1
|
Zhang B, Chu W, Li Z, Zhang Y, Zhen Q, Lv B, Liu J, Lu C, Zhao X. Circ-ATIC Serves as a Sponge of miR-326 to Accelerate Esophageal Squamous Cell Carcinoma Progression by Targeting ID1. Biochem Genet 2022; 60:1585-1600. [DOI: 10.1007/s10528-021-10167-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023]
|
2
|
Targeting Post-Translational Regulation of p53 in Colorectal Cancer by Exploiting Vulnerabilities in the p53-MDM2 Axis. Cancers (Basel) 2022; 14:cancers14010219. [PMID: 35008383 PMCID: PMC8750794 DOI: 10.3390/cancers14010219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/23/2021] [Accepted: 12/30/2021] [Indexed: 02/05/2023] Open
Abstract
The role played by the key tumor suppressor gene p53 and the implications of p53 mutations for the development and progression of neoplasia continue to expand. This review focuses on colorectal cancer and the regulators of p53 expression and activity identified over the past decade. These newly recognized regulatory mechanisms include (1) direct regulation of mouse double minute 2 homolog (MDM2), an E3 ubiquitin-protein ligase; (2) modulation of the MDM2-p53 interaction; (3) MDM2-independent p53 degradation; and (4) inhibition of p53 nuclear translocation. We positioned these regulatory mechanisms in the context of p53 missense mutations, which not only evade canonical p53 degradation machinery but also exhibit gain-of-function phenotypes that enhance tumor survival and metastasis. Lastly, we discuss current and potential therapeutic strategies directed against p53 mutant-bearing tumors.
Collapse
|
3
|
Zhu Y, Lam AK, Shum DK, Cui D, Zhang J, Yan DD, Li B, Xu WW, Lee NP, Chan KT, Law S, Tsao SW, Cheung AL. Significance of serglycin and its binding partners in autocrine promotion of metastasis in esophageal cancer. Theranostics 2021; 11:2722-2741. [PMID: 33456569 PMCID: PMC7806492 DOI: 10.7150/thno.49547] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/08/2020] [Indexed: 12/20/2022] Open
Abstract
Rationale: Little is known about the roles of proteoglycans in esophageal cancer. This study aims to investigate the roles and mechanisms of serglycin (SRGN) proteoglycan in promoting metastasis of esophageal squamous cell carcinoma (ESCC). Methods: Reverse phase protein array analysis was used to identify activated signaling pathways in SRGN-overexpressing cells. Chemokine array was used to identify differentially secreted factors from SRGN-overexpressing cells. Binding between SRGN and potential interacting partners was evaluated using proximity ligation assay and co-immunoprecipitation. The glycosaminoglycan (GAG) chains of SRGN were characterized using fluorophore-assisted carbohydrate electrophoresis. Tissue microarray and serum samples were used to determine the correlation of SRGN expression with clinicopathological parameters and patient survival. Results: In vitro and in vivo experiments showed that SRGN promoted invasion and metastasis in ESCC via activating ERK pathway, stabilizing c-Myc and upregulating the secretion of matrix metalloproteinases. SRGN-knockdown suppressed tumorigenic hallmarks. These SRGN-elicited functions were carried out in an autocrine manner by inducing the secretion of midkine (MDK), which was further identified as a novel binding partner of SRGN for the formation of a SRGN/MDK/CD44 complex. In addition, SRGN interacted with MDK and matrix metalloproteinase 2 in ESCC via its GAG chains, which were mainly decorated with chondroitin sulfate comprising of ∆di-4S and ∆di-6S CS. Clinically, high expression of serum SRGN in serum of patients with ESCC was an independent prognostic marker for poor survival. Conclusions: This study provides the first evidence that elevated serum SRGN has prognostic significance in patients with ESCC, and sheds light on the molecular mechanism by which elevated circulating SRGN in cancer patients might promote cancer progression.
Collapse
Affiliation(s)
- Yun Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Alfred K.Y. Lam
- Department of Pathology, Griffith Medical School, Queensland, Gold Coast, QLD, Australia
| | - Daisy K.Y. Shum
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Di Cui
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Jun Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Dong Dong Yan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, 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, Jinan University, Guangzhou, 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, China
| | - Nikki P.Y. Lee
- Department of Surgery, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Kin Tak Chan
- Department of Surgery, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Simon Law
- Department of Surgery, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Sai Wah Tsao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Annie L.M. Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
The HBx and HBc of hepatitis B virus can influence Id1 and Id3 by reducing their transcription and stability. Virus Res 2020; 284:197973. [PMID: 32305567 DOI: 10.1016/j.virusres.2020.197973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 01/28/2023]
Abstract
Hepatitis B virus (HBV) infection is closely related with the occurrence and development of hepatocellular carcinoma (HCC), in which Hepatitis B virus x protein (HBx) and core protein (HBc) play crucial roles. Additionally, inhibitors of differentiation (Id) proteins exhibited significant correlation with liver cancer development. Here, we identified that HBV dramatically inhibited the expression of Id1 and Id3 in both protein and transcriptional levels for the first time, whereas there was little effect of the virus on Id2. Additionally, two HBV coded protein, HBc and HBx, could reduce the expression of Id1 and Id3 distinctly, whereas the other two viral proteins, HBs and HBp were unable to affect Id1 and Id3 proteins. Both the activity inhibitors and activators further confirmed that HBc inhibited the expression of Id1 and Id3 by BMP/Smad signaling pathway. HBx could interact with both Id1 and Id3 at residues 112-136 of HBx protein, and it could inhibit the two Id proteins by accelerating their degradation. This is the first report about HBc and HBx regulating Id1 and Id3, whereas the detailed mechanism associated with above needed further experiments to clarify.
Collapse
|
5
|
Li J, Li Y, Wang B, Ma Y, Chen P. Id-1 promotes migration and invasion of non-small cell lung cancer cells through activating NF-κB signaling pathway. J Biomed Sci 2017; 24:95. [PMID: 29233161 PMCID: PMC5727929 DOI: 10.1186/s12929-017-0400-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
Background Numerous studies have shown that Id-1 (Inhibitor of differentiation 1) is upregulated in several cancers and associated with tumor malignant characters. However, the clinical significance and biological role of Id-1 in non-small cell lung cancer (NSCLC) remains unclear. Methods We used RT-PCR, Western blot and Immunohistochemistry to measure Id-1 expression in NSCLC tissues and matched adjacent noncancerous tissues. The expression pattern of Id-1 in NSCLC tissues was determined by scoring system of immunohistochemical analysis. The Kaplan-Meier method was used to calculate the survival curve, and log-rank test to determine statistical significance. The Id-1 gene was overexpressed or downreuglated with Lentiviral vectors in NSCLC cells. And, the migration ability of NSCLC cells was tested in a Transwell Boyden Chamber. Results We found that Id-1 is generally expressed higher in NSCLC tissues compared with matched adjacent noncancerous tissues. We also found that high Id-1 expression in tumor tissues is significantly correlated with tumor progression and poor survival in NSCLC patients. Furthermore, our experimental data revealed that knockdown of Id-1 significantly suppressed the proliferation, migration and invasion of NSCLC cells, whereas ectopic expression of Id-1 promoted the malignant phenotype of NSCLC cells. Mechanistic study showed that NF-κB signaling pathway contributed to the effects of Id-1 in NSCLC cells. Moreover, blocking the NF-κB pathway significantly inhibited the tumor-promoting actions of Id-1 in NSCLC cells. Conclusions We identified a tumorigenic role of Id-1 in NSCLC and provided a novel therapeutic target for NSCLC patients.
Collapse
Affiliation(s)
- Jie Li
- Department of Chest Surgery, The General Hospital of The People's Liberation Army, No. 28 Fuxing road, Beijing, 100853, China.
| | - Yingjie Li
- Department of Cardio-thoracic Surgery, First Affiliated Hospital, General Hospital of The People's Liberation Army, Beijing, China
| | - Bin Wang
- Department of Chest Surgery, The General Hospital of The People's Liberation Army, No. 28 Fuxing road, Beijing, 100853, China
| | - Yongfu Ma
- Department of Chest Surgery, The General Hospital of The People's Liberation Army, No. 28 Fuxing road, Beijing, 100853, China
| | - Ping Chen
- Department of Cardio-thoracic Surgery, First Affiliated Hospital, General Hospital of The People's Liberation Army, Beijing, China
| |
Collapse
|
6
|
Liu J, Lv L, Gong J, Tan Y, Zhu Y, Dai Y, Pan X, Huen MS, Li B, Tsao SW, Huo J, Cheung AL. Overexpression of F-box only protein 31 predicts poor prognosis and deregulates p38α- and JNK-mediated apoptosis in esophageal squamous cell carcinoma. Int J Cancer 2017; 142:145-155. [PMID: 28905993 DOI: 10.1002/ijc.31040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/21/2017] [Accepted: 08/30/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Jia Liu
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam; Hong Kong SAR People's Republic of China
- Center of Medical Research, The Second Xiangya Hospital of Central South University; Changsha Hunan People's Republic of China
| | - Liang Lv
- Department of Gastroenterology; The Second Xiangya Hospital of Central South University; Changsha Hunan People's Republic of China
| | - Jian Gong
- Department of Gastroenterology; The Second Xiangya Hospital of Central South University; Changsha Hunan People's Republic of China
| | - Yuyong Tan
- Department of Gastroenterology; The Second Xiangya Hospital of Central South University; Changsha Hunan People's Republic of China
| | - Yun Zhu
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam; Hong Kong SAR People's Republic of China
| | - Yinghuan Dai
- Department of Pathology; The Second Xiangya Hospital of Central South University; Changsha Hunan People's Republic of China
| | - Xin Pan
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Tai-Ping Road 27; Beijing 100850 People's Republic of China
| | - Michael S.Y. Huen
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam; Hong Kong SAR People's Republic of China
| | - Bin Li
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam; Hong Kong SAR People's Republic of China
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI); Hong Kong SAR People's Republic of China
| | - Sai Wah Tsao
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam; Hong Kong SAR People's Republic of China
| | - Jirong Huo
- Department of Gastroenterology; The Second Xiangya Hospital of Central South University; Changsha Hunan People's Republic of China
| | - Annie L.M. Cheung
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam; Hong Kong SAR People's Republic of China
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI); Hong Kong SAR People's Republic of China
| |
Collapse
|
7
|
He X, Liu Y, Li K, Yang A, Wang R, Liu S. Sildenafil suppresses the proliferation and enhances the apoptosis of hemangioma endothelial cells. Exp Ther Med 2017; 13:2645-2650. [PMID: 28587325 PMCID: PMC5450598 DOI: 10.3892/etm.2017.4306] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 12/09/2016] [Indexed: 01/16/2023] Open
Abstract
Treatment of infantile hemangiomas (IH) with propranolol was first reported in 2008. Regressions of lymphatic malformations combined with pulmonary hypertension was first reported in 2012 after three children received treatment with oral sildenafil, which serves as an antagonist of phosphodiesterase isoform-5 (PDE-5). A marked expression of endothelial cells in the cytoplasm of IH tissues was obtained in our previous study. Therefore, the present study hypothesized that the antagonist of PDE-5, sildenafil, may lead to the regression of hemangiomas. To assess this hypothesis, the proliferation and apoptosis of specimen-derived hemangioma endothelial cells (HemECs) was determined in vitro by an MTT assay and flow cytometry, respectively, following treatment with sildenafil. The potential mechanisms underlying the mRNA and protein expression levels of inhibitor of differentiation 1 (Id-1) were determined by reverse transcription-quantitative polymerase chain reaction and western blotting. The results demonstrated that 5 µM sildenafil suppressed the proliferation of HemECs and significantly enhanced the rate of apoptosis after 24 h. Additionally, the mRNA and protein expression levels of Id-1 were downregulated following treatment with sildenafil. Therefore, the present study concluded that PDE-5 may be a potential therapeutic target for hemangiomas and Id-1 may serve a vital role in the associated signaling transduction pathways.
Collapse
Affiliation(s)
- Xiaopo He
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Oral and Maxillofacial Surgery, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Yiran Liu
- School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Kai Li
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Oral and Maxillofacial Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Aijun Yang
- Library of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Rui Wang
- School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Oral and Maxillofacial Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shaohua Liu
- Department of Oral and Maxillofacial Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
8
|
Zhou S, Tang X, Tang F. Krüppel-like factor 17, a novel tumor suppressor: its low expression is involved in cancer metastasis. Tumour Biol 2016; 37:1505-1513. [PMID: 26662959 PMCID: PMC4842221 DOI: 10.1007/s13277-015-4588-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/03/2015] [Indexed: 12/18/2022] Open
Abstract
Krüppel-like factor (KLF) family is highly conserved zinc finger transcription factors that regulate cell proliferation, differentiation, apoptosis, and migration. KLF17 is a member of the KLF family. Recent studies have demonstrated that KLF17 low expression and inactivation are caused by microRNA, gene mutation, and loss of heterozygosity in human tumors, which participates in tumor progression. KLF17 low expression increases cancer metastatic viability; its mechanism is that low KLF17 mediates epithelial-mesenchymal transition (EMT) through regulating EMT-related genes expression; the reduced-KLF17 also increases cancer metastasis though upregulating inhibitor of DNA binding 1 (ID1). Additionally, mutant p53 proteins are capable of developing a complex with KLF17, which mediate the depletion of KLF17 inhibiting EMT gene transcription and increases cancer metastasis. KLF17 downregulation also mediates the activation of TGF-β pathway.
Collapse
Affiliation(s)
- Shan Zhou
- Medical Research Center and Clinical Laboratory, Zhuhai People’s Hospital and Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai, 519000 Guangdong China
| | - Xiaowei Tang
- Metallurgical Science and Engineering, Central South University, 21# Lushan South Road, Changsha, 410083 China
| | - Faqing Tang
- Medical Research Center and Clinical Laboratory, Zhuhai People’s Hospital and Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai, 519000 Guangdong China
| |
Collapse
|
9
|
Li J, Li B, Xu WW, Chan KW, Guan XY, Qin YR, Lee NPY, Chan KT, Law S, Tsao SW, Cheung ALM. Role of AMPK signaling in mediating the anticancer effects of silibinin in esophageal squamous cell carcinoma. Expert Opin Ther Targets 2015; 20:7-18. [DOI: 10.1517/14728222.2016.1121236] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
10
|
Yu H, Yue X, Zhao Y, Li X, Wu L, Zhang C, Liu Z, Lin K, Xu-Monette ZY, Young KH, Liu J, Shen Z, Feng Z, Hu W. LIF negatively regulates tumour-suppressor p53 through Stat3/ID1/MDM2 in colorectal cancers. Nat Commun 2014; 5:5218. [PMID: 25323535 PMCID: PMC4203416 DOI: 10.1038/ncomms6218] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 09/10/2014] [Indexed: 12/21/2022] Open
Abstract
Leukemia inhibitory factor (LIF) has been recently identified as a p53 target gene, which mediates the role of p53 in maternal implantation under normal physiological conditions. Here, we report that LIF is a negative regulator of p53; LIF downregulates p53 protein levels and function in human colorectal cancer (CRC) cells. The downregulation of p53 by LIF is mediated by the activation of Stat3, which transcriptionally induces ID1. ID1 upregulates MDM2, a key negative regulator of p53, and promotes p53 protein degradation. LIF is overexpressed in a large percentage of CRCs. LIF overexpression promotes cellular resistance towards chemotherapeutic agents in cultured CRC cells and colorectal xenograft tumors in a largely p53-dependent manner. Overexpression of LIF is associated with a poor prognosis in CRC patients. Taken together, LIF is a novel negative regulator of p53, overexpression of LIF is an important mechanism for the attenuation of p53, which promotes chemoresistance in CRCs.
Collapse
Affiliation(s)
- Haiyang Yu
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Xuetian Yue
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Yuhan Zhao
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Xiaoyan Li
- 1] Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA [2] Department of Breast Surgery, Qilu Hospital, Shandong University, Ji'nan 250012, China
| | - Lihua Wu
- 1] Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA [2] First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - Cen Zhang
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Zhen Liu
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Kevin Lin
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Zijun Y Xu-Monette
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ken H Young
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Juan Liu
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Zhiyuan Shen
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Zhaohui Feng
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Wenwei Hu
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08903, USA
| |
Collapse
|
11
|
Liu J, Han L, Li B, Yang J, Huen MSY, Pan X, Tsao SW, Cheung ALM. F-box only protein 31 (FBXO31) negatively regulates p38 mitogen-activated protein kinase (MAPK) signaling by mediating lysine 48-linked ubiquitination and degradation of mitogen-activated protein kinase kinase 6 (MKK6). J Biol Chem 2014; 289:21508-18. [PMID: 24936062 DOI: 10.1074/jbc.m114.560342] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The p38 MAPK signal transduction pathway plays an important role in inflammatory and stress responses. MAPKK6 (MKK6), a dual specificity protein kinase, is a p38 activator. Activation of the MKK6-p38 pathway is kept in check by multiple layers of regulations, including autoinhibition, dimerization, scaffold proteins, and Lys-63-linked polyubiquitination. However, the mechanisms underlying deactivation of MKK6-p38, which is crucial for maintaining the magnitude and duration of signal transduction, are not well understood. Lys-48-linked ubiquitination, which marks substrates for proteasomal degradation, is an important negative posttranslational regulatory machinery for signal pathway transduction. Here we report that the accumulation of F-box only protein 31 (FBXO31), a component of Skp1 · Cul1 · F-box protein E3 ligase, negatively regulated p38 activation in cancer cells upon genotoxic stresses. Our results show that FBXO31 binds to MKK6 and mediates its Lys-48-linked polyubiquitination and degradation, thereby functioning as a negative regulator of MKK6-p38 signaling and protecting cells from stress-induced cell apoptosis. Taken together, our findings uncover a new mechanism of deactivation of MKK6-p38 and substantiate a novel regulatory role of FBXO31 in stress response.
Collapse
Affiliation(s)
- Jia Liu
- From the Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China and
| | - Liang Han
- From the Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China and
| | - Bin Li
- From the Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China and
| | - Jie Yang
- From the Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China and
| | - Michael S Y Huen
- From the Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China and
| | - Xin Pan
- the Center for Molecular Medicine, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Sai Wah Tsao
- From the Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China and
| | - Annie L M Cheung
- From the Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China and
| |
Collapse
|
12
|
Li B, Tsao SW, Chan KW, Ludwig DL, Novosyadlyy R, Li YY, He QY, Cheung ALM. Id1-induced IGF-II and its autocrine/endocrine promotion of esophageal cancer progression and chemoresistance--implications for IGF-II and IGF-IR-targeted therapy. Clin Cancer Res 2014; 20:2651-62. [PMID: 24599933 DOI: 10.1158/1078-0432.ccr-13-2735] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the autocrine/endocrine role of Id1-induced insulin-like growth factor-II (IGF-II) in esophageal cancer, and evaluate the potential of IGF-II- and IGF-type I receptor (IGF-IR)-targeted therapies. EXPERIMENTAL DESIGN Antibody array-based screening was used to identify differentially secreted growth factors from Id1-overexpressing esophageal cancer cells. In vitro and in vivo assays were performed to confirm the induction of IGF-II by Id1, and to study the autocrine and endocrine effects of IGF-II in promoting esophageal cancer progression. Human esophageal cancer tissue microarray was analyzed for overexpression of IGF-II and its correlation with that of Id1 and phosphorylated AKT (p-AKT). The efficacy of intratumorally injected IGF-II antibody and intraperitoneally injected cixutumumab (fully human monoclonal IGF-IR antibody) was evaluated using in vivo tumor xenograft and experimental metastasis models. RESULTS Id1 overexpression induced IGF-II secretion, which promoted cancer cell proliferation, survival, and invasion by activating AKT in an autocrine manner. Overexpression of IGF-II was found in 21 of 35 (60%) esophageal cancer tissues and was associated with upregulation of Id1 and p-AKT. IGF-II secreted by Id1-overexpressing esophageal cancer xenograft could instigate the growth of distant esophageal tumors, as well as promote metastasis of circulating cancer cells. Targeting IGF-II and IGF-IR had significant suppressive effects on tumor growth and metastasis in mice. Cixutumumab treatment enhanced the chemosensitivity of tumor xenografts to fluorouracil and cisplatin. CONCLUSIONS The Id1-IGF-II-IGF-IR-AKT signaling cascade plays an important role in esophageal cancer progression. Blockade of IGF-II/IGF-IR signaling has therapeutic potential in the management of esophageal cancer.
Collapse
Affiliation(s)
- Bin Li
- Authors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New YorkAuthors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New York
| | - Sai Wah Tsao
- Authors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New YorkAuthors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New York
| | - Kwok Wah Chan
- Authors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New YorkAuthors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New York
| | - Dale L Ludwig
- Authors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New York
| | - Ruslan Novosyadlyy
- Authors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New York
| | - Yuk Yin Li
- Authors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New York
| | - Qing Yu He
- Authors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New York
| | - Annie L M Cheung
- Authors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New YorkAuthors' Affiliations: Department of Anatomy, Centre for Cancer Research; Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR; Institute of Life and Health Engineering, Jinan University, Guangzhou, China; and ImClone Systems Corporation, a wholly owned subsidiary of Eli Lilly & Co, New York, New York
| |
Collapse
|
13
|
Lasorella A, Benezra R, Iavarone A. The ID proteins: master regulators of cancer stem cells and tumour aggressiveness. Nat Rev Cancer 2014; 14:77-91. [PMID: 24442143 DOI: 10.1038/nrc3638] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inhibitor of DNA binding (ID) proteins are transcriptional regulators that control the timing of cell fate determination and differentiation in stem and progenitor cells during normal development and adult life. ID genes are frequently deregulated in many types of human neoplasms, and they endow cancer cells with biological features that are hijacked from normal stem cells. The ability of ID proteins to function as central 'hubs' for the coordination of multiple cancer hallmarks has established these transcriptional regulators as therapeutic targets and biomarkers in specific types of human tumours.
Collapse
Affiliation(s)
- Anna Lasorella
- Institute for Cancer Genetics, Department of Pathology and Pediatrics, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, 10032 New York, USA
| | - Robert Benezra
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 241, New York, 10065 New York, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Department of Pathology and Neurology, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, 10032 New York, USA
| |
Collapse
|
14
|
Lewis TC, Prywes R. Serum regulation of Id1 expression by a BMP pathway and BMP responsive element. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1147-59. [PMID: 23948603 DOI: 10.1016/j.bbagrm.2013.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 02/01/2023]
Abstract
Immediate early genes (IEGs) are expressed upon re-entry of quiescent cells into the cell cycle following serum stimulation. These genes are involved in growth control and differentiation and hence their expression is tightly controlled. Many IEGs are regulated through Serum Response Elements (SREs) in their promoters, which bind Serum Response Factor (SRF). However, many other IEGs do not have SREs in their promoters and their serum regulation is poorly understood. We have identified SRF-independent IEGs in SRF-depleted fibroblasts. One of these, Id1, was examined more closely. We mapped a serum responsive element in the Id1 promoter and find that it is identical to a BMP responsive element (BRE). The Id1 BRE is necessary and sufficient for the serum regulation of Id1. Inhibition of the BMP pathway by siRNA depletion of Smad 4, treatment with the BMP antagonist noggin, or the BMP receptor inhibitor dorsomorphin blocked serum induction of Id1. Further, BMP2 is sufficient to induce Id1 expression. Given reports that SRC inhibitors can block Id1 expression, we tested the SRC inhibitor, AZD0530, and found that it inhibits the serum activation of Id1. Surprisingly, this inhibition is independent of SRC or its family members. Rather, we show that AZD0530 directly inhibits the BMP type I receptors. Serum induction of the Id1 related gene Id3 also required the BMP pathway. Given these and other findings we conclude that the Id family of IEGs is regulated by BMPs in serum through similar BREs. This represents a second pathway for serum regulation of IEGs.
Collapse
Affiliation(s)
- Thera C Lewis
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA.
| | | |
Collapse
|
15
|
Liu H, Jia D, Li A, Chau J, He D, Ruan X, Liu F, Li J, He L, Li B. p53 regulates neural stem cell proliferation and differentiation via BMP-Smad1 signaling and Id1. Stem Cells Dev 2013. [PMID: 23199293 DOI: 10.1089/scd.2012.0370] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neural stem cells (NSCs) play essential roles in nervous system development and postnatal neuroregeneration and their deregulation underlies the development of neurodegenerative disorders. Yet how NSC proliferation and differentiation are controlled is not fully understood. Here we present evidence that tumor suppressor p53 regulates NSC proliferation and differentiation via the bone morphogenetic proteins (BMP)-Smad1 pathway and its target gene inhibitor of DNA binding 1 (Id1). p53 deficiency led to increased neurogenesis in vivo, and biased neuronal differentiation and augmented NSC proliferation of ex vivo NSCs. This is accompanied by elevated Smad1 expression/activation in the brain and NSC, which contributes to accelerated neuronal differentiation of p53(-/-) NSCs. p53 deficiency also leads to upregulation of Id1, whose expression is repressed by p53 in BMP-Smad1-dependent and -independent manners. Elevated Id1 expression contributes to augmented proliferation and, unexpectedly, accelerated neuronal differentiation of p53(-/-) NSCs as well. This study reveals a molecular mechanism by which tumor suppressor p53 controls NSC proliferation and differentiation and establishes a connection between p53 and Id1.
Collapse
Affiliation(s)
- Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
LIF maintains progenitor phenotype of endothelial progenitor cells via Krüppel-like factor 4. Microvasc Res 2012; 84:270-7. [PMID: 22835519 DOI: 10.1016/j.mvr.2012.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 06/22/2012] [Accepted: 07/16/2012] [Indexed: 11/24/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) participate in post-natal vasculogenesis. Maintaining the preliminary progenitor phenotype and good proliferation capacity of EPCs is key to their use in treating cardiovascular ischemic diseases. However, transcriptional regulation in EPCs remains largely unknown. We investigated the effect of leukemia inhibitory factor (LIF) combined with vascular endothelial growth factor (VEGF) on EPCs and the potential roles of Krüppel-like transcription factors (KLFs). METHODS AND RESULTS Co-treatment with LIF and VEGF (100 ng/ml each) (V+L) could increase EPC colony-forming units and CD34 expression, which reflects the EPC progenitor phenotype and alleviated differentiation of EPCs. The effect was associated with Akt activation and increased expression of KLF4. Upregulation of KLF4 induced by V+L could be inhibited by transfection with dominant-negative Akt adenovirus. Furthermore, overexpression of KLF4 in EPCs enhanced the expression of CD34 and alleviated cell differentiation but did not increase the phosphorylation of Akt. CONCLUSIONS LIF combined with VEGF can maintain the preliminary, progenitor phenotype of EPCs and alleviate cell differentiation by upregulating KLF4, which may provide new insights into transcriptional regulation in EPCs.
Collapse
|
17
|
Peng X, Wang Y, Kolli S, Deng J, Li L, Wang Z, Raj JU, Gou D. Physical and functional interaction between the ID1 and p65 for activation of NF-κB. Am J Physiol Cell Physiol 2012; 303:C267-77. [PMID: 22592405 DOI: 10.1152/ajpcell.00365.2011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Inhibitor of differentiation or DNA binding-1 (ID1) is an important helix-loop-helix (HLH) transcription factor involved in diverse biological functions including cell differentiation, proliferation, apoptosis, and senescence. Recently, it was reported that ID1 can activate the NF-κB signaling pathway in a variety of cancer cells and a T cell line, but the mechanisms involved in ID1-mediated transactivation of NF-κB are not clear. In this study, we demonstrate by both in vitro pull-down assays and a cell-based in vivo two-hybrid system that ID1-mediated NF-κB activation is due to its physical interaction with p65. We have identified that the transcriptional activation domain (TAD) in p65 and the HLH domain in ID1 are vital for their interaction. Interestingly, a single site mutation (Leu76) in the HLH domain of ID1 protein drastically decreased its ability to bind with p65. Using a dual-luciferase assay, we demonstrated that the interaction between ID1 and p65 modulates activation of the NF-κB signaling pathway in vivo. In addition, we demonstrated that, by affecting the nuclear translocation of p65, ID1 is essential in regulating TNF-α-induced p65 recruitment to its downstream target, the cellular inhibitor of apoptosis protein 2 (cIAP2) promoter.
Collapse
Affiliation(s)
- Xiao Peng
- College of Life Sciences, Shenzhen University, Shenzhen, China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
An essential role for the Id1/PI3K/Akt/NFkB/survivin signalling pathway in promoting the proliferation of endothelial progenitor cells in vitro. Mol Cell Biochem 2011; 363:135-45. [PMID: 22139302 PMCID: PMC3289789 DOI: 10.1007/s11010-011-1166-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 11/23/2011] [Indexed: 12/24/2022]
Abstract
The enhancement of re-endothelialisation is a critical therapeutic option for repairing injured blood vessels. Endothelial progenitor cells (EPCs) are the major source of cells that participate in endothelium repair and contribute to re-endothelialisation by reducing neointima formation after vascular injury. The over-expression of the inhibitor of differentiation or DNA binding 1 (Id1) significantly improved EPC proliferation. This study aimed to investigate the effects of Id1 on the phosphatidylinositol-3-kinase (PI3K)/Akt/nuclear factor kappa B (NFκB)/survivin signalling pathway and its significance in promoting EPC proliferation in vitro. Spleen-derived EPCs were cultured as previously described. Id1 was presented at low levels in EPCs, and was rapidly up-regulated by stimulation with vascular endothelial growth factor. We demonstrated that transient transfection of Id1 into EPCs activated the PI3K/Akt/NFκB/survivin signalling pathway and promoted EPC proliferation. The proliferation of EPCs was extensively inhibited by silencing of endogenous Id1, and knockdown of Id1 expression led to suppression of PI3K/Akt/NFκB/survivin signalling pathway in EPCs. In addition, blockade by the PI3K-specific inhibitor LY294002, Akt inhibitor, the NFκB inhibitor BAY 11-7082, the survivin inhibitor Curcumin, or the survivin inhibitor YM155 reduced the effects of Id1 transfection. These results suggest that the Id1/PI3K/Akt/NFκB/survivin signalling pathway plays a critical role in EPC proliferation. The Id1/PI3K/Akt/NFκB/survivin signalling pathway may represent a novel therapeutic target in the prevention of restenosis after vascular injury.
Collapse
|
19
|
Cheung PY, Yip YL, Tsao SW, Ching YP, Cheung ALM. Id-1 induces cell invasiveness in immortalized epithelial cells by regulating cadherin switching and Rho GTPases. J Cell Biochem 2011; 112:157-68. [PMID: 21053361 DOI: 10.1002/jcb.22911] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Epithelial-mesenchymal transition (EMT), characterized by cadherin switching, contributes to cancer metastasis. Our recent study showed that Id-1 (inhibitor of differentiation-1) promotes metastasis in esophageal cancer cells, but whether the invasive and metastatic dynamics can be induced early in the carcinogenesis process is still unclear. Immortalization is regarded as the initial stage in the malignant transformation of normal cells. In this study, we investigated the role and mechanisms of Id-1 in inducing EMT and cell invasiveness in immortalized esophageal epithelial cells. We found that immortalized epithelial cells expressed higher endogenous levels of Id-1 compared with normal cells. Ectopic Id-1 expression inhibited the differentiation of immortalized esophageal epithelial cells and promoted cadherin switching, which was accompanied by increased adhesiveness to extracellular matrix, cell motility, migratory potential and matrix metalloproteinase-dependent invasiveness. GTPase activity assays showed that over-expression or short-hairpin RNA knockdown of Id-1 led to corresponding changes in Rac1 activity, whereas RhoA activity was significantly decreased with Id-1 depletion. Inhibitors targeting Rac1, RhoA, and Rho kinase suppressed the invasiveness of Id-1-expressing NE2-hTERT cells. Knockdown of N-cadherin in Id-1-over-expressing cells inhibited cell invasiveness and down-regulated RhoA activity. These data suggest that the Id-1-induced invasive potential may be regulated through the N-cadherin-RhoA axis and Rac1 activation.
Collapse
Affiliation(s)
- P Y Cheung
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
20
|
Cheng YJ, Tsai JW, Hsieh KC, Yang YC, Chen YJ, Huang MS, Yuan SS. Id1 promotes lung cancer cell proliferation and tumor growth through Akt-related pathway. Cancer Lett 2011; 307:191-9. [PMID: 21536374 DOI: 10.1016/j.canlet.2011.04.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 04/06/2011] [Accepted: 04/07/2011] [Indexed: 11/24/2022]
Abstract
Overexpression of Id family proteins inhibits cell differentiation and enhances cell proliferation and invasiveness. Although Id1 is the Id family member mostly linked to tumorigenesis, its role in lung cancer is unclear. An elevated Id1 expression was observed in lung cancer cell lines as well as lung cancer tissues. Id1 overexpression increased cell proliferation while Id1 knockdown decreased cell proliferation, mostly through Akt-related pathway. Nude mice study further confirmed an increased tumor growth in Id1-overexpressing cells and a decreased tumor growth in Id1-knockdowned cells. In conclusion, inactivation of Id1 may provide a novel strategy for treatment of lung cancer patients.
Collapse
Affiliation(s)
- Yu-Jen Cheng
- Division of Thoracic Surgery, Department of Surgery, E-DA Hospital, No. 1 E-DA Road, Jiau-Shu, Kaohsiung 824, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
21
|
Gu Y, Fan S, Xiong Y, Peng B, Zheng G, Yu Y, Ouyang Y, He Z. Cloning and functional characterization of TCRP1, a novel gene mediating resistance to cisplatin in an oral squamous cell carcinoma cell line. FEBS Lett 2011; 585:881-7. [PMID: 21334329 DOI: 10.1016/j.febslet.2010.12.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 12/26/2010] [Accepted: 12/29/2010] [Indexed: 11/18/2022]
Abstract
To explore the mechanisms of chemotherapy resistance, we previously established a multi-drug resistant cell line, Tca8113/Pingyangmycin (Tca8113/PYM) and identified differential expression in known genes and ESTs using microarray analysis. From among those ESTs we have now identified a novel gene producing an mRNA of 1834 nucleotides translated into a protein having 235 amino acids. This gene was denominated as tongue cancer resistance-associated protein 1 gene (TCRP1, accession number: EF363480). We further determined its functional characteristics. The results demonstrate that TCRP1 mediates a specific resistance to cisplatin in Tca8113 cells by reducing the cisplatin-induced apoptosis. This suggests that TCRP1 might be a novel molecular target to develop agents to reverse cisplatin-induced chemoresistance.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cisplatin/pharmacology
- Cloning, Molecular
- Comet Assay
- Cytoplasm/metabolism
- DNA Damage
- DNA, Neoplasm/genetics
- Drug Resistance, Multiple/genetics
- Drug Resistance, Neoplasm/genetics
- Flow Cytometry
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Microscopy, Fluorescence
- Proteins/genetics
- Proteins/metabolism
- RNA Interference
- Reverse Transcriptase Polymerase Chain Reaction
- Tongue Neoplasms/genetics
- Tongue Neoplasms/metabolism
- Tongue Neoplasms/pathology
Collapse
Affiliation(s)
- Yixue Gu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha 410078, Hunan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Cheung PY, Deng W, Man C, Tse WW, Srivastava G, Law S, Tsao SW, Cheung ALM. Genetic alterations in a telomerase-immortalized human esophageal epithelial cell line: implications for carcinogenesis. Cancer Lett 2010; 293:41-51. [PMID: 20092939 DOI: 10.1016/j.canlet.2009.12.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2009] [Revised: 12/18/2009] [Accepted: 12/23/2009] [Indexed: 02/04/2023]
Abstract
Ectopic expression of viral oncoproteins disrupts cellular functions and limits the value of many existing immortalization models as models for carcinogenesis, especially for cancers without definitive viral etiology. Our newly established telomerase-immortalized human esophageal epithelial cell line, NE2-hTERT, retained nearly-diploid and non-tumorigenic characteristics, but exhibited genetic and genomic alterations commonly found in esophageal cancer, including progressive loss of the p16(INK4a) alleles, upregulation of anti-apoptotic proteins, epithelial-mesenchymal transition, whole-chromosome 7 gain and duplicated 5q arm. Our data also revealed a novel positive regulation of p16(INK4a) on cyclin D1. These findings probably represent early crucial events and mechanisms in esophageal carcinogenesis.
Collapse
Affiliation(s)
- Pak Yan Cheung
- Cancer Biology Group, Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Li B, Tsao SW, Li YY, Wang X, Ling MT, Wong YC, He QY, Cheung ALM. Id-1 promotes tumorigenicity and metastasis of human esophageal cancer cells through activation of PI3K/AKT signaling pathway. Int J Cancer 2009; 125:2576-85. [PMID: 19551863 DOI: 10.1002/ijc.24675] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Id-1 (inhibitor of differentiation or DNA binding) is a helix-loop-helix protein that is overexpressed in many types of cancer including esophageal squamous cell carcinoma (ESCC). We previously reported that ectopic Id-1 expression activates the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) signaling pathway in human esophageal cancer cells. In this study, we confirmed a positive correlation between Id-1 and phospho-AKT (Ser473) expressions in ESCC cell lines, as well as in ESCC on a tissue microarray. To investigate the significance of Id-1 in esophageal cancer progression, ESCC cells with stable ectopic Id-1 expression were inoculated subcutaneously into the flank of nude mice and were found to form larger tumors that showed elevated Ki-67 proliferation index and increased angiogenesis, as well as reduced apoptosis, compared with control cells expressing the empty vector.The Id-1-overexpressing cells also exhibited enhanced metastatic potential in the experimental metastasis assay. Treatment with the PI3K inhibitor LY294002 attenuated the tumor promotion effects of Id-1, indicating that the effects were mediated by the PI3K/AKT signaling pathway. In addition, our in vitro experiments showed that ectopic Id-1 expression altered the expression levels of markers associated with epithelial-mesenchymal transition and enhanced the migration ability of esophageal cancer cells. The Id-1-overexpressing ESCC cells also exhibited increased invasive potential, which was in part due to PI3K/AKT-dependent modulation of matrix metalloproteinase-9 expression. In conclusion, our results provide the first evidence that Id-1 promotes tumorigenicity and metastasis of human esophageal cancer in vivo and that the PI3K inhibitor LY294002 can attenuate these effects.
Collapse
Affiliation(s)
- Bin Li
- Cancer Biology Group, Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Song H, Guo B, Zhang J, Song C. Transforming Growth Factor-β Suppressed Id-1 Expression in a smad3-Dependent Manner in LoVo Cells. Anat Rec (Hoboken) 2009; 293:42-7. [DOI: 10.1002/ar.21012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Inhibitor of differentiation 1 (Id1) expression attenuates the degree of TiO2-induced cytotoxicity in H1299 non-small cell lung cancer cells. Toxicol Lett 2009; 189:191-9. [DOI: 10.1016/j.toxlet.2009.05.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 05/20/2009] [Accepted: 05/25/2009] [Indexed: 01/13/2023]
|
26
|
Li B, Li YY, Tsao SW, Cheung ALM. Targeting NF-kappaB signaling pathway suppresses tumor growth, angiogenesis, and metastasis of human esophageal cancer. Mol Cancer Ther 2009; 8:2635-44. [PMID: 19723887 DOI: 10.1158/1535-7163.mct-09-0162] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Esophageal cancer is the eighth most common malignancy, and one of the leading causes of cancer-related deaths worldwide. The overall 5-year survival rate of patients with esophageal cancer remains low at 10% to 40% due to late diagnosis, metastasis, and resistance of the tumor to radiotherapy and chemotherapy. NF-kappaB is involved in the regulation of cell growth, survival, and motility, but little is known about the role of this signaling pathway in the tumorigenesis of human esophageal squamous cell carcinoma (ESCC), the most common form of esophageal cancer. This study aims to explore the functions of NF-kappaB in human ESCC progression and to determine whether targeting the NF-kappaB signaling pathway might be of therapeutic value against ESCC. Our results from human ESCC cell lines and ESCC tissue indicated that NF-kappaB is constitutively active in ESCC. Exposure of ESCC cells to two NF-kappaB inhibitors, Bay11-7082 and sulfasalazine, not only reduced cancer cell proliferation, but also induced apoptosis and enhanced sensitivity to chemotherapeutic drugs, 5-fluorouracil, and cisplatin. In addition, Bay11-7082 and sulfasalazine suppressed the migration and invasive potential of ESCC cells. More importantly, the results from tumor xenograft and experimental metastasis models showed that Bay11-7082 had significant antitumor effects on ESCC xenografts in nude mice by promoting apoptosis, and inhibiting proliferation and angiogenesis, as well as reduced the metastasis of ESCC cells to the lungs without significant toxic effects. In summary, our data suggest that NF-kappaB inhibitors may be potentially useful as therapeutic agents for patients with esophageal cancer.
Collapse
Affiliation(s)
- Bin Li
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | | | | |
Collapse
|
27
|
Anticancer effect of celecoxib via COX-2 dependent and independent mechanisms in human gastric cancers cells. Dig Dis Sci 2009; 54:1418-24. [PMID: 18923901 DOI: 10.1007/s10620-008-0510-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Accepted: 08/22/2008] [Indexed: 12/15/2022]
Abstract
Cyclooxygenase-2 (COX-2) inhibitors cause growth inhibition of human gastric carcinoma cells, but it remains unclear whether this is both COX-2 dependent and independent. The related mechanisms remain to be determined. Both low COX-2 expressing gastric carcinoma and high COX-2 expressing gastric carcinoma cells were used to study the effect and mechanisms of celecoxib on gastric carcinoma cell growth. Celecoxib resulted in comparable growth inhibition in AGS cells with stable transfections of small interfering RNA (siRNA) against COX-2 (SAC) and negative control vector (NC) cells. Simultaneously, celecoxib resulted in significant reduction of Bcl-2 and significant increase of p21(WAF1) and p27(KIP1) in SAC and NC cells. The present study shows that celecoxib causes growth inhibition of gastric carcinoma cells by decreasing Bcl-2 of cyclooxygenase-2-dependent pathway, and by increasing p21(WAF1) and p27(KIP1) of cyclooxygenase-2-independent pathway. These data extend our knowledge on the effect and mechanisms of celecoxib-induced inhibition of gastric carcinoma cell growth.
Collapse
|
28
|
Chen L, Qiu J, Yang C, Yang X, Chen X, Jiang J, Luo X. Identification of a novel estrogen receptor β1 binding partner, inhibitor of differentiation-1, and role of ERβ1 in human breast cancer cells. Cancer Lett 2009; 278:210-219. [DOI: 10.1016/j.canlet.2009.01.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 12/26/2008] [Accepted: 01/06/2009] [Indexed: 02/07/2023]
|
29
|
Liu L, Lai CQ, Nie L, Ordovas J, Band M, Moser L, Meydani M. The modulation of endothelial cell gene expression by green tea polyphenol-EGCG. Mol Nutr Food Res 2009; 52:1182-92. [PMID: 18465779 DOI: 10.1002/mnfr.200700499] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Human and animal studies have shown that green tea consumption is associated with a reduced risk of some cancers. This has been attributed to its polyphenol components, in particular (-)-epigallocatechin gallate (EGCG). In addition to be a cancer chemopreventive agent, EGCG inhibits angiogenesis, thus reducing tumor growth and metastasis. We tested EGCG modulation on the gene expression profile of endothelial cells stimulated by VEGF using Affymetrix microarrays. A total of 421 genes were up-regulated and 72 genes were down-regulated at the false discovery rate of 5% by VEGF, EGCG, and EGCG pretreatment followed by VEGF stimulation. The changes in the expression of several pivotal genes were validated by real-time PCR. Furthermore, we have identified two signaling pathways (Wnt and Id) involved in cell proliferation were inhibited by EGCG treatment, suggesting the negative regulation of EGCG on cell proliferation. Our results also indicate that the antiangiogenesis effect of EGCG is partially mediated through its broad inhibition on endothelial cell proliferation. Our data further support earlier observations that the anticancer effect of EGCG is mediated through changes in the expression of genes that are associated with cell proliferation.
Collapse
Affiliation(s)
- Liping Liu
- Vascular Biology Laboratory, JM USDA Human Nutrition Center on Aging at Tufts University, Boston, MA 02111, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Kim H, Chung H, Kim HJ, Lee JY, Oh MY, Kim Y, Kong G. Id-1 regulates Bcl-2 and Bax expression through p53 and NF-κB in MCF-7 breast cancer cells. Breast Cancer Res Treat 2007; 112:287-96. [DOI: 10.1007/s10549-007-9871-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Accepted: 12/14/2007] [Indexed: 10/22/2022]
|
31
|
Hua H, Sarvetnick N. ID2 promotes the expansion and survival of growth-arrested pancreatic beta cells. Endocrine 2007; 32:329-37. [PMID: 18322822 DOI: 10.1007/s12020-008-9039-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 01/11/2008] [Accepted: 01/14/2008] [Indexed: 10/22/2022]
Abstract
Inhibitors of DNA binding proteins (Ids) are implicated in the control of proliferation and differentiation. Herein, we tested the hypothesis that Id2 could stimulate proliferation and survival in differentiated pancreatic beta cells. We showed that Id2-enhanced proliferation of a growth-arrested pancreatic beta cell line (BTC-tet). This was mediated by the Rb pathway, as shown by an E2F1-driven reporter assay and Western immunoblot of phosphorylated Rb protein. Id2 also induced expression of Bcl-2, accompanied by a significant reduction of critical mediators of cytokine stimulation, including p38 MAPK and NFkappaB, as well as apoptosis markers, caspase-3 and Annexin-V. Overall, our data suggest that Id2 enhances proliferation and survival of growth-arrested BTC-tet cells.
Collapse
Affiliation(s)
- Hong Hua
- Department of Immunology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., IMM-23, La Jolla, CA 92037, USA
| | | |
Collapse
|
32
|
Abstract
Id protein family consists of four members namely Id-1 to Id-4. Different from other basic helix-loop-helix transcription factors, they lack the DNA binding domain. Id proteins have been shown to be dysregulated in many different cancer types and their prognostic value has also been demonstrated. Recently, Id-1 has been shown to be upregulated in oesophageal squamous cell carcinoma (ESCC). However, the prognostic implications of Id proteins in ESCC have not been reported. We examined the expression of the Id proteins in ESCC cell lines and clinical ESCC specimens and found that Id protein expressions were dysregulated in both the ESCC cell lines and specimens. By correlating the expression levels of Id proteins and the clinicopathological data of our patient cohort, we found that M1 stage tumours had significantly higher nuclear Id-1 expression (P=0.012) while high nuclear Id-1 expression could predict development of distant metastasis within 1 year of oesophagectomy (P=0.005). In addition, high levels of Id-2 expression in both cytoplasmic and nuclear regions predicted longer patient survival (P=0.041). Multivariate analysis showed that high-level expression of Id-2 in both cytoplasmic and nuclear regions and lower level of nuclear Id-1 expression were independent favourable predictors of survival in our ESCC patients. Our results suggest that Id-1 may promote distant metastasis in ESCC, and both Id-1 and Id-2 may be used for prognostication for ESCC patients.
Collapse
|
33
|
Zhang H, Lawson WE, Polosukhin VV, Pozzi A, Blackwell TS, Litingtung Y, Chiang C. Inhibitor of differentiation 1 promotes endothelial survival in a bleomycin model of lung injury in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1113-26. [PMID: 17717145 PMCID: PMC1988863 DOI: 10.2353/ajpath.2007.070226] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The Id family of genes encodes negative regulators of basic helix-loop-helix transcription factors and has been implicated in diverse cellular processes such as proliferation, apoptosis, differentiation, and migration. However, the specific role of Id1 in lung injury has not been investigated. Bleomycin has been widely used to generate animal models of acute lung injury and fibrogenesis. In this study we found that, on bleomycin challenge, Id1 expression was significantly up-regulated in the lungs, predominantly in endothelial cells, as revealed by double immunolabeling and quantitative flow cytometric analysis. Mice with Id1 loss-of-function (Id1(-/-)) displayed increased vascular permeability and endothelial apoptosis in the lungs after bleomycin-induced injury. Cultured Id1(-/-) lung microvascular endothelial cells also showed decreased survival when exposed to bleomycin. We detected a decrease in the level of Bcl-2, a primary anti-apoptotic protein, in Id1(-/-) endothelial cells, suggesting that down-regulated Bcl-2 may promote endothelial apoptosis in the lung. Therefore, we propose that Id1 plays a crucial role in promoting endothelial survival in the adult lung on injury. In addition, bleomycin-exposed Id1(-/-) mice showed increased lung collagen accumulation and fibrogenesis, suggesting that Id1 up-regulation in the lung may play a critical role in lung homeostasis.
Collapse
Affiliation(s)
- Huimin Zhang
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, 4114 MRB III, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Ma G, Yang C, Qu Y, Wei H, Zhang T, Zhang N. The flavonoid component isorhamnetin in vitro inhibits proliferation and induces apoptosis in Eca-109 cells. Chem Biol Interact 2007; 167:153-60. [PMID: 17368593 DOI: 10.1016/j.cbi.2007.02.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2006] [Revised: 02/08/2007] [Accepted: 02/09/2007] [Indexed: 11/24/2022]
Abstract
Isorhamnetin is one member of flavonoid components which has been used in the treatment of heart disease. Recently the in vitro anti-cancer effect of isorhamnetin on human esophageal squamous carcinoma cell line Eca-109 was investigated in our lab. When Eca-109 cells were in vitro exposed to the graded doses of isorhamnetin (0-80 microg/ml) for 48 h, respectively, isorhamnetin exhibited cytostatic effect on the treated cells, with an IC(50) of 40+/-0.08 microg/ml as estimated by MTT assay. Inhibition on proliferation by isorhamnetin was detected by trypan blue exclusion assay, clone formation test, immunocytochemical assay of PCNA and (3)H-thymidine uptake analysis. Cell cycle distribution was measured by FCM. It was found that the viability of Eca-109 cells was significantly hampered by isorhamnetin. Compared with the negative control group, the treated group which was exposed to isorhamnetin had increased population in G(0)/G(1) phase from 74.6 to 84 while had a significant reduction in G(2)/M phase from 11.9 to 5.8. In addition to its cytostatic effect, isorhamnetin also showed stimulatory effect on apoptosis. Typical apoptotic morphology such as condensation and fragmentation of nuclei and blebbing membrane of the apoptotic cells could be observed through transmission electron microscope. Moreover, the sharp increase in apoptosis rate between the control and treated group were detected by FCM from 6.3 to 16.3. To explore the possible molecular mechanisms that underlie the growth inhibition and apoptosis stimulatory effects of isorhamnetin, the expressions of six proliferation- and death-related genes were detected by FCM. Expressions of bcl-2, c-myc and H-ras were downregulated whereas Bax, c-fos and p53 were upregulated. However, the in vivo experiments were required to further confirm the anti-cancer effects of isorhamnetin. In conclusion, isorhamnetin appears to be a potent drug against esophageal cancer due to its in vitro potential to not only inhibit proliferation but also induce apoptosis of Eca-109 cells.
Collapse
Affiliation(s)
- Gang Ma
- The Medicine-Biology Laboratory of Life Science, College of Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | | | | | | | | | | |
Collapse
|