1
|
Yang K, Chai S, Song H, Cao S, Gao F, Zhou C, Li L. Downregulation of ECRG4 by DNMT1 promotes EC growth via IRF3/IFN-γ/miR-29b/DNMT1/ECRG4 positive feedback loop. iScience 2025; 28:111614. [PMID: 39834855 PMCID: PMC11742825 DOI: 10.1016/j.isci.2024.111614] [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: 01/04/2024] [Revised: 10/10/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Esophageal carcinoma (EC) is one of the most common malignant tumors in the world. ECRG4 has been recently discovered to be downregulated in EC. However, the mechanism leading to reduced expression of ECRG4 in esophageal cancer remains obscure. Here, we found that ECRG4 expression was significantly downregulated in EC tissues and cell lines. ECRG4 overexpression led to a significant decrease in proliferation in vitro and in vivo. Mechanistically, ECRG4 can activate IRF3/IFN-γ pathway. IFN-γ can promote the expression of miR-29b. MiR-29b reduces the expression of DNMT1. DNMT1 may affect the expression of ECRG4 by affecting the methylation of ECRG4 promoter. These results reveal ECRG4/IRF3/IFN-γ/miR-29b/DNMT1 positive feedback loop in esophageal carcinoma cells, which may become a potential therapeutic target for esophageal carcinoma.
Collapse
Affiliation(s)
- Ke Yang
- Department of Oncology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, 450003 Henan, China
| | - Shuaining Chai
- Department of Oncology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, 450003 Henan, China
| | - Helong Song
- Department of Oncology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, 450003 Henan, China
| | - Sinan Cao
- Department of Oncology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, 450003 Henan, China
| | - Fangmiao Gao
- Department of Oncology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, 450003 Henan, China
| | - Chenxuan Zhou
- Department of Oncology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, 450003 Henan, China
| | - Linwei Li
- Department of Oncology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, 450003 Henan, China
- Department of Oncology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, 450003 Henan, China
| |
Collapse
|
2
|
Rao R, Gulfishan M, Kim MS, Kashyap MK. Deciphering Cancer Complexity: Integrative Proteogenomics and Proteomics Approaches for Biomarker Discovery. Methods Mol Biol 2025; 2859:211-237. [PMID: 39436604 DOI: 10.1007/978-1-0716-4152-1_12] [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] [Indexed: 10/23/2024]
Abstract
Proteomics has revolutionized the field of cancer biology because the use of a large number of in vivo (SILAC), in vitro (iTRAQ, ICAT, TMT, stable-isotope Dimethyl, and 18O) labeling techniques or label-free methods (spectral counting or peak intensities) coupled with mass spectrometry enables us to profile and identify dysregulated proteins in diseases such as cancer. These proteome and genome studies have led to many challenges, such as the lack of consistency or correlation between copy numbers, RNA, and protein-level data. This review covers solely mass spectrometry-based approaches used for cancer biomarker discovery. It also touches on the emerging role of oncoproteogenomics or proteogenomics in cancer biomarker discovery and how this new area is attracting the integration of genomics and proteomics areas to address some of the important questions to help impinge on the biology and pathophysiology of different malignancies to make these mass spectrometry-based studies more realistic and relevant to clinical settings.
Collapse
Affiliation(s)
- Rashmi Rao
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, UP, India
| | - Mohd Gulfishan
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, UP, India
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu-42988, Republic of Korea
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute (ASCI), Amity Medical School (AMS), Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
3
|
Xu Q, Zhang X, Hao M, Dang X, Xu Q, Cyganek L, Akin I, Tang D, Liao B, Zhou X, Lan H. Esophageal Cancer-Related Gene-4 Contributes to Lipopolysaccharide-Induced Ion Channel Dysfunction in hiPSC-Derived Cardiomyocytes. J Inflamm Res 2024; 17:10183-10197. [PMID: 39649417 PMCID: PMC11624686 DOI: 10.2147/jir.s470828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/22/2024] [Indexed: 12/10/2024] Open
Abstract
Background and Purpose Esophageal cancer-related gene-4 (ECRG4) participate in inflammation process and can interact with the innate immunity complex TLR4-MD2-CD14 on human granulocytes. In addition, ECRG4 participate in modulation of ion channel function and electrical activity of cardiomyocytes. However, the exact mechanism is unknown. This study aimed to test our hypothesis that ECRG4 contributes to inflammation-induced ion channel dysfunctions in cardiomyocytes. Methods Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) generated from three donors were treated with lipopolysaccharide (LPS) to establish an endotoxin-induced inflammatory model. Immunostaining, real-time PCR, and patch-clamp techniques were used for the study. Results ECRG4 was detected in hiPSC-CMs at different differentiation time. LPS treatment increased ECRG4 expression in hiPSC-CMs. Knockdown of ECRG4 decreased the expression level of Toll-Like-Receptor 4 (TLR4, a LPS receptor) and its associated genes and inflammatory cytokines. Furthermore, ECRG4 knockdown shortened the action potential duration (APD) and intercepted LPS-induced APD prolongation by enhancing ISK (small conductance calcium-activated K channel current) and attenuating INCX (Na/Ca exchanger current). Overexpression of ECRG4 mimicked LPS effects on ISK and INCX, which could be prevented by NFκB signaling blockers. Conclusion This study demonstrated that LPS effects on cardiac ion channel function were mediated by the upregulation of ECRG4, which affects NFκB signaling. Our findings support the roles of ECRG4 in inflammatory responses and the ion channel dysfunctions induced by LPS challenge.
Collapse
Affiliation(s)
- Qiang Xu
- School of Basic Medical Science, Southwest Medical University, Luzhou, People’s Republic of China
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Xiangjie Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - Maolin Hao
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - Xitong Dang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - QianQian Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Göttingen, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg, Mannheim, Germany
| | - Dan Tang
- The First People’s Hospital of Longquanyi District, Chengdu/West China Longquan Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Bin Liao
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Xiaobo Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg, Mannheim, Germany
| | - Huan Lan
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| |
Collapse
|
4
|
Wang C, He J, Chen C, Luo W, Dang X, Mao L. A potential role of human esophageal cancer-related gene-4 in cardiovascular homeostasis. Gene 2024; 894:147977. [PMID: 37956966 DOI: 10.1016/j.gene.2023.147977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/10/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Human esophageal cancer related gene-4 (ECRG-4) encodes a 148-aminoacid pre-pro-peptide that can be processed tissue-dependently into multiple small peptides possessing multiple functions distinct from, similar to, or opposite to the tumor suppressor function of the full-length Ecrg4. Ecrg-4 is covalently bound to the cell surface through its signal peptide, colocalized with the innate immunity complex (TLR4-CD14-MD2), and functions as a 'sentinel' molecule in the maintenance of epithelium and leukocyte homeostasis, meaning that the presence of Ecrg-4 on the cell surface signals the maintained homeostasis, whereas the loss of Ecrg-4 due to tissue injury activates pro-inflammatory and tissue proliferative responses, and the level of Ecrg-4 gradually returns to its pre-injury level upon wound healing. Interestingly, Ecrg-4 is also highly expressed in the heart and its conduction system, endothelial cells, and vascular smooth muscle cells. Accumulating evidence has shown that Ecrg-4 is involved in cardiac rate/rhythm control, the development of atrial fibrillation, doxorubicin-induced cardiotoxicity, the ischemic response of the heart and hypoxic response in the carotid body, the pathogenesis of atherosclerosis, and likely the endemic incidence of idiopathic dilated cardiomyopathy. These preliminary discoveries suggest that Ecrg-4 may function as a 'sentinel' molecule in cardiovascular system as well. Here, we briefly review the basic characteristics of ECRG-4 as a tumor suppressor gene and its regulatory functions on inflammation and apoptosis; summarize the discoveries about its distribution in cardiovascular system and involvement in the development of CVDs, and discuss its potential as a novel therapeutic target for the maintenance of cardiovascular system homeostasis.
Collapse
Affiliation(s)
- Chaoying Wang
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China
| | - Jianghui He
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China
| | - Chunyue Chen
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China
| | - Wenjun Luo
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China
| | - Xitong Dang
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China.
| | - Liang Mao
- The Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, China; Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
5
|
Richter M, Lalli E, Ruggiero C. Complex and pleiotropic signaling pathways regulated by the secreted protein augurin. Cell Commun Signal 2023; 21:69. [PMID: 37041625 PMCID: PMC10088197 DOI: 10.1186/s12964-023-01090-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/26/2023] [Indexed: 04/13/2023] Open
Abstract
The secreted protein augurin, the product of the tumor suppressor gene Ecrg4, has been identified as a peptide hormone in the human proteome in 2007. Since then, a number of studies have been carried out to highlight its structure and processing and its potential roles in physiopathology. Although augurin has been shown to be implicated in a variety of processes, ranging from tumorigenesis, inflammation and infection to neural stem cell proliferation, hypothalamo-pituitary adrenal axis regulation and osteoblast differentiation, the molecular mechanisms of its biological effects and the signaling pathways it regulates are still poorly characterized. Here we provide a comprehensive overview of augurin-dependent signal transduction pathways. Because of their secreted nature and the potential to be manipulated pharmacologically, augurin and its derived peptides represent attractive targets for diagnostic development and discovery of new therapeutic agents for the human diseases resulting from the deregulation of the signaling cascades they modulate. From this perspective, the characterization of the precise nature of augurin derived peptides and the identification of the receptor(s) on the cell surface conveying augurin signaling to downstream effectors are crucial to develop agonists and antagonists for this protein. Video abstract.
Collapse
Affiliation(s)
- Margaux Richter
- Institut de Pharmacologie Moleculaire et Cellulaire CNRS UMR 7275, Valbonne, France
- Universite Cote d'Azur, Valbonne, France
| | - Enzo Lalli
- Institut de Pharmacologie Moleculaire et Cellulaire CNRS UMR 7275, Valbonne, France
- Universite Cote d'Azur, Valbonne, France
- Inserm, Valbonne, France
| | - Carmen Ruggiero
- Institut de Pharmacologie Moleculaire et Cellulaire CNRS UMR 7275, Valbonne, France.
- Universite Cote d'Azur, Valbonne, France.
| |
Collapse
|
6
|
Zhang Z, Wang W, Zhang Y, You X, Wu J. A potential link between aberrant expression of ECRG4 and atrial fibrillation. Front Oncol 2023; 13:1031128. [PMID: 36910669 PMCID: PMC9992723 DOI: 10.3389/fonc.2023.1031128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Esophageal cancer-related gene-4 (ECRG4), a 148-amino acid propertied and new tumor suppressor, is initially cloned from the normal esophageal epithelium. ECRG4 was found to be expressed not only in esophageal tissues but also in cardiomyocytes. Previous studies demonstrated that ECRG4 is constitutively expressed in esophageal epithelial cells, and its degree of downregulation is directly proportional to prognosis in patients with esophageal cancer. In the heart, ECRG4 shows greater expression in the atria than in the ventricles, which accounts for its heterogeneity. Downregulation of ECRG4 expression level correlates with esophageal cancer, as well as myocardial injuries and arrhythmias. As a result, this review summarizes the possible susceptibility gene, ECRG4 and its associated molecular mechanisms in cancer patients with atrial fibrillation and myocardial injury. The review begins by describing ECRG4's biological background, discusses its expression in the cardiovascular system, lists the clinical and animal research related to the downregulation of ECRG4 in atrial fibrillation, and focuses on its potential role in atrial fibrillation. Downregulation of ECRG4 may increase the risk of atrial fibrillation by affecting ion channels, MMPs expression and inflammatory response. We will then discuss how ECRG4 can be used in the treatment of tumors and arrhythmias, and provide a novel possible strategy to reduce the occurrence of perioperative cardiovascular adverse events in patients with tumors such as esophageal cancer and gastric cancer.
Collapse
Affiliation(s)
- Zuojing Zhang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yuxin Zhang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xingji You
- School of Medicine, Shanghai University, Shanghai, China
| | - Jingxiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Loss of Ecrg4 improves calcium oxalate nephropathy. PLoS One 2022; 17:e0275972. [PMID: 36227903 PMCID: PMC9560046 DOI: 10.1371/journal.pone.0275972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Kidney stone is one of the most frequent urinary tract diseases, affecting 10% of the population and displaying a high recurrence rate. Kidney stones are the result of salt supersaturation, including calcium and oxalate. We have previously identified Esophageal cancer-related gene 4 (Ecrg4) as being modulated by hypercalciuria. Ecrg4 was initially described as a tumor suppressor gene in the esophagus. Lately, it was shown to be involved as well in apoptosis, cell senescence, cell migration, inflammation and cell responsiveness to chemotherapy. To the best of our knowledge, nothing is known about ECRG4's function in the renal tissue and its relationship with calciuria. We hypothesized that the increased expression of Ecrg4 mRNA is triggered by hypercalciuria and might modulate intratubular calcium-oxalate precipitation. In this study, we have first (i) validated the increased Ecrg4 mRNA in several types of hypercalciuric mouse models, then (ii) described the Ecrg4 mRNA expression along the nephron and (iii) assessed ECRG4's putative role in calcium oxalate nephropathy. For this, Ecrg4 KO mice were challenged with a kidney stone-inducing diet, rich in calcium and oxalate precursor. Taken together, our study demonstrates that Ecrg4's expression is restricted mainly to the distal part of the nephron and that the Ecrg4 KO mice develop less signs of tubular obstruction and less calcium-oxalate deposits. This promotes Ecrg4 as a modulator of renal crystallization and may open the way to new therapeutic possibilities against calcium oxalate nephropathy.
Collapse
|
8
|
Ruggiero C, Durand N, Jarjat M, Barhanin J, Ghirardello EJ, Dack MR, Williams GR, Bassett JD, Lalli E. The secreted protein augurin is a novel modulator of canonical Wnt signalling involved in osteoblast differentiation. CLINICAL AND TRANSLATIONAL DISCOVERY 2022; 2. [DOI: 10.1002/ctd2.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/02/2022] [Indexed: 01/21/2025]
Abstract
AbstractBackgroundECRG4/C2ORF40 is a tumour suppressor gene downregulated in several cancer types, which encodes the secreted protein augurin. A wide number of functions in health and disease have been assigned to augurin, but the signalling pathways it regulates are still poorly characterized. Augurin expression is strongly upregulated during in vitro differentiation of neonatal mouse osteoblasts.MethodsIn vitro differentiation assays of calvarial osteoblasts isolated from Ecrg4 ‐/‐ and wild‐type mice; transient transfection assays using reporters activated by Wnt signalling and other signal transduction pathways; Real‐time quantitative polymerase chain reaction for measurement of gene expression; protein expression in Chinese hamster ovary cells and Escherichia coli; in situ binding assays of proteins expressed as fusions to alkaline phosphatase with cells expressing various membrane receptors.ResultsOsteoblasts from Ecrg4 ‐/‐ mice have an accelerated differentiation compared to wild‐type and upregulation of Wnt markers. Augurin is a specific repressor of Wnt‐stimulated transcriptional activity, both when coexpressed together with the reporter and when added to the culture medium as a soluble protein. We confirmed the previously described binding of augurin to LOX1, a scavenger receptor, but an inhibitor of this molecule did not impair augurin repression of Wnt‐stimulated transcription specifically. Genome‐wide association studies showed an association of ECRG4 genomic variation with body height and osteoarthritis.ConclusionsOur study sheds new light on the wide spectrum of functions previously ascribed to augurin in brain function, stem cell biology, inflammation/immunity and cancer. Furthermore, our discovery paves the way to further characterization of the mechanisms involved in augurin repression of Wnt signalling and the development of agonists and antagonists for this protein, which have a wide array of potential applications in the clinic.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275 Valbonne France
- Université Côte d'Azur Valbonne France
| | - Nelly Durand
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275 Valbonne France
- Université Côte d'Azur Valbonne France
| | - Marielle Jarjat
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275 Valbonne France
- Université Côte d'Azur Valbonne France
| | - Jacques Barhanin
- Université Côte d'Azur Valbonne France
- LP2M CNRS UMR 7370 Nice France
| | - Elena J. Ghirardello
- Molecular Endocrinology Laboratory Department of Metabolism Digestion and Reproduction Imperial College London London UK
| | - Michael R.G. Dack
- Molecular Endocrinology Laboratory Department of Metabolism Digestion and Reproduction Imperial College London London UK
| | - Graham R. Williams
- Molecular Endocrinology Laboratory Department of Metabolism Digestion and Reproduction Imperial College London London UK
| | - J.H. Duncan Bassett
- Molecular Endocrinology Laboratory Department of Metabolism Digestion and Reproduction Imperial College London London UK
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275 Valbonne France
- Université Côte d'Azur Valbonne France
- Inserm Valbonne France
| |
Collapse
|
9
|
Yang Z, Ye X, Zhang Y, Huang Y, Chen J, Zeng Y, Chen J. ECRG4 acts as a tumor suppressor in nasopharyngeal carcinoma by suppressing the AKT/GSK3β/β-catenin signaling pathway. Cytotechnology 2022; 74:231-243. [PMID: 35464163 PMCID: PMC8976024 DOI: 10.1007/s10616-022-00520-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/17/2022] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED Nasopharyngeal carcinoma (NPC) is a malignant tumor with a poor prognosis. Studies have shown that esophageal carcinoma related gene 4 (ECRG4) is hypermethylated and significantly downregulated in NPC tissues. However, the role of ECRG4 in NPC, and in particular the underlying molecular mechanism, is largely unclear. In this study, using immunohistochemical staining of ECRG4 in NPC and normal specimens, we confirmed that ECRG4 was downregulated in human NPC tissues. In addition, various biological and molecular studies were carried out and the results showed that ECRG4 exerted anticancer effect in NPC, including inhibiting cell growth, migration, and invasion of NPC cells in vitro. Moreover, restoring ECRG4 expression suppressed the in vivo tumorigenesis of CNE2 cells. ECRG4 inhibited AKT/GSK3β/β-catenin signaling, as well as the downstream targets of β-catenin. LiCl treatment, which reduced GSK3β phosphorylation and upregulated β-catenin expression, restored the invasive ability of ECRG4-overexpressing NPC cells. Furthermore, we showed that the DNA methylation inhibitor 5-aza-dC reduced ECRG4 methylation and the invasive ability of negative control cells, but not that of ECRG4-overexpressing cells, suggesting that the inhibitory effect of 5-aza-dC depends on low expression of ECRG4. Collectively, our results demonstrated that ECRG4 downregulation contributed to NPC growth and invasion by activating AKT/GSK3β/β-catenin signaling pathway. ECRG4 could be a promising therapeutic target for the treatment of NPC. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s10616-022-00520-8.
Collapse
Affiliation(s)
- Zhengyuan Yang
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515031 People’s Republic of China
| | - Xiajun Ye
- Department of Medical Affairs, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515031 People’s Republic of China
| | - Yujie Zhang
- Department of Chemical Engineering, Guangdong Technion-Israel Institute of Technology, No. 241 Daxue Road, Shantou, 515031 People’s Republic of China
| | - Yiteng Huang
- Health Care Center, The First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041 People’s Republic of China
| | - Jian Chen
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. Raoping Road, Shantou, 515031 People’s Republic of China
| | - Yunzhu Zeng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515031 Guangdong Province People’s Republic of China
| | - Jiongyu Chen
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515031 Guangdong Province People’s Republic of China
| |
Collapse
|
10
|
Sharma A, Khan H, Singh TG, Grewal AK, Najda A, Kawecka-Radomska M, Kamel M, Altyar AE, Abdel-Daim MM. Pharmacological Modulation of Ubiquitin-Proteasome Pathways in Oncogenic Signaling. Int J Mol Sci 2021; 22:ijms222111971. [PMID: 34769401 PMCID: PMC8584958 DOI: 10.3390/ijms222111971] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/20/2022] Open
Abstract
The ubiquitin-proteasome pathway (UPP) is involved in regulating several biological functions, including cell cycle control, apoptosis, DNA damage response, and apoptosis. It is widely known for its role in degrading abnormal protein substrates and maintaining physiological body functions via ubiquitinating enzymes (E1, E2, E3) and the proteasome. Therefore, aberrant expression in these enzymes results in an altered biological process, including transduction signaling for cell death and survival, resulting in cancer. In this review, an overview of profuse enzymes involved as a pro-oncogenic or progressive growth factor in tumors with their downstream signaling pathways has been discussed. A systematic literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on modulation of ubiquitin-proteasome pathways in oncogenic signaling. Various in vitro, in vivo studies demonstrating the involvement of ubiquitin-proteasome systems in varied types of cancers and the downstream signaling pathways involved are also discussed in the current review. Several inhibitors of E1, E2, E3, deubiquitinase enzymes and proteasome have been applied for treating cancer. Some of these drugs have exhibited successful outcomes in in vivo studies on different cancer types, so clinical trials are going on for these inhibitors. This review mainly focuses on certain ubiquitin-proteasome enzymes involved in developing cancers and certain enzymes that can be targeted to treat cancer.
Collapse
Affiliation(s)
- Anmol Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (A.S.); (H.K.); (A.K.G.)
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (A.S.); (H.K.); (A.K.G.)
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (A.S.); (H.K.); (A.K.G.)
- Correspondence: or (T.G.S.); (M.M.A.-D.); Tel.: +91-9815951171 (T.G.S.); +966-580192142 (M.M.A.-D.)
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, India; (A.S.); (H.K.); (A.K.G.)
| | - Agnieszka Najda
- Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 50A Doświadczalna Street, 20-280 Lublin, Poland; (A.N.); (M.K.-R.)
| | - Małgorzata Kawecka-Radomska
- Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 50A Doświadczalna Street, 20-280 Lublin, Poland; (A.N.); (M.K.-R.)
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Ahmed E. Altyar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia;
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: or (T.G.S.); (M.M.A.-D.); Tel.: +91-9815951171 (T.G.S.); +966-580192142 (M.M.A.-D.)
| |
Collapse
|
11
|
You Y, Hu S. Dysregulation of ECRG4 is associated with malignant properties and of prognostic importance in human gastric cancer. Cancer Biomark 2021; 34:55-66. [PMID: 34657878 DOI: 10.3233/cbm-210334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND We have previously characterized esophageal carcinoma-related gene 4 (ECRG4) as a novel tumor suppressor gene, which is frequently inactivated in nasopharyngeal carcinoma and breast cancer. Nevertheless, the expression status and prognostic significance of ECRG4 maintain elusive in human gastric cancer. Herein, we examined ECRG4 expression profile in gastric cancer and assessed its association with clinicopathological characteristics and patient survival. METHODS Online data mining, real-time RT-PCR and immunohistochemistry were employed to determined ECRG4 expression at transcriptional and protein levels in tumors vs. noncancerous tissues. Statistical analyses including the Kaplan-Meier survival analysis and the Cox hazard model were utilized to detect the impact on clinical outcome. Moreover, ECRG4 expression was silenced in gastric cancer SGC7901 cells, and cell proliferation, colony formation and invasion assays were carried out. RESULTS ECRG4 mRNA and protein levels were obviously downregulated in cancer tissues than noncancerous tissues. Statistical analyses demonstrated that low ECRG4 expression was found in 34.5% (58/168) of primary gastric cancer tissues, which was associated with higher histological grade (P= 0.018), lymph node metastasis (P= 0.011), invasive depth (P= 0.020), advanced tumor stage (P= 0.002) and poor overall survival (P< 0.001). Multivariate analysis showed ECRG4 expression is an independent prognostic predictor (P< 0.001). Silencing ECRG4 expression promoted gastric cancer cell growth and invasion. Western blot analysis revealed the anti-metastatic functions of ECRG4 by downregulating of E-cadherin and α-Catenin, as well as upregulating N-cadherin and Vimentin. CONCLUSIONS Our observations reveal that ECRG4 expression is involved in gastric cancer pathogenesis and progression, and may serve as a candidate prognostic biomarker for this disease.
Collapse
Affiliation(s)
- Yanjie You
- Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, Ningxia Hui Autonomous Region, China.,Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, Ningxia Hui Autonomous Region, China
| | - Shengjuan Hu
- Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, Ningxia Hui Autonomous Region, China.,Endoscopy Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, Ningxia Hui Autonomous Region, China.,Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, Ningxia Hui Autonomous Region, China
| |
Collapse
|
12
|
Huo H, Yang S, Wu H, Sun Y, Zhao R, Ye R, Yan D, Shi X, Yang J. Brain endothelial cells-derived extracellular vesicles overexpressing ECRG4 inhibit glioma proliferation through suppressing inflammation and angiogenesis. J Tissue Eng Regen Med 2021; 15:1162-1171. [PMID: 34551201 DOI: 10.1002/term.3244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/22/2021] [Accepted: 09/02/2021] [Indexed: 12/27/2022]
Abstract
Esophageal cancer related gene-4 (ECRG4) has been shown to be a candidate tumor suppressor in many tumors, but its role in glioma remains poorly understood. This study aimed to explore whether extracellular vesicles (EVs) derived from brain endothelial cells which overexpressed ECRG4 have anti-tumor effect on gliomas in vivo and in vitro, as well as the possible mechanism. A constructed lentivirus expressing the ECRG4 gene was transfected into the hCMEC/D3 cell line. The EVs were isolated from the cells and characterized by Western blot with exosome markers of CD9, CD63, CD81, Alix. RT-PCR and Western blot were performed to verify ECRG4 expression. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and clone formation assays were applied to detect the proliferation of glioma cells incubated with EVs expressing the ECRG4 (ECRG4-exo). The level of inflammatory cytokines and angiogenesis related factors, including nuclear factor kappa-B (NF-κB), interleukin (IL)-1β, IL-6, IL-8, monocyte chemoattractant protein-1 (MCP-1), hypoxia-inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR2) levels were detected by ELISA. The T98G cell xenograft mouse model was established and treated with ECRG4-EV. The tumor volume and weight were recorded. p38-MAPK, p-p38-MAPK proteins were determined by Western blot in tumor tissues. As a result, EVs can be internalized into U87MG and T98G cells. ECRG4-EV inhibited U87MG and T98G cell proliferation. ECRG4-EV also inhibited the expression of factors involved in inflammation and angiogenesis. In addition, ECRG4-EVs suppressed tumor growth and decreased the production of inflammatory cytokines through inactivation of p38-MAPK signal pathway. In conclusion, ECRG4-EVsuppresses glioma proliferation through modulating the inflammation and angiogenesis.
Collapse
Affiliation(s)
- Haoran Huo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Song Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haotian Wu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuchen Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ranran Zhao
- Department of Hemodialysis, Traditional Chinese Medicine Hospital of Shi Jia Zhuang City, Shijiazhuang, Hebei, China
| | - Ruihao Ye
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dongdong Yan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xuefang Shi
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiankai Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
13
|
Zou Z, Zheng W, Fan H, Deng G, Lu SH, Jiang W, Yu X. Aspirin enhances the therapeutic efficacy of cisplatin in oesophageal squamous cell carcinoma by inhibition of putative cancer stem cells. Br J Cancer 2021; 125:826-838. [PMID: 34316020 PMCID: PMC8438052 DOI: 10.1038/s41416-021-01499-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 06/08/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are related to the patient's prognosis, recurrence and therapy resistance in oesophageal squamous cell carcinoma (ESCC). Although increasing evidence suggests that aspirin (acetylsalicylic acid, ASA) could lower the incidence and improve the prognosis of ESCC, the mechanism(s) remains to be fully understood. METHODS We investigated the role of ASA in chemotherapy/chemoprevention in human ESCC cell lines and an N-nitrosomethylbenzylamine-induced rat ESCC carcinogenesis model. The effects of combined treatment with ASA/cisplatin on ESCC cell lines were examined in vitro and in vivo. Sphere-forming cells enriched with putative CSCs (pCSCs) were used to investigate the effect of ASA in CSCs. Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) was performed to determine the alterations in chromatin accessibility caused by ASA in ESCC cells. RESULTS ASA inhibits the CSC properties and enhances cisplatin treatment in human ESCC cells. ATAC-seq indicates that ASA treatment results in remarkable epigenetic alterations on chromatin in ESCC cells, especially their pCSCs, through the modification of histone acetylation levels. The epigenetic changes activate Bim expression and promote cell death in CSCs of ESCC. Furthermore, ASA prevents the carcinogenesis of NMBzA-induced ESCC in the rat model. CONCLUSIONS ASA could be a potential chemotherapeutic adjuvant and chemopreventive drug for ESCC treatment.
Collapse
Affiliation(s)
- Zhigeng Zou
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Zheng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongjun Fan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guodong Deng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shih-Hsin Lu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Jiang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiying Yu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Prieto-Vila M, Yoshioka Y, Ochiya T. Biological Functions Driven by mRNAs Carried by Extracellular Vesicles in Cancer. Front Cell Dev Biol 2021; 9:620498. [PMID: 34527665 PMCID: PMC8435577 DOI: 10.3389/fcell.2021.620498] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 07/30/2021] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are extracellular nanovesicles released by most cells. EVs play essential roles in intercellular communication via the transport of a large variety of lipids, proteins, and nucleic acids to recipient cells. Nucleic acids are the most commonly found molecules inside EVs, and due to their small size, microRNAs and other small RNAs are the most abundant nucleic acids. However, longer molecules, such as messenger RNAs (mRNAs), have also been found. mRNAs encapsulated within EVs have been shown to be transferred to recipient cells and translated into proteins, altering the behavior of the cells. Secretion of EVs is maintained not only through multiple normal physiological conditions but also during aberrant pathological conditions, including cancer. Recently, the mRNAs carried by EVs in cancer have attracted great interest due to their broad roles in tumor progression and microenvironmental remodeling. This review focuses on the biological functions driven by mRNAs carried in EVs in cancer, which include supporting tumor progression by activating cancer cell growth, migration, and invasion; inducing microenvironmental remodeling via hypoxia, angiogenesis, and immunosuppression; and promoting modulation of the microenvironment at distant sites for the generation of a premetastatic niche, collectively inducing metastasis. Furthermore, we describe the potential use of mRNAs carried by EVs as a noninvasive diagnostic tool and novel therapeutic approach.
Collapse
Affiliation(s)
| | | | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
15
|
Glioblastoma-initiating cell heterogeneity generated by the cell-of-origin, genetic/epigenetic mutation and microenvironment. Semin Cancer Biol 2021; 82:176-183. [PMID: 33453403 DOI: 10.1016/j.semcancer.2020.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/27/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma (GBM) and other malignant tumours consist of heterogeneous cancer cells, including GBM-initiating cells (GICs). This heterogeneity is likely to arise from the following: different sets of genetic mutations and epigenetic modifications, which GICs gain in the transformation process; differences in cells of origin, such as stem cells, precursor cells or differentiated cells; and the cancer microenvironment, in which GICs communicate with neural cells, endothelial cells and immune cells. Furthermore, considering that various types of GICs can be generated at different time points of the transformation process, GBM very likely consists of heterogeneous GICs and their progeny. Because cancer cell heterogeneity is responsible for therapy resistance, it is crucial to develop methods of reducing such heterogeneity. Here, I summarize how GIC heterogeneity is generated in the transformation process and present how cell heterogeneity in cancer can be addressed based on recent findings.
Collapse
|
16
|
Lin L, Cheng X, Yin D. Aberrant DNA Methylation in Esophageal Squamous Cell Carcinoma: Biological and Clinical Implications. Front Oncol 2020; 10:549850. [PMID: 33194605 PMCID: PMC7645039 DOI: 10.3389/fonc.2020.549850] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/18/2020] [Indexed: 12/20/2022] Open
Abstract
Almost all cancer cells possess multiple epigenetic abnormalities, which cooperate with genetic alterations to enable the acquisition of cancer hallmarks during tumorigenesis. As the most frequently found epigenetic change in human cancers, aberrant DNA methylation manifests at two major forms: global genomic DNA hypomethylation and locus-specific promoter region hypermethylation. It has been recognized as a critical contributor to esophageal squamous cell carcinoma (ESCC) malignant transformation. In ESCC, DNA methylation alterations affect genes involved in cell cycle regulation, DNA damage repair, and cancer-related signaling pathways. Aberrant DNA methylation patterns occur not only in ESCC tumors but also in precursor lesions. It adds another layer of complexity to the ESCC heterogeneity and may serve as early diagnostic, prognostic, and chemo-sensitive markers. Characterization of the DNA methylome in ESCC could help better understand its pathogenesis and develop improved therapies. We herein summarize the current research and knowledge about DNA methylation in ESCC and its clinical significance in diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Lehang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xu Cheng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Liao L, Yao Z, Fang W, He Q, Xu WW, Li B. Epigenetics in Esophageal Cancer: From Mechanisms to Therapeutics. SMALL METHODS 2020; 4:2000391. [DOI: 10.1002/smtd.202000391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Indexed: 02/05/2023]
Affiliation(s)
- Long Liao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes Institute of Life and Health Engineering College of Life Science and Technology Jinan University Guangzhou 510632 China
| | - Zi‐Ting Yao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes Institute of Life and Health Engineering College of Life Science and Technology Jinan University Guangzhou 510632 China
| | - Wang‐Kai Fang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area Department of Biochemistry and Molecular Biology Shantou University Medical College Shantou 515041 China
| | - Qing‐Yu He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes Institute of Life and Health Engineering College of Life Science and Technology Jinan University Guangzhou 510632 China
| | - Wen Wen Xu
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine National Engineering Research Center of Genetic Medicine Institute of Biomedicine College of Life Science and Technology Jinan University Guangzhou 510632 China
| | - Bin Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes Institute of Life and Health Engineering College of Life Science and Technology Jinan University Guangzhou 510632 China
| |
Collapse
|
18
|
Liang X, Gao J, Wang Q, Hou S, Wu C. ECRG4 Represses Cell Proliferation and Invasiveness via NFIC/OGN/NF-κB Signaling Pathway in Bladder Cancer. Front Genet 2020; 11:846. [PMID: 32922434 PMCID: PMC7456849 DOI: 10.3389/fgene.2020.00846] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022] Open
Abstract
Bladder cancer (BCa) is a malignant tumor in the urinary system with high cancer-related mortality worldwide. However, the molecular mechanisms of many genes dysregulated in BCa are still unclear. Herein, we showed that esophageal cancer-related gene-4 (ECRG4), which is downregulated in BCa tissues and cell lines, has a positive correlation with osteoglycin (OGN). Further functional experimental studies suggested that both ECRG4 and OGN inhibit cell proliferation, migration, and invasion in BCa cells. Moreover, ECRG4 acts as a tumor repressor and promotes the expression of OGN via the upregulation of nuclear factor 1 C-type (NFIC), which can bind to the promoter region of OGN and regulate its transcription. Bioinformatics analysis revealed that NFIC is downregulated in BCa tissues and has a positive correlation with ECRG4 or OGN. Esophageal cancer-related gene-4 could positively regulate the protein levels of NFIC in BCa cells. In addition, we demonstrated for the first time that ECRG4 inhibits the nuclear factor (NF)-κB signaling pathway via the upregulation of OGN in BCa cells. Overall, these findings provide evidence that both ECRG4 and OGN function as tumor repressors and that overexpression of ECRG4 inhibits the NF-κB signaling pathway by promoting NFIC/OGN signaling in BCa cells. Our results reveal the molecular regulatory mechanisms of the ECRG4-mediated repression of the NFIC/OGN/NF-κB signaling pathway in BCa and provide potential biomarkers or therapeutic targets for BCa.
Collapse
Affiliation(s)
- Xin Liang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.,Department of Urology, Qingdao Municipal Hospital, Qingdao, China
| | - Jiangang Gao
- Department of Urology, Qingdao Municipal Hospital, Qingdao, China
| | - Quan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao, China
| | - Sichuan Hou
- Department of Urology, Qingdao Municipal Hospital, Qingdao, China
| | - Changli Wu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
19
|
Fan H, Yu X, Zou Z, Zheng W, Deng X, Guo L, Jiang W, Zhan Q, Lu SH. Metformin suppresses the esophageal carcinogenesis in rats treated with NMBzA through inhibiting AMPK/mTOR signaling pathway. Carcinogenesis 2020; 40:669-679. [PMID: 30445633 DOI: 10.1093/carcin/bgy160] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/01/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
Metformin is a widely used antidiabetic drug for the management of type 2 diabetes mellitus. Recently, epidemiological studies demonstrate that metformin has anticancer effects on esophageal squamous cell carcinoma (ESCC) and other cancers. However, the effects and potential mechanisms of metformin on ESCC remain elusive. In this study, we used N-nitroso-N-methylbenzylamine (NMBzA), a special carcinogen for esophagi, to develop a rat ESCC model, in which the carcinogenesis progression of ESCC in rat was induced and promoted. We investigated the effects of metformin on carcinogenesis of ESCC in this model. Our results revealed that metformin significantly decreased the incidence and precancerous lesions of ESCC and inhibited proliferation and promoted apoptosis of esophageal epithelial cells in rat treated with NMBzA. Moreover, metformin also increased apoptosis and inhibited migration, colony formation and tumor sphere formation of human ESCC cells in vitro. Immunohistochemistry and western blotting showed that without interfering the metabolism of NMBzA, metformin inhibited the inflammation of esophagi via reducing the expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and interleukin-6 (IL-6). Treatment of metformin led to activation of AMP-activated protein kinase (AMPK) and attenuated signaling of the downstream molecules such as p-mTOR, p-p70S6K and cyclin D1 expression both in vivo and in vitro. Taken together, our study demonstrated that metformin suppressed the carcinogenesis of ESCC through inhibiting AMPK/mammalian target of the rapamycin (mTOR) signaling pathway, resulting in its chemopreventive effects on the carcinogenesis of ESCC.
Collapse
Affiliation(s)
- Hongjun Fan
- Department of Etiology and Carcinogenesis and State Key Laboratory of Molecular Oncology.,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiying Yu
- Department of Etiology and Carcinogenesis and State Key Laboratory of Molecular Oncology.,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhigeng Zou
- Department of Etiology and Carcinogenesis and State Key Laboratory of Molecular Oncology.,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Zheng
- Department of Etiology and Carcinogenesis and State Key Laboratory of Molecular Oncology.,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Department of Etiology and Carcinogenesis and State Key Laboratory of Molecular Oncology.,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liping Guo
- Department of Etiology and Carcinogenesis and State Key Laboratory of Molecular Oncology.,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Jiang
- Department of Etiology and Carcinogenesis and State Key Laboratory of Molecular Oncology
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shih-Hsin Lu
- Department of Etiology and Carcinogenesis and State Key Laboratory of Molecular Oncology.,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Rose M, Bringezu S, Godfrey L, Fiedler D, Gaisa NT, Koch M, Bach C, Füssel S, Herr A, Hübner D, Ellinger J, Pfister D, Knüchel R, Wirth MP, Böhme M, Dahl E. ITIH5 and ECRG4 DNA Methylation Biomarker Test (EI-BLA) for Urine-Based Non-Invasive Detection of Bladder Cancer. Int J Mol Sci 2020; 21:ijms21031117. [PMID: 32046186 PMCID: PMC7036997 DOI: 10.3390/ijms21031117] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023] Open
Abstract
Bladder cancer is one of the more common malignancies in humans and the most expensive tumor for treating in the Unites States (US) and Europe due to the need for lifelong surveillance. Non-invasive tests approved by the FDA have not been widely adopted in routine diagnosis so far. Therefore, we aimed to characterize the two putative tumor suppressor genes ECRG4 and ITIH5 as novel urinary DNA methylation biomarkers that are suitable for non-invasive detection of bladder cancer. While assessing the analytical performance, a spiking experiment was performed by determining the limit of RT112 tumor cell detection (range: 100-10,000 cells) in the urine of healthy donors in dependency of the processing protocols of the RWTH cBMB. Clinically, urine sediments of 474 patients were analyzed by using quantitative methylation-specific PCR (qMSP) and Methylation Sensitive Restriction Enzyme (MSRE) qPCR techniques. Overall, ECRG4-ITIH5 showed a sensitivity of 64% to 70% with a specificity ranging between 80% and 92%, i.e., discriminating healthy, benign lesions, and/or inflammatory diseases from bladder tumors. When comparing single biomarkers, ECRG4 achieved a sensitivity of 73%, which was increased by combination with the known biomarker candidate NID2 up to 76% at a specificity of 97%. Hence, ITIH5 and, in particular, ECRG4 might be promising candidates for further optimizing current bladder cancer biomarker panels and platforms.
Collapse
Affiliation(s)
- Michael Rose
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.B.); (L.G.); (D.F.); (N.T.G.); (M.K.); (R.K.)
- RWTH Centralized Biomaterial Bank (RWTH cBMB), Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
- Correspondence: (M.R.); (E.D.); Tel.: +49-241-808-9715 (M.R.); +49-241-808-8431 (E.D.); Fax: +49-241-808-2439 (M.R.); +49-241-808-2439 (E.D.)
| | - Sarah Bringezu
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.B.); (L.G.); (D.F.); (N.T.G.); (M.K.); (R.K.)
| | - Laura Godfrey
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.B.); (L.G.); (D.F.); (N.T.G.); (M.K.); (R.K.)
| | - David Fiedler
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.B.); (L.G.); (D.F.); (N.T.G.); (M.K.); (R.K.)
| | - Nadine T. Gaisa
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.B.); (L.G.); (D.F.); (N.T.G.); (M.K.); (R.K.)
| | - Maximilian Koch
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.B.); (L.G.); (D.F.); (N.T.G.); (M.K.); (R.K.)
| | - Christian Bach
- Department of Urology, RWTH Aachen University, 52074 Aachen, Germany; (C.B.); (D.P.)
| | - Susanne Füssel
- Department of Urology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.F.); (D.H.); (M.P.W.)
| | | | - Doreen Hübner
- Department of Urology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.F.); (D.H.); (M.P.W.)
| | - Jörg Ellinger
- Department of Urology, University Hospital Bonn, 53105 Bonn, Germany;
| | - David Pfister
- Department of Urology, RWTH Aachen University, 52074 Aachen, Germany; (C.B.); (D.P.)
- Department of Urology, Uro-Oncology, Robot Assisted and Reconstructive Urologic Surgery, University Hospital Cologne, 50937 Cologne, Germany
| | - Ruth Knüchel
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.B.); (L.G.); (D.F.); (N.T.G.); (M.K.); (R.K.)
| | - Manfred P. Wirth
- Department of Urology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.F.); (D.H.); (M.P.W.)
| | - Manja Böhme
- Biotype GmbH, 01109 Dresden, Germany; (A.H.); (M.B.)
| | - Edgar Dahl
- Institute of Pathology, RWTH Aachen University, 52074 Aachen, Germany; (S.B.); (L.G.); (D.F.); (N.T.G.); (M.K.); (R.K.)
- RWTH Centralized Biomaterial Bank (RWTH cBMB), Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
- Correspondence: (M.R.); (E.D.); Tel.: +49-241-808-9715 (M.R.); +49-241-808-8431 (E.D.); Fax: +49-241-808-2439 (M.R.); +49-241-808-2439 (E.D.)
| |
Collapse
|
21
|
Li D, Liu X, Liu T, Liu H, Tong L, Jia S, Wang YF. Neurochemical regulation of the expression and function of glial fibrillary acidic protein in astrocytes. Glia 2019; 68:878-897. [PMID: 31626364 DOI: 10.1002/glia.23734] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/27/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022]
Abstract
Glial fibrillary acidic protein (GFAP), a type III intermediate filament, is a marker of mature astrocytes. The expression of GFAP gene is regulated by many transcription factors (TFs), mainly Janus kinase-2/signal transducer and activator of transcription 3 cascade and nuclear factor κ-light-chain-enhancer of activated B cell signaling. GFAP expression is also modulated by protein kinase and other signaling molecules that are elicited by neuronal activity and hormones. Abnormal expression of GFAP proteins occurs in neuroinflammation, neurodegeneration, brain edema-eliciting diseases, traumatic brain injury, psychiatric disorders and others. GFAP, mainly in α-isoform, is the major component of cytoskeleton and the scaffold of astrocytes, which is essential for the maintenance of astrocytic structure and shape. GFAP also has highly morphological plasticity because of its quick changes in assembling and polymerizing states in response to environmental challenges. This plasticity and its corresponding cellular morphological changes endow astrocytes the functions of physical barrier between adjacent neurons and stabilizer of extracellular environment. Moreover, GFAP colocalizes and even molecularly associates with many functional molecules. This feature allows GFAP to function as a platform for direct interactions between different molecules. Last, GFAP involves transportation and localization of other functional proteins and thus serves as a protein transport guide in astrocytes. This guiding role of GFAP involves an elastic retraction and extension cytoskeletal network that couples with GFAP reassembling, transporting, and membrane protein recycling machinery. This paper reviews our current understanding of the expression and functions of GFAP as well as their regulation.
Collapse
Affiliation(s)
- Dongyang Li
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Xiaoyu Liu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Tianming Liu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Haitao Liu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Li Tong
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Shuwei Jia
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Yu-Feng Wang
- Department of Physiology, Harbin Medical University, Harbin, China
| |
Collapse
|
22
|
Potential functions of esophageal cancer-related gene-4 in the cardiovascular system. Front Med 2019; 13:639-645. [PMID: 31468282 DOI: 10.1007/s11684-019-0701-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/21/2019] [Indexed: 02/07/2023]
Abstract
Esophageal cancer-related gene-4 (Ecrg4) is cloned from the normal epithelium of the esophagus. It is constitutively expressed in quiescent epithelial cells and downregulated during tumorigenesis, and Ecrg4 expression levels are inversely correlated with the malignant phenotype of tumor cells, validating that Ecrg4 is a real tumor suppressor gene. Unlike other tumor suppressor genes that usually encode membrane or intracellular proteins, Ecrg4 encodes a 148-amino acid pre-pro-peptide that is tethered on the cell surface in epithelial cells, specialized epithelial cells, and human leukocytes, where it can be processed tissue dependently into several small peptides upon cell activation. Ecrg4 is expressed in a wide variety of other cells/tissues, including cardiomyocytes and conduction system of the heart, the glomus cells of the carotid body, adrenal glands, choroid plexus, and leukocytes among others, where it exerts distinct functions, such as promoting/suppressing inflammation, inducing neuron senescence, stimulating the hypothalamus-pituitary-adrenal axis, maintaining the stemness of stem cells, participating in the rhythm and rate control of the heart, and possibly gauging the responsiveness of the cardiovascular system (CVS) to hypoxia, in addition to tumor suppression. Here, we briefly review the latest discoveries on Ecrg4 and its underlying molecular mechanisms as a tumor suppressor and focus on the emerging roles of Ecrg4 in the CVS.
Collapse
|
23
|
Huang W, Zhou R, Mao L, Deng C, Dang X. Esophageal cancer related gene-4 inhibits the migration and proliferation of oral squamous cell carcinoma through BC200 lncRNA/MMP-9 and -13 signaling pathway. Cell Signal 2019; 62:109327. [PMID: 31152845 DOI: 10.1016/j.cellsig.2019.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/22/2022]
Abstract
Esophageal cancer related gene-4 (ECRG4) inhibits the malignant phenotype of oral squamous cell carcinoma. However, the molecular mechanisms remain to be explored. Using the tongue carcinoma cell line, TCA8113 as a cell model, we showed that forced expression of ECRG4 down-regulated the expression of the BC200 long non-coding RNA (lncRNA) and matrix metalloproteinases (MMP-9 and MMP-13). Restoration of BC200 lncRNA rescued ECRG4-mediated down-regulation of MMP-9 and -13. Furthermore, over-expression of Ecrg4 inhibited cell proliferation and migration, which was abolished by forced expression of BC200 lncRNA in TCA8113 cells. Our results indicate that ECRG4 inhibits the malignant phenotype of TCA8113 cells most likely through suppression of BC200 lncRNA/MMPs signaling pathway, rationalizing that BC200 lncRNA may be a potential target for oral squamous cell carcinoma (OSCC) therapy.
Collapse
Affiliation(s)
- Wenjun Huang
- The Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Rui Zhou
- The Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Liang Mao
- The Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chenliang Deng
- Department of Plastic Surgery, Shanghai 6th People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200237, China.
| | - Xitong Dang
- The Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
24
|
Nakatani Y, Kiyonari H, Kondo T. Ecrg4 deficiency extends the replicative capacity of neural stem cells in a Foxg1-dependent manner. Development 2019; 146:dev.168120. [PMID: 30745428 DOI: 10.1242/dev.168120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 01/28/2019] [Indexed: 02/03/2023]
Abstract
The self-renewal activity of neural stem cells (NSCs) has been suggested to decrease with aging, resulting in age-dependent declines in brain function, such as presbyopia and memory loss. The molecular mechanisms underlying decreases in NSC proliferation with age need to be elucidated in more detail to develop treatments that promote brain function. We have previously reported that the expression of esophageal cancer-related gene 4 (Ecrg4) was upregulated in aged NSCs, whereas its overexpression decreased NSC proliferation, suggesting a functional relationship between Ecrg4 and NSC aging. Using Ecrg4-deficient mice in which the Ecrg4 locus was replaced with the lacZ gene, we here show that Ecrg4 deficiency recovered the age-dependent decline in NSC proliferation and enhanced spatial learning and memory in the Morris water-maze paradigm. We demonstrate that the proliferation of Ecrg4-deficient NSCs was partly maintained by the increased expression of Foxg1. Collectively, these results determine Ecrg4 as a NSC aging factor.
Collapse
Affiliation(s)
- Yuka Nakatani
- Division of Bio-Function Dynamics Imaging, Center for Life Science Technology, RIKEN, Kobe, Hyogo 650-0047, Japan
| | - Hiroshi Kiyonari
- Animal Resource Development Unit and Genetic Engineering Team, Center for Life Science Technology, RIKEN, Kobe, Hyogo 650-0047, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| |
Collapse
|
25
|
Deng P, Chang XJ, Gao ZM, Xu XY, Sun AQ, Li K, Dai DQ. Downregulation and DNA methylation of ECRG4 in gastric cancer. Onco Targets Ther 2018; 11:4019-4028. [PMID: 30034241 PMCID: PMC6049055 DOI: 10.2147/ott.s161200] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Esophageal cancer-related gene 4 (ECRG4) is a novel candidate tumor suppressor gene. Our study investigated the expression and function of ECRG4 in gastric cancer and highlighted the role of DNA hypermethylation at the promoter in silencing the ECRG4 expression. Methods The GSE63089 data set was obtained from the Gene Expression Omnibus and analyzed for differentially expressed genes. Carcinoma and para-carcinoma tissues of 102 patients with gastric cancer were collected from January 2010 to July 2011. Immunohistochemistry, real-time polymerase chain reaction (PCR), and western blot analyses were performed to evaluate the expression of ECRG4. After measuring the change in the level of ECRG4 expression, CCK-8, Transwell, and flow cytometric cell cycle assays were performed. In addition, methylation-specific PCR was performed to detect the methylation state of ECRG4, and 5-aza-2′-deoxycytidine was used for demethylation of ECRG4. All statistical analyses were performed using the SPSS 17.0 software. Results We found that ECRG4 expression was downregulated in gastric cancer, and this was closely related to lymph node metastasis. After ECRG4 was silenced using a specific small interfering RNA, the BGC-823 cell line became highly aggressive and proliferative. In addition, we verified whether downregulation of ECRG4 was highly correlated with DNA methylation of the ECRG4 promoter and found that the demethylating agent 5-aza-2′-deoxycytidine could effectively enhance ECRG4 expression. Conclusion The aberrant expression of ECRG4 is associated with hypermethylation in the promoter region and plays an important role in the malignancy of gastric cancer. Therefore, ECRG4 may be a potential biomarker for molecular diagnosis of gastric cancer, and the use of 5-Aza-dC to reverse the hypermethylation of ECRG4 may be a new approach to the treatment of gastric cancer.
Collapse
Affiliation(s)
- Peng Deng
- Department of Gastrointestinal Surgery and Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, China,
| | - Xiao-Jing Chang
- Department of Gastrointestinal Surgery and Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, China,
| | - Zi-Ming Gao
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Yang Xu
- Department of Gastrointestinal Surgery and Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, China,
| | - An-Qi Sun
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Kai Li
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dong-Qiu Dai
- Department of Gastrointestinal Surgery and Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, China,
| |
Collapse
|
26
|
ECRG4: a new potential target in precision medicine. Front Med 2018; 13:540-546. [PMID: 30003403 DOI: 10.1007/s11684-018-0637-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/27/2018] [Indexed: 12/28/2022]
Abstract
Given the rapid development in precision medicine, tremendous efforts have been devoted to discovering new biomarkers for disease diagnosis and treatment. Esophageal cancer-related gene-4 (ECRG4), which is initially known as a new candidate tumor suppressor gene, is emerging as a sentinel molecule for gauging tissue homeostasis. ECRG4 is unique in its cytokine-like functional pattern and epigenetically-regulated gene expression pattern. The gene can be released from the cell membrane upon activation and detected in liquid biopsy, thus offering considerable potential in precision medicine. This review provides an updated summary on the biology of ECRG4, with emphasis on its important roles in cancer diagnosis and therapy. The future perspectives of ECRG4 as a potential molecular marker in precision medicine are also discussed in detail.
Collapse
|
27
|
Mao L, Li X, Gong S, Yuan H, Jiang Y, Huang W, Sun X, Dang X. Serum exosomes contain ECRG4 mRNA that suppresses tumor growth via inhibition of genes involved in inflammation, cell proliferation, and angiogenesis. Cancer Gene Ther 2018; 25:248-259. [PMID: 29983418 DOI: 10.1038/s41417-018-0032-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 05/14/2018] [Accepted: 05/17/2018] [Indexed: 02/07/2023]
Abstract
Esophageal cancer related gene-4 (Ecrg4) has been shown to be a tumor suppressor in many organs. Exosomes are naturally secreted nanosized particles that carry signal molecules including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and messenger RNAs (mRNAs) among others. Upon internalization, exosomes unload their cargos that in turn modulate the biology of the recipient cells. Mounting evidence has shown that exosomal miRNAs are functional. However, reports that exosomes carry functional mRNAs remain scarce. We found that serum exosomes contain ECRG4 open reading frame. To simulate serum exosomal ECRG4, stable cell line expressing ECRG4 was created, from which exosomes were isolated and characterized, and the internalization and the resulting biological effects of exosomal ECRG4 were evaluated. Results showed that serum exosomes contain higher levels of ECRG4 mRNA in healthy individuals than their cancer counterparts. Exosomal ECRG4 can be internalized and unload the encapsulated ECRG4 into recipient cells, which subsequently suppressed cell proliferation in vitro, and inhibited tumor growth in a xenograft mouse model. Mechanistically, ECRG4-containing exosomes, when internalized, suppressed the expression of genes commonly implicated in inflammation, cell proliferation, and angiogenesis. Given that exosome is an ideal vehicle for therapeutics delivery and that ECRG4 is a tumor suppressor gene, the exosomal ECRG4 can be exploited as a formulation for cancer gene therapy.
Collapse
Affiliation(s)
- Liang Mao
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xue Li
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shu Gong
- Department of Pathophysiology, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Haiyang Yuan
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yu Jiang
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Wenjun Huang
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xingwang Sun
- Department of Pathology, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xitong Dang
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
28
|
Li C, Zhang P, Jiang A, Mao JH, Wei G. A short synthetic peptide fragment of human C2ORF40 has therapeutic potential in breast cancer. Oncotarget 2018; 8:41963-41974. [PMID: 28410214 PMCID: PMC5522041 DOI: 10.18632/oncotarget.16713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022] Open
Abstract
C2ORF40 encodes a secreted protein which is cleaved to generate soluble peptides by proteolytic processing and this process is believed to be necessary for C2ORF40 to exert cell type specific biological activity. Here, we reported a short mimic peptide of human C2ORF40 acts potential therapeutic efficacy in human cancer cells in vitro and in vivo. We synthesized a short peptide of human C2ORF40, named C2ORF40 mimic peptide fragment and assessed its biological function on cancer cell growth, migration and tumorigenesis. Cell growth assay showed that C2ORF40 mimic peptide fragment significantly suppressed cell proliferation of breast and lung cancer cells. Moreover, C2ORF40 mimic peptide fragment significantly inhibited the migration and invasion of breast cancer cells. Furthermore, we showed that this peptide suppressed tumorigenesis in breast tumor xenograft model. Cell cycle assay indicated that the C2ORF40 mimic peptide fragment suppressed the growth of tumor cells through inducing mitotic phase arrest. In conclusion, our results firstly suggested that this short synthetic peptide of human C2ORF40 may be a candidate tumor therapeutic agent.
Collapse
Affiliation(s)
- Chaoyang Li
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Pengju Zhang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Anli Jiang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Guangwei Wei
- Department of Human Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, Shandong, 250012, P.R. China
| |
Collapse
|
29
|
Abstract
Esophageal cancer-related gene 4 (Ecrg4) encodes a hormone-like peptide that is believed to be involved in a variety of physiological phenomena, including tumour suppression. Recent progress in the study of Ecrg4 has shown that Ecrg4 is a proinflammatory factor and induces the expression of several cytokines and chemokines in macrophages/microglia. However, the detailed molecular mechanisms of Ecrg4 signalling, especially the Ecrg4 receptors, remain poorly understood. Here, using retrovirus-mediated expression cloning, we identified lectin-like oxidised low-density lipoprotein receptor-1 (LOX-1) as a membrane protein that binds amino acid residues 71–132 of Ecrg4 (Ecrg4(71–132)). Moreover, in addition to LOX-1, several scavenger receptors, such as Scarf1, Cd36 and Stabilin-1, facilitated the efficient internalisation of Ecrg4(71–132) into cells. A broad competitive inhibitor of scavenger receptors, polyinosinic acid, reduced both the binding of Ecrg4(71–132) and the activation of NF-κB in microglia. This activation was dependent on MyD88, an adaptor protein that recruits signalling proteins to Toll-like receptors (TLRs), with the consequent induction of various immune responses. These data suggest that multiple scavenger receptors recognise Ecrg4(71–132) and transduce its signals, together with TLRs, in microglia.
Collapse
|
30
|
Chung J, Zhang X, Allen M, Wang X, Ma Y, Beecham G, Montine TJ, Younkin SG, Dickson DW, Golde TE, Price ND, Ertekin-Taner N, Lunetta KL, Mez J, Mayeux R, Haines JL, Pericak-Vance MA, Schellenberg G, Jun GR, Farrer LA. Genome-wide pleiotropy analysis of neuropathological traits related to Alzheimer's disease. Alzheimers Res Ther 2018; 10:22. [PMID: 29458411 PMCID: PMC5819208 DOI: 10.1186/s13195-018-0349-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Simultaneous consideration of two neuropathological traits related to Alzheimer's disease (AD) has not been attempted in a genome-wide association study. METHODS We conducted genome-wide pleiotropy analyses using association summary statistics from the Beecham et al. study (PLoS Genet 10:e1004606, 2014) for AD-related neuropathological traits, including neuritic plaque (NP), neurofibrillary tangle (NFT), and cerebral amyloid angiopathy (CAA). Significant findings were further examined by expression quantitative trait locus and differentially expressed gene analyses in AD vs. control brains using gene expression data. RESULTS Genome-wide significant pleiotropic associations were observed for the joint model of NP and NFT (NP + NFT) with the single-nucleotide polymorphism (SNP) rs34487851 upstream of C2orf40 (alias ECRG4, P = 2.4 × 10-8) and for the joint model of NFT and CAA (NFT + CAA) with the HDAC9 SNP rs79524815 (P = 1.1 × 10-8). Gene-based testing revealed study-wide significant associations (P ≤ 2.0 × 10-6) for the NFT + CAA outcome with adjacent genes TRAPPC12, TRAPPC12-AS1, and ADI1. Risk alleles of proxy SNPs for rs79524815 were associated with significantly lower expression of HDAC9 in the brain (P = 3.0 × 10-3), and HDAC9 was significantly downregulated in subjects with AD compared with control subjects in the prefrontal (P = 7.9 × 10-3) and visual (P = 5.6 × 10-4) cortices. CONCLUSIONS Our findings suggest that pleiotropy analysis is a useful approach to identifying novel genetic associations with complex diseases and their endophenotypes. Functional studies are needed to determine whether ECRG4 or HDAC9 is plausible as a therapeutic target.
Collapse
Affiliation(s)
- Jaeyoon Chung
- Bioinformatics Graduate Program, Boston University, Boston, MA USA
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
| | - Xue Wang
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL USA
| | - Yiyi Ma
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
| | - Gary Beecham
- Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL USA
| | | | | | | | - Todd E. Golde
- Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL USA
| | - Nathan D. Price
- Institute for Systems Biology, University of Washington, Seattle, WA USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL USA
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Jesse Mez
- Department of Neurology, Boston University School of Medicine, Boston, MA USA
| | - Alzheimer’s Disease Genetics Consortium
- Bioinformatics Graduate Program, Boston University, Boston, MA USA
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL USA
- Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL USA
- Department of Pathology, University of Washington, Seattle, WA USA
- Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL USA
- Institute for Systems Biology, University of Washington, Seattle, WA USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
- Department of Neurology, Boston University School of Medicine, Boston, MA USA
- Department of Neurology and Sergievsky Center, Columbia University, New York, NY USA
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA USA
- Neurogenetics and Integrated Genomics, Andover Innovative Medicines Institute, Eisai Inc., Andover, MA USA
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA USA
| | - Richard Mayeux
- Department of Neurology and Sergievsky Center, Columbia University, New York, NY USA
| | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH USA
| | | | - Gerard Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Gyungah R. Jun
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
- Neurogenetics and Integrated Genomics, Andover Innovative Medicines Institute, Eisai Inc., Andover, MA USA
| | - Lindsay A. Farrer
- Bioinformatics Graduate Program, Boston University, Boston, MA USA
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
- Department of Neurology, Boston University School of Medicine, Boston, MA USA
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA USA
| |
Collapse
|
31
|
The unrecognized role of tumor suppressor genes in atrial fibrillation. Gene 2018; 642:26-31. [DOI: 10.1016/j.gene.2017.11.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/25/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
|
32
|
Li L, Li X, Wang W, Gao T, Shi Z. UBE2C is involved in the functions of ECRG4 on esophageal squamous cell carcinoma. Biomed Pharmacother 2018; 98:201-206. [PMID: 29268240 DOI: 10.1016/j.biopha.2017.12.066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/05/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023] Open
|
33
|
Dang X, Zeng X, Coimbra R, Eliceiri BP, Baird A. Counter regulation of ECRG4 gene expression by hypermethylation-dependent inhibition and the Sp1 transcription factor-dependent stimulation of the c2orf40 promoter. Gene 2017; 636:103-111. [PMID: 28870864 DOI: 10.1016/j.gene.2017.08.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022]
Abstract
The human cytokine precursor ECRG4 has been associated with multiple physiological, developmental and pathophysiological processes involving cell proliferation, cell migration, innate immunity, inflammation, cancer progression and metastases. Although down-regulation of ECRG4 gene expression has been largely attributed to hypermethylation of CpG islands in the 5'untranslated region of the ECRG4 promoter, the mechanisms that underlie the dynamics of its regulation have never been systematically described. Here we show that the ECRG4 gene is widely expressed in human tissues and report that its core promoter lies between the -780 to +420 base pairs relative to the ATG start codon of the ECRG4 open reading frame. This sequence, which contains several CpG islands, also includes multiple overlapping Sp1 consensus binding sequences and a putative binding site for NF-kB activation. 5'RACE of mRNA derived from human leukocytes shows that ECRG4 transcription initiates from the guanidine at -11 from the initiation ATG of the ECRG4 open reading frame. While there is no canonical TATA- or CAAT-boxes proximal to this translational initiation site, there is a distal TATA-sequence in the 5'UTR. This region was identified as the sequence targeted by hypermethylation because in vitro methylation of plasmids encoding the ECRG4 promoter abolish promoter activity and the treatment of Jurkat cells (which naturally express ECRG4) with the methylation inhibitor 5-AzaC, increases endogenous ECRG4 expression. Because ChIP assays show that Sp1 binds the ECRG4 promoter, that forced Sp1 expression trans-activates the ECRG4 promoter and Sp1 inhibition with mithramycin inhibits ECRG4 expression, we conclude that the dynamic positive and negative regulatory elements controlling ECRG4 expression include a counter regulation between promoter methylation and Sp1 activation.
Collapse
Affiliation(s)
- Xitong Dang
- Department of Surgery, UC San Diego School of Medicine, University of California San Diego, CA 92139, USA; Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Xiaorong Zeng
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Raul Coimbra
- Department of Surgery, UC San Diego School of Medicine, University of California San Diego, CA 92139, USA
| | - Brian P Eliceiri
- Department of Surgery, UC San Diego School of Medicine, University of California San Diego, CA 92139, USA
| | - Andrew Baird
- Department of Surgery, UC San Diego School of Medicine, University of California San Diego, CA 92139, USA.
| |
Collapse
|
34
|
Deng L, Xiang X, Yang F, Xiao D, Liu K, Chen Z, Zhang R, Feng G. Functional evidence that the self-renewal gene NANOG regulates esophageal squamous cancer development. Biochem Biophys Res Commun 2017; 490:161-168. [PMID: 28601640 DOI: 10.1016/j.bbrc.2017.06.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/06/2017] [Indexed: 10/19/2022]
Abstract
Cancer cell molecular mimicry of stem cells (SC) follows with enhanced proliferative and renewal capacities. In support, numerous mediators of SC self-renewal have been evinced to exhibit oncogenic potential. More and more researches showed that the embryonic stem cell self-renewal genes express in various cancer cells. In this study, we sought to test the tumorigenic functions of NANOG, particularly, in esophageal cancer (EC). Using quantitative RT-PCR and western blotting, we confirmed that EC cells highly express NANOG mRNA and protein. We then constructed a shRNA-mediated plasmid to knockdown of NANOG mRNA. We observed that NANOG deficiency in Eca109 cells decreased clone formation, cell proliferation, and showed G1 arrest. To further investigate the functions and mechanisms of NANOG in Eca109 cells, we detected the changes of multiple signaling molecules when NANOG deficiency. We foud that NANOG deficiency affected multiple genes, particularly, supressed drug-resistance via down-regulated ABCG2 in Eca109 cells, and caused G1 arrest by down-regulated cyclin D1 (CCND1) expression. The present loss-of-function work, establish the integral role for NANOG in Eca109 cell proliferation, drug resistance, and shed light on its mechanisms of action.
Collapse
Affiliation(s)
- Li Deng
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Xiaocong Xiang
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Fei Yang
- Orthopedics, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Dongqin Xiao
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Kang Liu
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Zhu Chen
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Ruolan Zhang
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Gang Feng
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan 637000, China.
| |
Collapse
|
35
|
A Potential Role of Esophageal Cancer Related Gene-4 for Atrial Fibrillation. Sci Rep 2017; 7:2717. [PMID: 28578429 PMCID: PMC5457405 DOI: 10.1038/s41598-017-02902-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 04/20/2017] [Indexed: 11/17/2022] Open
Abstract
Epidemiological studies have shown a strong correlation between tumor and AF. However, the molecular link between tumor and AF remains unknown. ECRG4, a tumor suppressor gene that is expressed in the A-V node and in sporadic ventricular myocytes, inhibits tumorigenesis and monitors tissue homeostasis by functioning as a ‘sentinel’ molecule gauging inflammatory and cell proliferative responses. To explore the potential physiological function of Ecrg4 in heart, we evaluated its distribution in heart, analyzed its expression in patients with persistent AF and in a canine AF model, and dissected the molecular events downstream of Ecrg4. The results showed that the level of Ecrg4 expression is homogenously high in atria and the conduction systems and in sporadic ventricular myocytes. Importantly, the expression of Ecrg4 was significantly decreased in atrial appendages of AF patients than patients with SR. Moreover, in rapid pacing canine AF models, the expression of ECRG4 in atria was significantly decreased compared to that of the controls. Mechanistically, knockdown ECRG4 in atrial myocytes significantly shortened the APDs, inhibited the expression of Gja1, and activated pro-inflammatory cascades and genes involved in cardiac remodeling. These results suggest that Ecrg4 may play a critical role in the pathogenesis of AF.
Collapse
|
36
|
Weinberger P, Ponny SR, Xu H, Bai S, Smallridge R, Copland J, Sharma A. Cell Cycle M-Phase Genes Are Highly Upregulated in Anaplastic Thyroid Carcinoma. Thyroid 2017; 27:236-252. [PMID: 27796151 PMCID: PMC5314727 DOI: 10.1089/thy.2016.0285] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) accounts for only 3% of thyroid cancers, yet strikingly, it accounts for almost 40% of thyroid cancer deaths. Currently, no effective therapies exist. In an effort to identify ATC-specific therapeutic targets, we analyzed global gene expression data from multiple studies to identify ATC-specific dysregulated genes. METHODS The National Center for Biotechnology Information Gene Expression Omnibus database was searched for high-throughput gene expression microarray studies from human ATC tissue along with normal thyroid and/or papillary thyroid cancer (PTC) tissue. Gene expression levels in ATC were compared with normal thyroid or PTC using seven separate comparisons, and an ATC-specific gene set common in all seven comparisons was identified. We investigated these genes for their biological functions and pathways. RESULTS There were three studies meeting inclusion criteria, (including 32 ATC patients, 69 PTC, and 75 normal). There were 259 upregulated genes and 286 downregulated genes in ATC with at least two-fold change in all seven comparisons. Using a five-fold filter, 36 genes were upregulated in ATC, while 40 genes were downregulated. Of the 10 top globally upregulated genes in ATC, 4/10 (MMP1, ANLN, CEP55, and TFPI2) are known to play a role in ATC progression; however, 6/10 genes (TMEM158, CXCL5, E2F7, DLGAP5, MME, and ASPM) had not been specifically implicated in ATC. Similarly, 3/10 (SFTA3, LMO3, and C2orf40) of the most globally downregulated genes were novel in this context, while 7/10 genes (SLC26A7, TG, TSHR, DUOX2, CDH1, PDE8B, and FOXE1) have been previously identified in ATC. We experimentally validated a significant correlation for seven transcription factors (KLF16, SP3, ETV6, FOXC1, SP1, EGFR1, and MAFK) with the ATC-specific genes using microarray analysis of ATC cell lines. Ontology clustering of globally altered genes revealed that "mitotic cell cycle" is highly enriched in the globally upregulated gene set (44% of top upregulated genes, p-value <10-30). CONCLUSIONS By focusing on globally altered genes, we have identified a set of consistently altered biological processes and pathways in ATC. Our data are consistent with an important role for M-phase cell cycle genes in ATC, and may provide direction for future studies to identify novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Paul Weinberger
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Otolaryngology, Medical College of Georgia at Augusta University, Augusta, Georgia
- Augusta University Cancer Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Sithara Raju Ponny
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Hongyan Xu
- Department of Biostatistics and Epidemiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | | | - John Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Biostatistics and Epidemiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
37
|
Chen JY, Wu X, Hong CQ, Chen J, Wei XL, Zhou L, Zhang HX, Huang YT, Peng L. Downregulated ECRG4 is correlated with lymph node metastasis and predicts poor outcome for nasopharyngeal carcinoma patients. Clin Transl Oncol 2017; 19:84-90. [PMID: 27119734 DOI: 10.1007/s12094-016-1507-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/25/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Esophageal cancer-related gene 4 (ECRG4) is a new candidate tumor suppressor gene. In this retrospective study, we evaluated ECRG4 protein expression in patients with nasopharyngeal carcinoma (NPC) under curative treatment and examined its association with pathological features and clinical outcomes as a possible biomarker for diagnosis and prognosis of NPC. METHODS We enrolled 122 patients with a first diagnosis between January 2001 and December 2003. Tumor tissue and control tissue from biopsies underwent immunohistochemical staining for ECRG4. ECRG4 expression was analyzed by clinicopathological variables. After Kaplan-Meier survival analysis, we used Cox proportional hazards regression to estimate the predictive effect of ECRG4 expression on overall survival. RESULTS ECRG4 protein level was lower in NPC than control tissue (P < 0.01). It was inversely related to node status (P < 0.001) and clinical stage (P = 0.027). ECRG4 expression was associated with overall survival, and downregulated ECRG4 expression was an independent prognostic factor of poor survival (hazard ratio = 0.677, 95 % confidence interval 0.463-0.989, P = 0.044). CONCLUSIONS A significant NPC patients showed downregulated ECRG4 expression, which is correlated with lymph node metastasis. The marker could be an independent prognostic factor for NPC patients. The precise function of ECRG4 in the progression of NPC, especially for lymphatic metastasis, deserves further investigation, which would bring a new target for personalized therapy.
Collapse
Affiliation(s)
- J-Y Chen
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - X Wu
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - C-Q Hong
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - J Chen
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - X-L Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - L Zhou
- Department of Gynecologic Surgery, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - H-X Zhang
- Health Care Center, The First Affiliated Hospital of Shantou University Medical College, 3/F, Science and Education Building, 52 Southern Dongxia Road, Shantou, 515041, Guangdong Province, People's Republic of China
| | - Y-T Huang
- Health Care Center, The First Affiliated Hospital of Shantou University Medical College, 3/F, Science and Education Building, 52 Southern Dongxia Road, Shantou, 515041, Guangdong Province, People's Republic of China.
| | - L Peng
- Clinical Laboratory, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, Guangdong, 515041, People's Republic of China.
- Environmental Medicine and Developmental Toxicity, Shantou University Medical College, Shantou, Guangdong, People's Republic of China.
| |
Collapse
|
38
|
Moriguchi T, Kaneumi S, Takeda S, Enomoto K, Mishra SK, Miki T, Koshimizu U, Kitamura H, Kondo T. Ecrg4 contributes to the anti-glioma immunosurveillance through type-I interferon signaling. Oncoimmunology 2016; 5:e1242547. [PMID: 28123880 DOI: 10.1080/2162402x.2016.1242547] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/23/2016] [Accepted: 09/23/2016] [Indexed: 01/14/2023] Open
Abstract
Esophageal cancer-related gene 4 (Ecrg4), a hormone-like peptide, is thought to be a tumor suppressor, however, little is known about the mechanism of how Ecrg4 suppresses tumorigenesis. Here, we show that the ecrg4 null glioma-initiating cell (GIC) line, which was generated from neural stem cells of ecrg4 knockout (KO) mice, effectively formed tumors in the brains of immunocompetent mice, whereas the transplanted ecrg4 wild type-GIC line GIC(+/+) was frequently eliminated. This was caused by host immune system including adaptive T cell responses, since depletion of CD4+, CD8+, or NK cells by specific antibodies in vivo recovered tumorigenicity of GIC(+/+). We demonstrate that Ecrg4 fragments, amino acid residues 71-132 and 133-148, which are produced by the proteolitic cleavage, induced the expression of pro-inflammatory cytokines in microglia in vitro. Moreover, blockades of type-I interferon (IFN) signaling in vivo, either depleting IFN-α/β receptor 1 or using stat1 KO mice, abrogated the Ecrg4-dependent antitumor activity. Together, our findings indicate a major antitumor function of Ecrg4 in enhancing host immunity via type-I IFN signaling, and suggest its potential as a clinical candidate for cancer immunotherapy.
Collapse
Affiliation(s)
- Tetsuo Moriguchi
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University , Sapporo, Hokkaido, Japan
| | - Shun Kaneumi
- Division of Functional immunology, Institute for Genetic Medicine, Hokkaido University , Sapporo, Hokkaido, Japan
| | | | | | - Shyam Kumar Mishra
- Department of Anti-aging and Genomics, Ehime University Proteo-Medicine Research Center , To-on, Ehime, Japan
| | - Tetsuro Miki
- Department of Anti-aging and Genomics, Ehime University Proteo-Medicine Research Center , To-on, Ehime, Japan
| | | | - Hidemitsu Kitamura
- Division of Functional immunology, Institute for Genetic Medicine, Hokkaido University , Sapporo, Hokkaido, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University , Sapporo, Hokkaido, Japan
| |
Collapse
|
39
|
Li L, Li X, Wang W, Gao T, Zhou Y, Lu S. Soluble purified recombinant C2ORF40 protein inhibits tumor cell growth in vivo by decreasing telomerase activity in esophageal squamous cell carcinoma. Oncol Lett 2016; 12:2820-2824. [PMID: 27698864 DOI: 10.3892/ol.2016.4935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/05/2016] [Indexed: 02/02/2023] Open
Abstract
The chromosome 2 open reading frame 40 (C2ORF40) gene is a candidate tumor suppressor gene for a variety of tumors. Previous results by the present authors revealed that the C2ORF40 protein is a secreted protein. However, the exact biological function of secreted C2ORF40 protein in carcinogenesis has not been thoroughly investigated. In the present study, the signal peptide sequence of the C2ORF40 cDNA was initially removed to produce secreted recombinant human C2ORF40 protein (rhC2ORF40). Soluble rhC2ORF40 was successfully expressed and purified, which was evaluated for the first time, to the best of our knowledge, for tumor-suppressing function in vivo in esophageal cancer. The present results revealed that soluble purified rhC2ORF40 was concentrated with a purity of >95%. Furthermore, rhC2ORF40 inhibited esophageal cancer cell growth in vivo in a dose-dependent manner compared with a control group (P<0.05). In addition, the present study demonstrated for the first time that rhC2ORF40 decreased telomerase activity using telomeric repeat amplification protocol-enzyme-linked immunosorbent assay (P<0.05), without affecting the expression levels of telomerase-component RNA (P>0.05), as shown with polymerase chain reaction. Overall, the present results demonstrated that soluble rhC2ORF40 inhibited tumor cell growth in vivo by decreasing telomerase activity in esophageal squamous cell carcinoma. Therefore, soluble rhC2ORF40 with a high purity and biological activity may be a potential biological therapy drug for esophageal cancer.
Collapse
Affiliation(s)
- Linwei Li
- Oncology Department, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, Henan 450003, P.R. China
| | - Xiaoyan Li
- Oncology Department, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, Henan 450003, P.R. China
| | - Wenyu Wang
- Oncology Department, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, Henan 450003, P.R. China
| | - Tianhui Gao
- Oncology Department, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, Henan 450003, P.R. China
| | - Yun Zhou
- Oncology Department, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou, Henan 450003, P.R. China
| | - Shixin Lu
- State Key Laboratory of Molecular Oncology and Department of Etiology and Carcinogenesis, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, P.R. China
| |
Collapse
|
40
|
Ma K, Cao B, Guo M. The detective, prognostic, and predictive value of DNA methylation in human esophageal squamous cell carcinoma. Clin Epigenetics 2016; 8:43. [PMID: 27110300 PMCID: PMC4840959 DOI: 10.1186/s13148-016-0210-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/14/2016] [Indexed: 12/17/2022] Open
Abstract
Esophageal cancer is one of the most common malignancies in the world. Squamous cell carcinoma accounts for approximately 90 % of esophageal cancer cases. Genetic and epigenetic changes have been found to accumulate during the development of various cancers, including esophageal squamous carcinoma (ESCC). Tobacco smoking and alcohol consumption are two major risk factors for ESCC, and both tobacco and alcohol were found to induce methylation changes in ESCC. Growing evidence demonstrates that aberrant epigenetic changes play important roles in the multiple-step processes of carcinogenesis and tumor progression. DNA methylation may occur in the key components of cancer-related signaling pathways. Aberrant DNA methylation affects genes involved in cell cycle, DNA damage repair, Wnt, TGF-β, and NF-κB signaling pathways, including P16, MGMT, SFRP2, DACH1, and ZNF382. Certain genes methylated in precursor lesions of the esophagus demonstrate that DNA methylation may serve as esophageal cancer early detection marker, such as methylation of HIN1, TFPI-2, DACH1, and SOX17. CHFR methylation is a late stage event in ESCC and is a sensitive marker for taxanes in human ESCC. FHIT methylation is associated with poor prognosis in ESCC. Aberrant DNA methylation changes may serve as diagnostic, prognostic, and chemo-sensitive markers. Characterization of the DNA methylome in ESCC will help to better understand its mechanisms and develop improved therapies.
Collapse
Affiliation(s)
- Kai Ma
- />Department of Thoracic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Baoping Cao
- />Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853 China
| | - Mingzhou Guo
- />Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853 China
| |
Collapse
|
41
|
Cai Z, Liang P, Xuan J, Wan J, Guo H. ECRG4 as a novel tumor suppressor gene inhibits colorectal cancer cell growth in vitro and in vivo. Tumour Biol 2016; 37:9111-20. [DOI: 10.1007/s13277-015-4775-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/29/2015] [Indexed: 12/26/2022] Open
|
42
|
You Y, Li H, Qin X, Ran Y, Wang F. Down-regulated ECRG4 expression in breast cancer and its correlation with tumor progression and poor prognosis--A short Report. Cell Oncol (Dordr) 2015; 39:89-95. [PMID: 26631111 DOI: 10.1007/s13402-015-0260-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Recently, we identified the esophageal carcinoma related gene 4 (ECRG4) as a novel candidate tumor suppressor gene and a promising therapeutic target in nasopharyngeal carcinoma (NPC). In addition, we found that reduced ECRG4 expression in NPC was associated with promoter hypermethylation. The aim of the current study was to assess the expression status of the ECRG4 protein in breast cancer and to clarify its clinicopathological significance and potential prognostic implications. METHODS Western blotting was used to examine ECRG4 protein levels in 20 paired breast cancer tissues and adjacent noncancerous tissues. In addition, we performed ECRG4 immunohistochemistry on 113 clinicopathologically well-characterized breast cancer samples and assessed putative associations between its expression and overall patient survival rates. RESULTS We found that ECRG4 protein expression was significantly reduced in the breast cancer tissues compared to the noncancerous tissues. Clinicopathological analyses revealed that loss of ECRG4 protein expression, observed in 41.6 % (47/113) of the primary breast cancer tissues tested, was significantly correlated with lymph node metastasis (P = 0.026), advanced tumor stage (P = 0.042) and unfavorable overall survival (P = 0.004). Additional multivariate analyses revealed that ECRG4 protein expression may serve as an independent prognostic factor for the prediction of patient survival (P = 0.033). CONCLUSION Our data suggest that loss of ECRG4 protein expression may be involved in tumor progression and may serve as a prognostic biomarker for breast cancer.
Collapse
Affiliation(s)
- Yanjie You
- Pathological Examination and Research Center, Luohe Medical College, Luohe, 462002, China
- Department of Pharmacy, Luohe Medical College, Luohe, 462002, China
- Luohe Key Laboratory of Medical Bioengineering, Luohe Medical College, 148 Daxue-Road, Luohe, 462002, China
| | - Haijun Li
- Department of Radiation Oncology, The Second People's Hospital of Neijiang City, Neijiang, 641000, China
| | - Xin Qin
- Medical College, Hubei University of Arts and Science, Xiangyang, 441053, China
| | - Yonggang Ran
- Department of Teaching and Training, Bethune Military Medical NCO Academy of PLA, Shijiazhuang, 050081, China
| | - Fei Wang
- Luohe Key Laboratory of Medical Bioengineering, Luohe Medical College, 148 Daxue-Road, Luohe, 462002, China.
- Bioengineering Laboratory, Luohe Medical College, Luohe, 462002, China.
| |
Collapse
|
43
|
Downregulated ECRG4 is associated with poor prognosis in renal cell cancer and is regulated by promoter DNA methylation. Tumour Biol 2015; 37:1121-9. [PMID: 26276361 DOI: 10.1007/s13277-015-3913-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 08/06/2015] [Indexed: 12/17/2022] Open
Abstract
Esophageal cancer-related gene 4 (ECRG4) has been proposed as a putative tumor suppressor gene in several tumors. However, the role and regulation of ECRG4 in the pathogenesis of human renal cancer remain largely unknown. Our current study revealed that expression of ECRG4 is downregulated in renal cell lines and renal cancer tissues. ECRG4 expression was significantly associated with histological grade of tumors (p < 0.001), primary tumor stage (p = 0.017), and distant metastasis (p = 0.017). Low expression of ECRG4 was an independent prognostic indicator for survival of renal cancer patients. Silencing of ECRG4 expression in renal cell lines was associated with its promoter methylation. Moreover, ectopic expression of ECRG4 markedly inhibited cell proliferation and invasion in renal cancer cell lines. These results indicated that ECRG4 is frequently silenced by the methylation of promoter in renal cell cancers. ECRG4 may be a tumor suppressor in renal cancer and serve as a prognostic marker.
Collapse
|
44
|
Jia J, Dai S, Sun X, Sang Y, Xu Z, Zhang J, Cui X, Song J, Guo X. A preliminary study of the effect of ECRG4 overexpression on the proliferation and apoptosis of human laryngeal cancer cells and the underlying mechanisms. Mol Med Rep 2015; 12:5058-64. [PMID: 26165988 PMCID: PMC4581775 DOI: 10.3892/mmr.2015.4059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 06/03/2015] [Indexed: 12/18/2022] Open
Abstract
Human esophageal cancer-related gene 4 (ECRG4) is a potential tumor suppressor gene isolated from human esophageal epithelial cells. Studies have shown that ECRG4 effectively inhibits the proliferation of tumor cells and induces apoptosis. However, the role of ECRG4 in laryngeal cancer has not yet been clearly defined. In this study, a human laryngeal cancer cell line stably overexpressing ECRG4 was established. The effect of ECRG4 on the proliferation and apoptosis of laryngeal cancer cells and the associated mechanisms were investigated. The Hep-2 human laryngeal carcinoma cell line exhibited a low basal level of ECRG4 expression and was selected for the present study. The eukaryotic expression plasmid pcDNA3.1-ECRG4 was constructed and introduced into Hep-2 cells by transfection reagents. Western blot analysis, reverse transcription-quantitative polymerase chain reaction and immunofluorescence staining confirmed high-level expression of ECRG4. The 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and colony formation assay showed that ECRG4 over-expression suppressed the proliferative capacity of laryngeal cancer cells in vitro. Cell cycle analysis showed that ECRG4 induced cell cycle arrest at the G0/G1 phase. Flow cytometric analysis and Hoechst staining demonstrated that overexpres-sion of ECRG4 significantly induced apoptosis. Western blot analysis confirmed that Bcl-2-associated X protein, cleaved-caspase-3 and cleaved-poly (ADP-ribose) polymerase were upregulated in the apoptotic process, whereas B-cell lymphoma 2 was downregulated. In conclusion, overexpression of ECRG4 inhibited laryngeal cancer cell proliferation and induced cancer cell apoptosis. Therefore, ECRG4 exhibits potential as an effective target in gene therapy for laryngeal cancer.
Collapse
Affiliation(s)
- Jianping Jia
- Department of Otolaryngology‑Head and Neck Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Song Dai
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xinghe Sun
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Yuehong Sang
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Zhenming Xu
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Jie Zhang
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xiaofeng Cui
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Jinhui Song
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xing Guo
- Department of Otolaryngology‑Head and Neck Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
45
|
Li X, Li L, Wang W, Yang Y, Zhou Y, Lu S. Soluble purified recombinant C2ORF40 protein inhibits esophageal cancer cell proliferation by inducing cell cycle G 1 phase block. Oncol Lett 2015; 10:1593-1596. [PMID: 26622716 DOI: 10.3892/ol.2015.3429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 06/11/2015] [Indexed: 02/04/2023] Open
Abstract
Chromosome 2 open reading frame 40 (C2ORF40) plays a significant role in numerous processes, including cell differentiation, senescence, apoptosis, inflammation and neuroendocrine hormone regulation. Moreover, C2ORF40 is a candidate tumor suppressor gene in a variety of tumors, and is closely associated with prognosis. Bioinformatics analysis has indicated that pro-C2ORF40 is a secreted protein with a signal peptide. Secreted C2ORF40 protein (sC2ORF40) exists in cancer cell medium. However, thus far, the exact biological function of sC2ORF40 in carcinogenesis has not been thoroughly researched. In the present study, the signal peptide sequence of the C2ORF40 complementary DNA was initially cut off to produce secreted recombinant human C2ORF40 protein (rhC2ORF40). The soluble rhC2ORF40 was expressed, purified and examined for tumor-suppressing function for the first time. The results revealed that the soluble purified rhC2ORF40 protein was concentrated with a purity of >95%. Furthermore, the rhC2ORF40 inhibited esophageal cancer cell proliferation in vitro (P<0.05) and caused cell cycle G1 phase block, as determined by flow cytometric analysis (P<0.05). Overall, the soluble rhC2ORF40 protein with high purity and biological activity was obtained, which suppressed esophageal cancer cells proliferation by inducing cell cycle G1 phase block in vitro. Therefore, the soluble rhC2ORF40 protein could be potential biological therapy drug for esophageal carcinoma.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Oncology, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Linwei Li
- Department of Oncology, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Wenyu Wang
- Department of Oncology, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yang Yang
- State Key Laboratory of Molecular Oncology and Department of Etiology and Carcinogenesis, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Yun Zhou
- Department of Oncology, Zhengzhou University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Shixin Lu
- State Key Laboratory of Molecular Oncology and Department of Etiology and Carcinogenesis, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| |
Collapse
|
46
|
Porzionato A, Rucinski M, Macchi V, Sarasin G, Malendowicz LK, De Caro R. ECRG4 expression in normal rat tissues: expression study and literature review. Eur J Histochem 2015; 59:2458. [PMID: 26150152 PMCID: PMC4503965 DOI: 10.4081/ejh.2015.2458] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/14/2015] [Accepted: 03/18/2015] [Indexed: 01/19/2023] Open
Abstract
The Esophageal Cancer Related Gene 4 (ECRG4) is a highly conserved tumour suppressor gene encoding various peptides (augurin, CΔ16 augurin, ecilin, argilin, CΔ16 argilin) which can be processed and secreted. In the present work, we examined ECRG4 expression and location in a wide range of rat organs and reviewed the available literature. ECRG4 mRNA was identified in all examined tissues by quantitative PCR (qPCR). ECRG4 immunoreaction was mainly cytoplasmic, and was detected in heart and skeletal muscles, smooth muscle cells showing only weak reactions. In the digestive system, ECRG4 immunostaining was stronger in the esophageal epithelium, bases of gastric glands, hepatocytes and pancreatic acinar epithelium. In the lymphatic system, immunoreactive cells were detectable in the thymus cortex, lymph node medulla and splenic red pulp. In the central and peripheral nervous systems, different neuronal groups showed different reaction intensities. In the endocrine system, ECRG4 immunoreaction was detected in the hypothalamic paraventricular and supraoptic nuclei, hypophysis, thyroid and parathyroid glands, adrenal zona glomerularis and medulla and Leydig cells, as well as in follicular and luteal cells of the ovary. In the literature, ECRG4 has been reported to inhibit cell proliferation and increase apoptosis in various cell types. It is down-regulated, frequently due to hypermethylation, in esophageal, prostate, breast and colon cancers, together with glioma (oncosuppressor function), although it is up-regulated in papillary thyroid cancer (oncogenic role). ECRG4 expression is also higher in non-proliferating cells of the lymphatic system. In conclusion, our identification of ECRG4 in many structures suggests the involvement of ECRG4 in the tumorigenesis of other organs and also the need for further research. In addition, on the basis of the location of ECRG4 in neurons and endocrine cells and the fact that it can be secreted, its role as a neurotransmitter/neuromodulator and endocrine factor must be examined in depth in the future.
Collapse
|
47
|
You Y, Yang W, Qin X, Wang F, Li H, Lin C, Li W, Gu C, Zhang Y, Ran Y. ECRG4 acts as a tumor suppressor and as a determinant of chemotherapy resistance in human nasopharyngeal carcinoma. Cell Oncol (Dordr) 2015; 38:205-14. [PMID: 25707757 DOI: 10.1007/s13402-015-0223-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Human nasopharyngeal carcinoma (NPC) is a malignant type of cancer with an increasing incidence. As yet, however, molecular biomarkers with a strong diagnostic impact and a major therapeutic promise have remained elusive. Here, we identified the esophageal carcinoma related gene 4 (ECRG4) as a novel candidate tumor suppressor gene and a promising therapeutic target for NPC. METHODS RT-PCR, Western blotting, methylation-specific PCR and bisulfite sequencing were performed to assess the expression and methylation status of the ECRG4 gene in primary NPC samples, NPC-derived cell lines and patient-derived peripheral blood samples. The NPC-derived cell line CNE1 was selected for treatment with a methylation inhibitor to restore ECRG4 expression. In addition, cell proliferation, invasion and colony formation assays were performed to assess the inhibitory effects of exogenous ECRG4 expression in CNE1 cells. RESULTS Down-regulated ECRG4 expression was found to occur in 82.5% (33/40) of the primary NPC biopsies tested. This down-regulation was significantly correlated with its tumor-specific promoter methylation status (72.5%, 29/40) and was also observed in the matching peripheral blood samples from the NPC patients (57.5%, 23/40). Pharmacologic demethylation through 5-aza-dC treatment led to gene reactivation in ECRG4 methylated and silenced NPC cell lines. Moreover, exogenous expression of ECRG4 in the CNE1 cell line strongly inhibited its growth and invasive capacities, as well as its enhanced chemosensitivity to cisplatin through autophagy induction. CONCLUSION Our data suggest that methylation-mediated suppression of the ECRG4 gene occurs frequently in NPC and that restoration of its expression may have therapeutic benefits.
Collapse
Affiliation(s)
- Yanjie You
- Department of Pharmacy, Luohe Medical College, 148 Daxue-Road, Luohe, 462002, China,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kao S, Shaterian A, Cauvi DM, Dang X, Chun HB, De Maio A, Costantini TW, Coimbra R, Eliceiri BP, Baird A. Pulmonary preconditioning, injury, and inflammation modulate expression of the candidate tumor suppressor gene ECRG4 in lung. Exp Lung Res 2014; 41:162-72. [PMID: 25513848 DOI: 10.3109/01902148.2014.983282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE The human c2orf40 gene encodes a candidate tumor suppressor called Esophageal Cancer-Related Gene-4 (ECRG4) that is a cytokine-like epigenetically-regulated protein that is characteristically downregulated in cancer, injury, inflammation, and infection. Here, we asked whether ECRG4 gene expression is detectable in lung epithelial cells and if its expression changes with inflammation, infection, and/or protective preconditioning. MATERIALS AND METHODS We used immunoblotting, PCR, and quantitative PCR to measure ECRG4 and either inhalation anesthesia preconditioning, lipopolysaccharide injection, or laparotomy to modulate lung inflammation. RESULTS Immunoblotting establishes the presence of the full-length 14 kDa ECRG4 peptide in mouse lung. Immunohistochemistry localizes ECRG4 to type l alveolar epithelial cells. Basal ECRG4 mRNA is greater than TNF-α, IL-1β, and IL-6 but following inflammatory lung injury, TNF-α, IL-1β, IL-6, and IL-10 are upregulated while ECRG4 gene expression is decreased. Similar findings are observed after an intravenous administration of lipopolysaccharide. In contrast, lung preconditioning with isoflurane anesthesia increases lung ECRG4 gene expression. Over-expression of ECRG4 in human lung epithelial cells in vitro decreases cell proliferation implying that a loss of ECRG4 in vivo would be permissive to cell growth. CONCLUSIONS This study supports the hypothesis that ECRG4 acts as a sentinel growth inhibitor in lung alveolar epithelial cells. Its downregulation by injury, infection, and inflammation and upregulation by preconditioning supports a role for ECRG4 in regulating the alveolar epithelium response to injury and inflammation. By extension, the findings support a functional consequence to its inhibition by promoter hypermethylation (i.e. lung cancer) and suggest potential benefits to its upregulation.
Collapse
Affiliation(s)
- Steven Kao
- Department of Surgery Division of Trauma, Surgical Critical Care, Burn and Acute Care Surgery, School of Medicine, University of California in San Diego, La Jolla, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Podvin S, Dang X, Meads M, Kurabi A, Costantini T, Eliceiri BP, Baird A, Coimbra R. Esophageal cancer-related gene-4 (ECRG4) interactions with the innate immunity receptor complex. Inflamm Res 2014; 64:107-18. [PMID: 25511108 DOI: 10.1007/s00011-014-0789-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/04/2014] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE AND DESIGN The human c2orf40 gene encodes a tumor suppressor gene called esophageal cancer-related gene-4 (ECRG4) with pro- and anti-inflammatory activities that depend on cell surface processing. Here, we investigated its physical and functional association with the innate immunity receptor complex. METHODS Interactions between ECRG4 and the innate immunity receptor complex were assessed by flow cytometry, immunohistochemistry, confocal microscopy, and co-immunoprecipitation. Phage display was used for ligand targeting to cells that overexpress the TLR4-MD2-CD14. RESULTS Immunoprecipitation and immunohistochemical studies demonstrate a physical interaction between ECRG4 and TLR4-MD2-CD14 on human granulocytes. Flow cytometry shows ECRG4 on the cell surface of a subset of CD14(+) and CD16(+) leukocytes. In a cohort of trauma patients, the C-terminal 16 amino acid domain of ECRG4 (ECRG4(133-148)) appears to be processed and shed, presumably at a thrombin-like consensus sequence. Phage targeting this putative ligand shows that this peptide sequence internalizes into cells through the TLR4/CD14/MD2 complex, but modulates inflammation through non-canonical, NFκB signal transduction. CONCLUSIONS ECRG4 is present on the surface of human monocytes and granulocytes. Its interaction with the human innate immunity receptor complex supports a role for cell surface activation of ECRG4 during inflammation and implicates this receptor in its mechanism of action.
Collapse
Affiliation(s)
- Sonia Podvin
- Division of Trauma, Surgical Critical Care, Burns, and Acute Care Surgery, Department of Surgery, University of California, San Diego, San Diego, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Lee J, Dang X, Borboa A, Coimbra R, Baird A, Eliceiri BP. Thrombin-processed Ecrg4 recruits myeloid cells and induces antitumorigenic inflammation. Neuro Oncol 2014; 17:685-96. [PMID: 25378632 DOI: 10.1093/neuonc/nou302] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 09/28/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Extensive infiltration of brain tumors by microglia and macrophages is a hallmark of tumor progression, and yet the overall tumor microenvironment is characterized by an immunosuppressive phenotype. Here we identify esophageal cancer-related gene 4 (Ecrg4) as a novel thrombin-processed monocyte chemoattractant that recruits myeloid cells, promotes their activation, and leads to a blockade of tumor progression. METHODS Both xenograft glioma and syngeneic glioma models were used to measure orthotopic tumor progression and overall survival. Flow cytometry and immunohistochemical analyses were performed to assess myeloid cell localization, recruitment, and activation. RESULTS Ecrg4 promotes monocyte recruitment and activation of microglia in a T-/B-cell-independent mechanism, which leads to a reduction in glioma tumor burden and increased survival. Mutational analysis reveals that the biological activity of Ecrg4 is dependent on a thrombin-processing site at the C-terminus, inducing monocyte invasion in vivo and in vitro. Furthermore, tumor-induced myeloid cell recruitment is impaired in Ecrg4 knockout mice, leading to increased tumor burden and decreased survival. CONCLUSIONS Together, these results identify Ecrg4 as a paracrine factor that activates microglia and is chemotactic for monocytes, with potential as an antitumor therapeutic.
Collapse
Affiliation(s)
- Jisook Lee
- Department of Surgery, University of California San Diego School of Medicine, San Diego, California (J.L., X.D., A.B., R.C., A.B., B.P.E.)
| | - Xitong Dang
- Department of Surgery, University of California San Diego School of Medicine, San Diego, California (J.L., X.D., A.B., R.C., A.B., B.P.E.)
| | - Alexandra Borboa
- Department of Surgery, University of California San Diego School of Medicine, San Diego, California (J.L., X.D., A.B., R.C., A.B., B.P.E.)
| | - Raul Coimbra
- Department of Surgery, University of California San Diego School of Medicine, San Diego, California (J.L., X.D., A.B., R.C., A.B., B.P.E.)
| | - Andrew Baird
- Department of Surgery, University of California San Diego School of Medicine, San Diego, California (J.L., X.D., A.B., R.C., A.B., B.P.E.)
| | - Brian P Eliceiri
- Department of Surgery, University of California San Diego School of Medicine, San Diego, California (J.L., X.D., A.B., R.C., A.B., B.P.E.)
| |
Collapse
|