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive malignant tumor of bile duct epithelia. Recent evidence suggests the impact of cancer stem cells (CSC) on the therapeutic resistance of CCA; however, the knowledge of CSC in CCA is limited due to the lack of a CSC model. In this study, we successfully established a stable sphere-forming CCA stem-like cell, KKU-055-CSC, from the original CCA cell line, KKU-055. The KKU-055-CSC exhibits CSC characteristics, including: (1) the ability to grow stably and withstand continuous passage for a long period of culture in the stem cell medium, (2) high expression of stem cell markers, (3) low responsiveness to standard chemotherapy drugs, (4) multilineage differentiation, and (5) faster and constant expansive tumor formation in xenograft mouse models. To identify the CCA-CSC-associated pathway, we have undertaken a global proteomics and functional cluster/network analysis. Proteomics identified the 5925 proteins in total, and the significantly upregulated proteins in CSC compared with FCS-induced differentiated CSC and its parental cells were extracted. Network analysis revealed that high mobility group A1 (HMGA1) and Aurora A signaling through the signal transducer and activator of transcription 3 pathways were enriched in KKU-055-CSC. Knockdown of HMGA1 in KKU-055-CSC suppressed the expression of stem cell markers, induced the differentiation followed by cell proliferation, and enhanced sensitivity to chemotherapy drugs including Aurora A inhibitors. In silico analysis indicated that the expression of HMGA1 was correlated with Aurora A expressions and poor survival of CCA patients. In conclusion, we have established a unique CCA stem-like cell model and identified the HMGA1-Aurora A signaling as an important pathway for CSC-CCA.
Collapse
Affiliation(s)
- Orasa Panawan
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
- Department of Biochemistry, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
| | - Atit Silsirivanit
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
- Department of Biochemistry, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Cholangiocarcinoma Research Institute, Khon Kaen UniversityKhon KaenThailand
| | - Chih‐Hsiang Chang
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Siyaporn Putthisen
- Department of Biochemistry, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
| | - Piyanard Boonnate
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus InfectionKumamoto UniversityKumamotoJapan
| | - Taro Yokota
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Yuki Nishisyama‐Ikeda
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Marutpong Detarya
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
- Department of Biochemistry, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Cholangiocarcinoma Research Institute, Khon Kaen UniversityKhon KaenThailand
| | - Kanlayanee Sawanyawisuth
- Department of Biochemistry, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Cholangiocarcinoma Research Institute, Khon Kaen UniversityKhon KaenThailand
| | - Worasak Kaewkong
- Department of Biochemistry, Faculty of Medical SciencesNaresuan UniversityPhitsanulokThailand
| | - Kanha Muisuk
- Department of Forensic Medicine, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
| | - Sukanya Luang
- Department of Biochemistry, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Cholangiocarcinoma Research Institute, Khon Kaen UniversityKhon KaenThailand
| | - Kulthida Vaeteewoottacharn
- Department of Biochemistry, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Cholangiocarcinoma Research Institute, Khon Kaen UniversityKhon KaenThailand
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus InfectionKumamoto UniversityKumamotoJapan
| | - Ryusho Kariya
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus InfectionKumamoto UniversityKumamotoJapan
| | - Hiromu Yano
- Department of Cell Pathology, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Kunimasa Ohta
- Department of Stem Cell Biology, Faculty of Arts and ScienceKyushu UniversityFukuokaJapan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus InfectionKumamoto UniversityKumamotoJapan
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Center for Translational Medicine, Faculty of MedicineKhon Kaen UniversityKhon KaenThailand
| | - Norie Araki
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
3
|
Wang L, Zhang J, Xia M, Liu C, Zu X, Zhong J. High Mobility Group A1 (HMGA1): Structure, Biological Function, and Therapeutic Potential. Int J Biol Sci 2022; 18:4414-4431. [PMID: 35864955 PMCID: PMC9295051 DOI: 10.7150/ijbs.72952] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022] Open
Abstract
High mobility group A1 (HMGA1) is a nonhistone chromatin structural protein characterized by no transcriptional activity. It mainly plays a regulatory role by modifying the structure of DNA. A large number of studies have confirmed that HMGA1 regulates genes related to tumours in the reproductive system, digestive system, urinary system and haematopoietic system. HMGA1 is rare in adult cells and increases in highly proliferative cells such as embryos. After being stimulated by external factors, it will produce effects through the Wnt/β-catenin, PI3K/Akt, Hippo and MEK/ERK pathways. In addition, HMGA1 also affects the ageing, apoptosis, autophagy and chemotherapy resistance of cancer cells, which are linked to tumorigenesis. In this review, we summarize the mechanisms of HMGA1 in cancer progression and discuss the potential clinical application of targeted HMGA1 therapy, indicating that targeted HMGA1 is of great significance in the diagnosis and treatment of malignancy.
Collapse
Affiliation(s)
- Lu Wang
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Ji Zhang
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Min Xia
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Chang Liu
- Department of Endocrinology and Metabolism, The First People's Hospital of Chenzhou, First School of Clinical Medicine, University of Southern Medical, Guangzhou 510515, Guangdong, China
| | - Xuyu Zu
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jing Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| |
Collapse
|
4
|
Ghosh S, Guimaraes JC, Lanzafame M, Schmidt A, Syed AP, Dimitriades B, Börsch A, Ghosh S, Mittal N, Montavon T, Correia AL, Danner J, Meister G, Terracciano LM, Pfeffer S, Piscuoglio S, Zavolan M. Prevention of dsRNA-induced interferon signaling by AGO1x is linked to breast cancer cell proliferation. EMBO J 2020; 39:e103922. [PMID: 32812257 PMCID: PMC7507497 DOI: 10.15252/embj.2019103922] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 06/27/2020] [Accepted: 07/07/2020] [Indexed: 01/05/2023] Open
Abstract
Translational readthrough, i.e., elongation of polypeptide chains beyond the stop codon, was initially reported for viral RNA, but later found also on eukaryotic transcripts, resulting in proteome diversification and protein‐level modulation. Here, we report that AGO1x, an evolutionarily conserved translational readthrough isoform of Argonaute 1, is generated in highly proliferative breast cancer cells, where it curbs accumulation of double‐stranded RNAs (dsRNAs) and consequent induction of interferon responses and apoptosis. In contrast to other mammalian Argonaute protein family members with primarily cytoplasmic functions, AGO1x exhibits nuclear localization in the vicinity of nucleoli. We identify AGO1x interaction with the polyribonucleotide nucleotidyltransferase 1 (PNPT1) and show that the depletion of this protein further augments dsRNA accumulation. Our study thus uncovers a novel function of an Argonaute protein in buffering the endogenous dsRNA‐induced interferon responses, different than the canonical function of AGO proteins in the miRNA effector pathway. As AGO1x expression is tightly linked to breast cancer cell proliferation, our study thus suggests a new direction for limiting tumor growth.
Collapse
Affiliation(s)
- Souvik Ghosh
- Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Joao C Guimaraes
- Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Manuela Lanzafame
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Afzal Pasha Syed
- Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Beatrice Dimitriades
- Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Anastasiya Börsch
- Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Shreemoyee Ghosh
- Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Nitish Mittal
- Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Thomas Montavon
- Architecture et Réactivité de l'ARN, Institut de biologie moléculaire et cellulaire du CNRS, Université de Strasbourg, Strasbourg, France
| | - Ana Luisa Correia
- Department of Biomedicine, University of Basel/University Hospital Basel, Basel, Switzerland
| | - Johannes Danner
- Department of Biochemistry, Department of Biology and Preclinical Medicine, University of Regensburg, Regensburg, Germany
| | - Gunter Meister
- Department of Biochemistry, Department of Biology and Preclinical Medicine, University of Regensburg, Regensburg, Germany
| | | | - Sébastien Pfeffer
- Architecture et Réactivité de l'ARN, Institut de biologie moléculaire et cellulaire du CNRS, Université de Strasbourg, Strasbourg, France
| | - Salvatore Piscuoglio
- Institute of Pathology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel/University Hospital Basel, Basel, Switzerland
| | - Mihaela Zavolan
- Computational and Systems Biology, Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
5
|
Clinical Implications of Extracellular HMGA1 in Breast Cancer. Int J Mol Sci 2019; 20:ijms20235950. [PMID: 31779212 PMCID: PMC6928815 DOI: 10.3390/ijms20235950] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 02/06/2023] Open
Abstract
The unconventional secretion of proteins is generally caused by cellular stress. During the tumorigenesis, tumor cells experience high levels of stress, and the secretion of some theoretically intracellular proteins is activated. Once in the extracellular space, these proteins play different paracrine and autocrine roles and could represent a vulnerability of cancer. One of these proteins is the high mobility group A1 (HMGA1), which is frequently overexpressed in tumors and presents a low expression in normal adult tissues. We have recently described that HMGA1 establishes an autocrine loop in invasive triple-negative breast cancer (TNBC) cells. The secretion of HMGA1 and its binding to the receptor for advanced glycation end products (RAGE) mediates the migration, invasion, and metastasis of TNBC cells and predicts the onset of metastasis in these patients. In this review, we summarized different strategies to exploit the novel tumorigenic phenotype mediated by extracellular HMGA1. We envisioned future clinical applications where the association between its change in subcellular localization and breast cancer progression could be used to predict tumor aggressiveness and guide treatment decisions. Furthermore, we proposed that targeting extracellular HMGA1 as monotherapy using monoclonal antibodies, or in combination with chemotherapy and other targeted therapies, could bring new therapeutic options for TNBC patients.
Collapse
|
6
|
Zeindler J, Angehrn F, Droeser R, Däster S, Piscuoglio S, Ng CKY, Kilic E, Mechera R, Meili S, Isaak A, Weber WP, Muenst S, Soysal SD. Infiltration by myeloperoxidase-positive neutrophils is an independent prognostic factor in breast cancer. Breast Cancer Res Treat 2019; 177:581-589. [PMID: 31267330 DOI: 10.1007/s10549-019-05336-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/22/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Myeloperoxidase (MPO) is an enzyme secreted by neutrophil granulocytes as a result of phagocytosis during inflammation. In colorectal cancer, tumour infiltration by MPO expressing cells has been shown to be independently associated with a favourable prognosis. In this study, we explored the role of MPO-positive cell infiltration and its prognostic significance in invasive breast cancer. METHODS We performed immunohistochemical staining for MPO on multiple tissue microarrays comprising a total of 928 human breast cancer samples with detailed clinical-pathological annotation and outcome data. RESULTS MPO-positive cell infiltration (≥ 5 cells/tissue punch) was found in 150 (16%) of the 928 evaluable breast cancer cases. In univariate survival analyses, infiltration by MPO-positive cells was associated with a significantly better overall survival (p < 0.001). In subset univariate analyses, the infiltration by MPO-positive cells was associated with significantly better overall survival in the Luminal B/HER2-negative subtype (p = 0.005), the HER2 enriched subtype (p = 0.011), and the Triple Negative subtype (p < 0.001). In multivariate analysis, MPO expression proved to be an independent prognostic factor for improved overall survival (p < 0.001). CONCLUSIONS This is the first study to show that infiltration of MPO-positive cells is an independent prognostic biomarker for improved overall survival in human breast cancer.
Collapse
Affiliation(s)
- Jasmin Zeindler
- Breast Cancer Center, University Hospital Basel and University of Basel, 4031, Basel, Switzerland
| | - Fiorenzo Angehrn
- Department of Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Raoul Droeser
- Department of Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Silvio Däster
- Department of Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Salvatore Piscuoglio
- Visceral Surgery Research Laboratory, Clarunis, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Charlotte K Y Ng
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Ergin Kilic
- Institute of Pathology, Hospital Leverkusen, Leverkusen, Germany
| | - Robert Mechera
- Department of Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Samuel Meili
- Department of Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland
| | - Andrej Isaak
- Department of Vascular and Endovascular Surgery, University Hospital Basel, Basel, Switzerland
| | - Walter P Weber
- Breast Cancer Center, University Hospital Basel and University of Basel, 4031, Basel, Switzerland
| | - Simone Muenst
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Savas Deniz Soysal
- Department of Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases Basel, Basel, Switzerland. .,Visceral Surgery Research Laboratory, Clarunis, Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
7
|
Qin MM, Chai X, Huang HB, Feng G, Li XN, Zhang J, Zheng R, Liu XC, Pu C. let-7i inhibits proliferation and migration of bladder cancer cells by targeting HMGA1. BMC Urol 2019; 19:53. [PMID: 31196036 PMCID: PMC6567622 DOI: 10.1186/s12894-019-0485-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 06/03/2019] [Indexed: 12/20/2022] Open
Abstract
Background Let-7 is one of the earliest discovered microRNAs(miRNAs) and has been reported to be down-regulated in multiple malignant tumors. The effects and molecular mechanisms of let-7i in bladder cancer are still unclear. This study was to investigate the effects and potential mechanisms of let-7i on bladder cancer cells. Methods Total RNA was extracted from bladder cancer cell lines. The expression levels of let-7i and HMGA1 were examined by quantitative real-time PCR. Cell viability was detected using the CCK-8 and colony formation assays, while transwell and wound healing assays were used to evaluate migration ability. Luciferase reporter assay and western blot were used to confirm the target gene of let-7i. Results Compared with the SV-40 immortalized human uroepithelial cell line (SV-HUC-1), bladder cancer cell lines T24 and 5637 had low levels of let-7i expression, but high levels of high mobility group protein A1 (HMGA1) expression. Transfection of cell lines T24 and 5637 with let-7i mimic suppressed cell proliferation and migration. Luciferase reporter assay confirmed HMGA1 may be one of the target genes of let-7i-5p. Protein and mRNA expression of HMGA1 was significantly downregulated in let-7i mimic transfected cell lines T24 and 5637. Conclusions Up-regulation of let-7i suppressed proliferation and migration of the human bladder cancer cell lines T24 and 5637 by targeting HMGA1. These findings suggest that let-7i might be considered as a novel therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- M-M Qin
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No.2, West Zheshan Road, Wuhu, 241001, Anhui, China
| | - X Chai
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - H-B Huang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - G Feng
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No.2, West Zheshan Road, Wuhu, 241001, Anhui, China
| | - X-N Li
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No.2, West Zheshan Road, Wuhu, 241001, Anhui, China
| | - J Zhang
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No.2, West Zheshan Road, Wuhu, 241001, Anhui, China
| | - R Zheng
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No.2, West Zheshan Road, Wuhu, 241001, Anhui, China
| | - X-C Liu
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No.2, West Zheshan Road, Wuhu, 241001, Anhui, China
| | - C Pu
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No.2, West Zheshan Road, Wuhu, 241001, Anhui, China.
| |
Collapse
|
8
|
Mechera R, Soysal SD, Piscuoglio S, Ng CKY, Zeindler J, Mujagic E, Däster S, Glauser P, Hoffmann H, Kilic E, Droeser RA, Weber WP, Muenst S. Expression of RET is associated with Oestrogen receptor expression but lacks prognostic significance in breast cancer. BMC Cancer 2019; 19:41. [PMID: 30621641 PMCID: PMC6325785 DOI: 10.1186/s12885-018-5262-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 12/28/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The Rearranged during Transfection (RET) protein is overexpressed in a subset of Estrogen Receptor (ER) positive breast cancer, with both signalling pathways functionally interacting. This cross-talk plays a pivotal role in the resistance of breast cancer cells to anti-endocrine therapies, and RET expression is assumed to correlate with poor prognosis based on findings in small patient cohorts. The aim of our study was to investigate the impact of RET expression on patient outcome in human breast cancer. METHODS We performed an immunohistochemical analysis of RET protein expression on a tissue microarray encompassing 990 breast cancer patients and correlated its expression with clinicopathological parameters and survival data. RESULTS Expression of RET was detected in 409 out of 990 cases (41.3%). RET and ER expression significantly correlated (p < 0.0001). The Luminal B HER2-positive subtype showed the highest expression rate (48.9%). In univariate and multivariate survival analyses, RET expression had no impact on overall survival. CONCLUSION We confirmed the co-expression of RET and ER, but we did not find RET expression to be an independent prognostic factor in human breast cancer. Clinical trials with newly developed RET inhibitors are needed to evaluate if RET inhibition has a beneficial impact on patient survival in ER positive breast cancer.
Collapse
Affiliation(s)
- Robert Mechera
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Savas D Soysal
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland.
| | - Salvatore Piscuoglio
- Institute of Pathology, University Hospital Basel, Schoenbeinstrasse 40, 4031, Basel, Switzerland
| | - Charlotte K Y Ng
- Institute of Pathology, University Hospital Basel, Schoenbeinstrasse 40, 4031, Basel, Switzerland
| | - Jasmin Zeindler
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Edin Mujagic
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Silvio Däster
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Philippe Glauser
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Henry Hoffmann
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Ergin Kilic
- Institute of Pathology, Klinikum Leverkusen, Am Gesundheitspark 11, 51375, Leverkusen, Germany
| | - Raoul A Droeser
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Walter P Weber
- Department of Surgery, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Simone Muenst
- Institute of Pathology, University Hospital Basel, Schoenbeinstrasse 40, 4031, Basel, Switzerland
| |
Collapse
|
9
|
Ohe K, Miyajima S, Abe I, Tanaka T, Hamaguchi Y, Harada Y, Horita Y, Beppu Y, Ito F, Yamasaki T, Terai H, Mori M, Murata Y, Tanabe M, Ashida K, Kobayashi K, Enjoji M, Yanase T, Harada N, Utsumi T, Mayeda A. HMGA1a induces alternative splicing of estrogen receptor alpha in MCF-7 human breast cancer cells. J Steroid Biochem Mol Biol 2018; 182:21-26. [PMID: 29678492 DOI: 10.1016/j.jsbmb.2018.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/23/2017] [Accepted: 04/13/2018] [Indexed: 12/11/2022]
Abstract
The high-mobility group A protein 1a (HMGA1a) protein is known as an oncogene whose expression level in cancer tissue correlates with the malignant potential, and known as a component of senescence-related structures connecting it to tumor suppressor networks in fibroblasts. HMGA1 protein binds to DNA, but recent studies have shown it exerts novel functions through RNA-binding. Our previous studies have shown that sequence-specific RNA-binding of HMGA1a induces exon-skipping of Presenilin-2 exon 5 in sporadic Alzheimer disease. Here we show that HMGA1a induced exon-skipping of the estrogen receptor alpha (ERα) gene and increased ERα46 mRNA expression in MCF-7 breast cancer cells. An RNA-decoy of HMGA1a efficiently blocked this event and reduced ERα46 protein expression. Blockage of HMGA1a RNA-binding property consequently induced cell growth through reduced ERα46 expression in MCF-7 cells and increased sensitivity to tamoxifen in the tamoxifen-resistant cell line, MCF-7/TAMR1. Stable expression of an HMGA1a RNA-decoy in MCF-7 cells exhibited decreased ERα46 protein expression and increased estrogen-dependent tumor growth when these cells were implanted in nude mice. These results show HMGA1a is involved in alternative splicing of the ERα gene and related to estrogen-related growth as well as tamoxifen sensitivity in MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Kenji Ohe
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan.
| | - Shinsuke Miyajima
- Department of Breast Surgery, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Ichiro Abe
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University Chikushi Hospital, Chikushino city, 818-8502, Japan
| | - Tomoko Tanaka
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Yuriko Hamaguchi
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Yoshihiro Harada
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Yuta Horita
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Yuki Beppu
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Fumiaki Ito
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Takafumi Yamasaki
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Hiroki Terai
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Masayoshi Mori
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Yusuke Murata
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Makito Tanabe
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Kenji Ashida
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kunihisa Kobayashi
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University Chikushi Hospital, Chikushino city, 818-8502, Japan
| | - Munechika Enjoji
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Toshihiko Yanase
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-180, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Toshiaki Utsumi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Akila Mayeda
- Division of Gene Expression Mechanism, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Aichi, Toyoake, 470-1192, Japan
| |
Collapse
|
10
|
High Mobility Group A (HMGA) proteins: Molecular instigators of breast cancer onset and progression. Biochim Biophys Acta Rev Cancer 2018. [DOI: 10.1016/j.bbcan.2018.03.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
11
|
Qi C, Cao J, Li M, Liang C, He Y, Li Y, Li J, Zheng X, Wang L, Wei B. HMGA1 Overexpression is Associated With the Malignant Status and Progression of Breast Cancer. Anat Rec (Hoboken) 2018; 301:1061-1067. [PMID: 29316384 DOI: 10.1002/ar.23777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/20/2017] [Accepted: 11/23/2017] [Indexed: 12/30/2022]
Abstract
Breast cancer is the most common malignant tumor among women, and the incidence and mortality of breast cancer has rapidly increased in recent years. Studies have indicated that high mobility group A1 (HMGA1), an important member of the HMGA family, plays a role in the pathogenesis and progression of malignant tumors, including breast cancer. This study aims to evaluate the effect of HMGA1 in breast cancer. Interestingly, we found that HMGA1 expression was significantly higher in breast cancer tissues than in adenoma tissues and closely correlated with the clinical stage and histological grade in breast cancer patients. Further study showed that HMGA1 knockdown inhibited the proliferation and migration of breast cancer cells. Thus, the results demonstrated that HMGA1 could act as an independent prognostic indicator in breast cancer. HMGA1 expression was closely correlated with the clinical stage, histological grade, and tumor size in breast cancer patients and breast cancer progression in transgenic MMTV-PyMT mice. Anat Rec, 301:1061-1067, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cuiling Qi
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jinghua Cao
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Mengshi Li
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chenghua Liang
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Yajun He
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuanyuan Li
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jialin Li
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaoming Zheng
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Lijing Wang
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bo Wei
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
12
|
De Martino M, Forzati F, Arra C, Fusco A, Esposito F. HMGA1-pseudogenes and cancer. Oncotarget 2017; 7:28724-35. [PMID: 26895108 PMCID: PMC5053758 DOI: 10.18632/oncotarget.7427] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/05/2016] [Indexed: 12/25/2022] Open
Abstract
Pseudogenes are DNA sequences with high homology to the corresponding functional gene, but, because of the accumulation of various mutations, they have lost their initial functions to code for proteins. Consequently, pseudogenes have been considered until few years ago dysfunctional relatives of the corresponding ancestral genes, and then useless in the course of genome evolution. However, several studies have recently established that pseudogenes are owners of key biological functions. Indeed, some pseudogenes control the expression of functional genes by competitively binding to the miRNAs, some of them generate small interference RNAs to negatively modulate the expression of functional genes, and some of them even encode functional mutated proteins. Here, we concentrate our attention on the pseudogenes of the HMGA1 gene, that codes for the HMGA1a and HMGA1b proteins having a critical role in development and cancer progression. In this review, we analyze the family of HMGA1 pseudogenes through three aspects: classification, characterization, and their possible function and involvement in cancer.
Collapse
Affiliation(s)
- Marco De Martino
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Floriana Forzati
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Claudio Arra
- Istituto Nazionale dei Tumori, Fondazione Pascale, Naples, Italy
| | - Alfredo Fusco
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Francesco Esposito
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Scuola di Medicina e Chirurgia di Napoli, Università degli Studi di Napoli "Federico II", Naples, Italy
| |
Collapse
|
13
|
Quintavalle C, Burmeister K, Piscuoglio S, Quagliata L, Karamitopoulou E, Sepe R, Fusco A, Terracciano LM, Andersen JB, Pallante P, Matter MS. High mobility group A1 enhances tumorigenicity of human cholangiocarcinoma and confers resistance to therapy. Mol Carcinog 2017; 56:2146-2157. [PMID: 28467612 DOI: 10.1002/mc.22671] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/01/2017] [Indexed: 12/16/2022]
Abstract
High mobility group A1 (HMGA1) protein has been described to play an important role in numerous types of human carcinoma. By the modulation of several target genes HMGA1 promotes proliferation and epithelial-mesenchymal transition of tumor cells. However, its role in cholangiocarcinoma (CCA) has not been addressed yet. Therefore, we determined HMGA1 mRNA expression in CCA samples in a transcriptome array (n = 104) and a smaller cohort (n = 13) by qRT-PCR. Protein expression was evaluated by immunohistochemistry in a tissue microarray (n = 67). In addition, we analyzed changes in cell proliferation, colony formation, response to gemcitabine treatment, and target gene expression after modulation of HMGA1 expression in CCA cell lines. mRNA levels of HMGA1 were found to be upregulated in 15-62% depending on the cohort analyzed. Immunohistochemistry showed HMGA1 overexpression in 51% of CCA specimens. Integration with clinico-pathological data revealed that high HMGA1 expression was associated with reduced time to recurrence and a positive lymph node status in extrahepatic cholangiocellular carcinoma. In vitro experiments showed that overexpression of HMGA1 in CCA cell lines promoted cell proliferation, whereas its suppression reduced growth rate. HMGA1 further promoted colony formation in an anchorage independent growth and conferred resistance to gemcitabine treatment. Finally, HMGA1 modulated the expression of two genes involved in CCA carcinogenesis, iNOS and ERBB2. In conclusion, our findings indicate that HMGA1 expression is increased in a substantial number of CCA specimens. HMGA1 further promotes CCA tumorigenicity and confers resistance to chemotherapy.
Collapse
Affiliation(s)
- Cristina Quintavalle
- Division of Molecular Pathology, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Katharina Burmeister
- Division of Molecular Pathology, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Salvatore Piscuoglio
- Division of Molecular Pathology, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Luca Quagliata
- Division of Molecular Pathology, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Eva Karamitopoulou
- Translational Research Unit, Institute of Pathology, University of Bern, Bern, Switzerland.,Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Romina Sepe
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale (IEOS) "G. Salvatore", Consiglio Nazionale delle Ricerche (CNR), Napoli, Italy
| | - Alfredo Fusco
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale (IEOS) "G. Salvatore", Consiglio Nazionale delle Ricerche (CNR), Napoli, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Luigi M Terracciano
- Division of Molecular Pathology, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Jesper B Andersen
- Department of Health and Medical Sciences, Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Pierlorenzo Pallante
- Division of Molecular Pathology, Institute of Pathology, University of Basel, Basel, Switzerland.,Istituto per l'Endocrinologia e l'Oncologia Sperimentale (IEOS) "G. Salvatore", Consiglio Nazionale delle Ricerche (CNR), Napoli, Italy
| | - Matthias S Matter
- Division of Molecular Pathology, Institute of Pathology, University of Basel, Basel, Switzerland
| |
Collapse
|
14
|
Andreozzi M, Quintavalle C, Benz D, Quagliata L, Matter M, Calabrese D, Tosti N, Ruiz C, Trapani F, Tornillo L, Fusco A, Heim MH, Ng CK, Pallante P, Terracciano LM, Piscuoglio S. HMGA1 Expression in Human Hepatocellular Carcinoma Correlates with Poor Prognosis and Promotes Tumor Growth and Migration in in vitro Models. Neoplasia 2016; 18:724-731. [PMID: 27855356 PMCID: PMC5110473 DOI: 10.1016/j.neo.2016.10.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/17/2016] [Accepted: 10/17/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND HMGA1 is a non-histone nuclear protein that regulates cellular proliferation, invasion and apoptosis and is overexpressed in many carcinomas. In this study we sought to explore the expression of HMGA1 in HCCs and cirrhotic tissues, and its effect in in vitro models. METHODS We evaluated HMGA1 expression using gene expression microarrays (59 HCCs, of which 37 were matched with their corresponding cirrhotic tissue and 5 normal liver donors) and tissue microarray (192 HCCs, 108 cirrhotic tissues and 79 normal liver samples). HMGA1 expression was correlated with clinicopathologic features and patient outcome. Four liver cancer cell lines with stable induced or knockdown expression of HMGA1 were characterized using in vitro assays, including proliferation, migration and anchorage-independent growth. RESULTS HMGA1 expression increased monotonically from normal liver tissues to cirrhotic tissue to HCC (P<.01) and was associated with Edmondson grade (P<.01). Overall, 51% and 42% of HCCs and cirrhotic tissues expressed HMGA1, respectively. Patients with HMGA1-positive HCCs had earlier disease progression and worse overall survival. Forced expression of HMGA1 in liver cancer models resulted in increased cell growth and migration, and vice versa. Soft agar assay showed that forced expression of HMGA1 led to increased foci formation, suggesting an oncogenic role of HMGA1 in hepatocarcinogenesis. CONCLUSIONS HMGA1 is frequently expressed in cirrhotic tissues and HCCs and its expression is associated with high Edmondson grade and worse prognosis in HCC. Our results suggest that HMGA1 may act as oncogenic driver of progression, implicating it in tumor growth and migration potential in liver carcinogenesis.
Collapse
Affiliation(s)
| | | | - David Benz
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Luca Quagliata
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Matthias Matter
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Diego Calabrese
- Department of Biomedicine, Hepatology Laboratory, University of Basel, Basel, Switzerland
| | - Nadia Tosti
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Christian Ruiz
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Francesca Trapani
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Luigi Tornillo
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Alfredo Fusco
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), and Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples "Federico II, Naples, Italy; National Cancer Institute-INCA, Rua Andrè Cavalcanti, 37-Centro, Rio de Janeiro, Brazil
| | - Markus H Heim
- Department of Biomedicine, Hepatology Laboratory, University of Basel, Basel, Switzerland
| | - Charlotte Ky Ng
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Pierlorenzo Pallante
- Institute of Pathology, University Hospital Basel, Basel, Switzerland; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), and Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples "Federico II, Naples, Italy
| | | | | |
Collapse
|