1
|
Jeon EH, Park SY, Park KU, Lee YH. Ribosomal Protein L9 Maintains Stemness of Colorectal Cancer via an ID-1 Dependent Mechanism. J Cancer Prev 2024; 29:25-31. [PMID: 38957590 PMCID: PMC11215338 DOI: 10.15430/jcp.24.004] [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: 04/30/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
The identification of therapeutic target genes that are functionally involved in stemness is crucial to effectively cure patients with metastatic carcinoma. We have previously reported that inhibition of ribosomal protein L9 (RPL9) expression suppresses the growth of colorectal cancer (CRC) cells by inactivating the inhibitor of DNA-binding 1 (ID-1) signaling axis, which is functionally associated with cancer cell survival. In addition to cell proliferation, ID-1 is also involved in the maintenance of cancer stemness. Thus, we aimed in this study to investigate whether the function of RPL9 could correlate with CRC stem cell-like properties. Here, we demonstrated that siRNA silencing of RPL9 reduced the invasiveness and migrative capabilities of HT29 and HCT116 parental cell populations and the capacity for sphere formation in the HT29 parental cell population. CD133+ cancer stem cells (CSCs) were then separated from CD133- cancer cells of the HT29 parental cell culture and treated with RPL9-specific siRNAs to verify the effects of RPL9 targeting on stemness. As a result, knockdown of RPL9 significantly suppressed the proliferative potential of CD133+ colorectal CSCs, accompanied by a reduction in CD133, ID-1, and p-IκBα levels. In line with these molecular alterations, targeting RPL9 inhibited the invasion, migration, and sphere-forming capacity of CD133+ HT29 CSCs. Taken together, these findings suggest that RPL9 promotes CRC stemness via ID-1 and that RPL9 could be a potential therapeutic target for both primary CRC treatment and the prevention of metastasis and/or recurrence.
Collapse
Affiliation(s)
- Eun-Hye Jeon
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - So-Young Park
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Keon Uk Park
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Yun-Han Lee
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
2
|
Nguyen A, Sung Y, Lee SH, Martin CE, Srikanth S, Chen W, Kang MK, Kim RH, Park NH, Gwack Y, Kim Y, Shin KH. Orai3 Calcium Channel Contributes to Oral/Oropharyngeal Cancer Stemness through the Elevation of ID1 Expression. Cells 2023; 12:2225. [PMID: 37759448 PMCID: PMC10527097 DOI: 10.3390/cells12182225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Emerging evidence indicates that intracellular calcium (Ca2+) levels and their regulatory proteins play essential roles in normal stem cell proliferation and differentiation. Cancer stem-like cells (CSCs) are subpopulations of cancer cells that retain characteristics similar to stem cells and play an essential role in cancer progression. Recent studies have reported that the Orai3 calcium channel plays an oncogenic role in human cancer. However, its role in CSCs remains underexplored. In this study, we explored the effects of Orai3 in the progression and stemness of oral/oropharyngeal squamous cell carcinoma (OSCC). During the course of OSCC progression, the expression of Orai3 exhibited a stepwise augmentation. Notably, Orai3 was highly enriched in CSC populations of OSCC. Ectopic Orai3 expression in non-tumorigenic immortalized oral epithelial cells increased the intracellular Ca2+ levels, acquiring malignant growth and CSC properties. Conversely, silencing of the endogenous Orai3 in OSCC cells suppressed the CSC phenotype, indicating a pivotal role of Orai3 in CSC regulation. Moreover, Orai3 markedly increased the expression of inhibitor of DNA binding 1 (ID1), a stemness transcription factor. Orai3 and ID1 exhibited elevated expression within CSCs compared to their non-CSC counterparts, implying the functional importance of the Orai3/ID1 axis in CSC regulation. Furthermore, suppression of ID1 abrogated the CSC phenotype in the cell with ectopic Orai3 overexpression and OSCC. Our study reveals that Orai3 is a novel functional CSC regulator in OSCC and further suggests that Orai3 plays an oncogenic role in OSCC by promoting cancer stemness via ID1 upregulation.
Collapse
Affiliation(s)
- Anthony Nguyen
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Youngjae Sung
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Sung Hee Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Charlotte Ellen Martin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Wei Chen
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Mo K. Kang
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Reuben H. Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Yong Kim
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- Laboratory of Stem Cell and Cancer Epigenetics, UCLA School of Dentistry, Los Angeles, CA 90095, USA
- UCLA Broad Stem Cell Research Center, Los Angeles, CA 90095, USA
| | - Ki-Hyuk Shin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Lin Z, Liu Y, Xu T, Su T, Yang Y, Liang R, Gu S, Li J, Song X, Liang B, Leng Z, Li Y, Meng L, Luo Y, Chang X, Huang D, Xie L. STAT3-Mediated Promoter-Enhancer Interaction Up-Regulates Inhibitor of DNA Binding 1 ( ID1) to Promote Colon Cancer Progression. Int J Mol Sci 2023; 24:10041. [PMID: 37373188 DOI: 10.3390/ijms241210041] [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: 05/18/2023] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND High expression of inhibitor of DNA binding 1 (ID1) correlates with poor prognosis in colorectal cancer (CRC). Aberrant enhancer activation in regulating ID1 transcription is limited. METHODS Immunohistochemistry (IHC), quantitative RT-PCR (RT-qPCR) and Western blotting (WB) were used to determine the expression of ID1. CRISPR-Cas9 was used to generate ID1 or enhancer E1 knockout cell lines. Dual-luciferase reporter assay, chromosome conformation capture assay and ChIP-qPCR were used to determine the active enhancers of ID1. Cell Counting Kit 8, colony-forming, transwell assays and tumorigenicity in nude mice were used to investigate the biological functions of ID1 and enhancer E1. RESULTS Human CRC tissues and cell lines expressed a higher level of ID1 than normal controls. ID1 promoted CRC cell proliferation and colony formation. Enhancer E1 actively regulated ID1 promoter activity. Signal transducer and activator of transcription 3 (STAT3) bound to ID1 promoter and enhancer E1 to regulate their activity. The inhibitor of STAT3 Stattic attenuated ID1 promoter and enhancer E1 activity and the expression of ID1. Enhancer E1 knockout down-regulated ID1 expression level and cell proliferation in vitro and in vivo. CONCLUSIONS Enhancer E1 is positively regulated by STAT3 and contributes to the regulation of ID1 to promote CRC cell progression and might be a potential target for anti-CRC drug studies.
Collapse
Affiliation(s)
- Zhike Lin
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Ying Liu
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Tian Xu
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Ting Su
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Yingying Yang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Runhua Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Songgang Gu
- Department of Hepatobiliary Surgery, Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jie Li
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Xuhong Song
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Bin Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Zhijun Leng
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Yangsihan Li
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Lele Meng
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Yijing Luo
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Xiaolan Chang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou 515041, China
| | - Lingzhu Xie
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
4
|
Liu F, Chen S, Yu Y, Huang C, Chen H, Wang L, Zhang W, Wu J, Ye Y. Inhibitor of DNA binding 2 knockdown inhibits the growth and liver metastasis of colorectal cancer. Gene 2022; 819:146240. [PMID: 35114275 DOI: 10.1016/j.gene.2022.146240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Liver metastasis of colorectal cancer (CRC) remains high mortality and the mechanism is still unknown. Here we investigated the effects of inhibitor of DNA binding 2 (Id2) on growth and liver metastasis of CRC. METHODS qPCR and western blotting were used to demonstrate mRNA and protein expressions in Id2-knockdown HCT116 cells. Cell growth was observed by cell proliferation assay, colony formation assay and flow cytometry. Cell migration and invasion were observed with wound healing assay and transwell migration and invasion assay. The effects of Id2 knockdown on tumor growth and liver metastasis in vivo were evaluated respectively with subcutaneous tumor model and colorectal liver metastasis model by injecting HCT116 cells into the mesentery triangle of cecum in mice. RESULTS Id2 overexpression was found in CRC cell lines. Id2 knockdown resulted in a reduction in the proliferation, colony formation, migration and invasion of HCT116 cells. The suppression of cell proliferation was accompanied by the cell cycle arrest in the G0/G1 phase with down-regulation of Cyclin D1, Cyclin E, p-Cdk2/3, Cdk6, p-p27 and up-regulation of p21 and p27. Id2 knockdown reversed epithelial-mesenchymal transition (EMT) through increasing E-Cadherin and inhibiting N-Cadherin, Vimentin, β-catenin, Snail and Slug. Id2 was also found to inhibit CRC metastasis via MMP2, MMP9 and TIMP-1. Furthermore, Id2 knockdown suppressed CRC liver metastasis in vivo. CONCLUSION Id2 promotes CRC growth through activation of the PI3K/AKT signaling pathway, and triggers EMT to enhance CRC migration and invasion.
Collapse
Affiliation(s)
- Fang Liu
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Shuping Chen
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Yue Yu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Chuanzhong Huang
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Huijing Chen
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Ling Wang
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Wanping Zhang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Junxin Wu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China.
| | - Yunbin Ye
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China.
| |
Collapse
|
5
|
Niu B, Liu J, Lv B, Lin J, Li X, Wu C, Jiang X, Zeng Z, Zhang XK, Zhou H. Interplay between transforming growth factor-β and Nur77 in dual regulations of inhibitor of differentiation 1 for colonic tumorigenesis. Nat Commun 2021; 12:2809. [PMID: 33990575 PMCID: PMC8121807 DOI: 10.1038/s41467-021-23048-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/14/2021] [Indexed: 01/04/2023] Open
Abstract
The paradoxical roles of transforming growth factor-β (TGFβ) signaling and nuclear receptor Nur77 in colon cancer development are known but the underlying mechanisms remain obscure. Inhibitor of differentiation 1 (ID1) is a target gene of TGFβ and a key promoter for colon cancer progression. Here, we show that Nur77 enhances TGFβ/Smad3-induced ID1 mRNA expression through hindering Smurf2-mediated Smad3 mono-ubiquitylation, resulting in ID1 upregulation. In the absence of TGFβ, however, Nur77 destabilizes ID1 protein by promoting Smurf2-mediated ID1 poly-ubiquitylation, resulting in ID1 downregulation. Interestingly, TGFβ stabilizes ID1 protein by switching Nur77 interaction partners to inhibit ID1 ubiquitylation. This also endows TGFβ with an active pro-tumorigenic action in Smad4-deficient colon cancers. Thus, TGFβ converts Nur77's role from destabilizing ID1 protein and cancer inhibition to inducing ID1 mRNA expression and cancer promotion, which is highly relevant to colon cancer stemness, metastasis and oxaliplatin resistance. Our data therefore define the integrated duality of Nur77 and TGFβ signaling in regulating ID1 expression and provide mechanistic insights into the paradoxical roles of TGFβ and Nur77 in colon cancer progression.
Collapse
MESH Headings
- Animals
- Carcinogenesis
- Cell Line, Tumor
- Colonic Neoplasms/etiology
- Colonic Neoplasms/genetics
- Colonic Neoplasms/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- HCT116 Cells
- HT29 Cells
- Humans
- Inhibitor of Differentiation Protein 1/genetics
- Inhibitor of Differentiation Protein 1/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Models, Biological
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Protein Stability
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Smad3 Protein/metabolism
- Smad4 Protein/deficiency
- Smad4 Protein/metabolism
- Transforming Growth Factor beta/metabolism
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitination
Collapse
Affiliation(s)
- Boning Niu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, China
| | - Jie Liu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, China
| | - Ben Lv
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, China
| | - Jiacheng Lin
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xin Li
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, China
| | - Chunxiao Wu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, China
| | - Xiaohua Jiang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, China
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
6
|
Leong SP, Witz IP, Sagi-Assif O, Izraely S, Sleeman J, Piening B, Fox BA, Bifulco CB, Martini R, Newman L, Davis M, Sanders LM, Haussler D, Vaske OM, Witte M. Cancer microenvironment and genomics: evolution in process. Clin Exp Metastasis 2021; 39:85-99. [PMID: 33970362 DOI: 10.1007/s10585-021-10097-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
Cancer heterogeneity is a result of genetic mutations within the cancer cells. Their proliferation is not only driven by autocrine functions but also under the influence of cancer microenvironment, which consists of normal stromal cells such as infiltrating immune cells, cancer-associated fibroblasts, endothelial cells, pericytes, vascular and lymphatic channels. The relationship between cancer cells and cancer microenvironment is a critical one and we are just on the verge to understand it on a molecular level. Cancer microenvironment may serve as a selective force to modulate cancer cells to allow them to evolve into more aggressive clones with ability to invade the lymphatic or vascular channels to spread to regional lymph nodes and distant sites. It is important to understand these steps of cancer evolution within the cancer microenvironment towards invasion so that therapeutic strategies can be developed to control or stop these processes.
Collapse
Affiliation(s)
- Stanley P Leong
- California Pacific Medical Center and Research Institute, San Francisco, USA
| | - Isaac P Witz
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Orit Sagi-Assif
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Izraely
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Jonathan Sleeman
- European Center for Angioscience, Medizinische Fakultät Mannheim der Universität Heidelberg, Heidelberg, Germany
| | | | | | | | - Rachel Martini
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA.,Department of Genetics, University of Georgia, Athens, GA, USA
| | - Lisa Newman
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA
| | - Melissa Davis
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA.
| | - Lauren M Sanders
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz and UC Santa Cruz Genomics Institute, Santa Cruz, USA
| | - David Haussler
- UC Santa Cruz Genomics Institute and Howard Hughes Medical Institute, University of California Santa Cruz, Santa Cruz, USA.
| | - Olena M Vaske
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz and UC Santa Cruz Genomics Institute, Santa Cruz, USA
| | - Marlys Witte
- Department of Surgery, Neurosurgery and Pediatrics, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| |
Collapse
|
7
|
Id4 Suppresses the Growth and Invasion of Colorectal Cancer HCT116 Cells through CK18-Related Inhibition of AKT and EMT Signaling. JOURNAL OF ONCOLOGY 2021; 2021:6660486. [PMID: 33936204 PMCID: PMC8060092 DOI: 10.1155/2021/6660486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/19/2021] [Accepted: 04/02/2021] [Indexed: 01/05/2023]
Abstract
Id4 is one of the inhibitors of DNA-binding proteins (Id) and involved in the pathogenesis of numerous cancers. The specific mechanism underlying the Id4-mediated regulation of proliferation, invasion, and metastasis of colorectal cancer (CRC) cells is still largely unclear. In the present study, results showed CRC cells had a lower baseline Id4 expression than normal intestinal epithelial NCM460 cells. In order to explore the role of Id4 in the tumorigenicity, CRC HCT116 cells with stable Id4 expression were used, and results showed Id4 overexpression arrested the cell cycle at the G0/G1 phase, inhibited the cell proliferation and the colony formation, as well as suppressed the migration and invasion. In the in vivo model, Id4 overexpression inhibited the tumor growth and metastasis in the nude mice. Furthermore, Id4 overexpression upregulated the expression of proteins associated with cell proliferation, inhibited the PI3K/AKT pathway, and suppressed epithelial-mesenchymal transition (EMT) of HCT116 cells. Moreover, Id4 significantly decreased cytokeratin 18 (CK18) expression, but CK18 overexpression in Id4 expressing HCT116-Id4 cells rescued the activation of AKT, p-AKT, MMP2, MMP7, and E-cadherin. Collectively, our study indicated Id4 may inhibit CRC growth and metastasis through inhibiting the PI3K/AKT pathway in a CK18-dependent manner and suppressing EMT. Id4 may become a target for the treatment of CRC.
Collapse
|
8
|
Kim HR, Moon JH, Lee JH, Lim YC. Inhibitor of DNA Binding 2 (ID2): A Novel Marker for Lymph Node Metastasis in Head and Neck Squamous Cell Carcinoma. Ann Surg Oncol 2021; 28:6479-6488. [PMID: 33783641 DOI: 10.1245/s10434-021-09832-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 02/19/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Although aggressive invasion and sequential lymph node metastasis (LNM) significantly affect the prognosis of patients with head and neck squamous cell carcinoma (HNSCC), studies on identifying the factors that regulate this process remain scarce. This study found an inhibitor of DNA binding 2 (ID2) as a novel molecule involved in the regulation of invasion and LNM of HNSCC and further verified its functional role. METHODS The study examined the translational significance between ID2 expression levels and the presence of LNM as well as the prognosis for 119 patients with HNSCC after treatment. In addition, in vitro and in vivo experiments were performed using ID2 gene-modulated HNSCC cell lines to determine the functional role of ID2 in the invasion and LNM of HNSCC. RESULTS Elevated levels of ID2 expression were closely associated with the presence of LNM in 119 patients with HNSCC, resulting in a poor prognosis. Overexpression of ID2-induced invasion and LNM of HNSCC cells was observed in vitro and in vivo. By contrast, knockdown of the ID2 gene diminished invasion and LNM of HNSCC cells. In addition, the ID2 expression level increased the expression level of matrix metalloproteinase 1 (MMP1), a molecule downstream to ID2. Furthermore, silencing of MMP1 in ID2-overexpressed HNSCC cells rescued the elevated invasion and LNM capabilities of these cells, suggesting that ID2 enhances invasion and LNM partly via MMP1 activation. CONCLUSION In the invasion and LNM of HNSCC, ID2 plays an important role by modulating MMP1 expression, suggesting ID2-MMP1 axis to be a novel alternative therapeutic target for invasion and LNM of HNSCC.
Collapse
Affiliation(s)
- Hye Ryun Kim
- Department of Otorhinolaryngology - Head and Neck Surgery, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Jung Hwa Moon
- Department of Otorhinolaryngology - Head and Neck Surgery, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Jun Hwan Lee
- Department of Otorhinolaryngology - Head and Neck Surgery, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Young Chang Lim
- Department of Otorhinolaryngology - Head and Neck Surgery, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Sedlmeier G, Al‐Rawi V, Buchert J, Yserentant K, Rothley M, Steshina A, Gräßle S, Wu R, Hurrle T, Richer W, Decraene C, Thiele W, Utikal J, Abuillan W, Tanaka M, Herten D, Hill CS, Garvalov BK, Jung N, Bräse S, Sleeman JP. Id1 and Id3 Are Regulated Through Matrix‐Assisted Autocrine BMP Signaling and Represent Therapeutic Targets in Melanoma. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Georg Sedlmeier
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Vanessa Al‐Rawi
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Justyna Buchert
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Klaus Yserentant
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- College of Medical and Dental Sciences & School of Chemistry University of Birmingham Birmingham UK
- Centre of Membrane Proteins and Receptors (COMPARE) Universities of Birmingham and Nottingham UK
| | - Melanie Rothley
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Anastasia Steshina
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Simone Gräßle
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Ruo‐Lin Wu
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Thomas Hurrle
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
| | - Wilfrid Richer
- CNRS UMR144 Translational Research Department Institut Curie PSL Research University 26 rue d'Ulm Paris Cedex 05 75248 France
| | - Charles Decraene
- CNRS UMR144 Translational Research Department Institut Curie PSL Research University 26 rue d'Ulm Paris Cedex 05 75248 France
| | - Wilko Thiele
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Jochen Utikal
- Skin Cancer Unit German Cancer Research Center (DKFZ) Im Neuenheimer Feld 280 69120 Heidelberg Germany
- Department of Dermatology, Venereology and Allergology University Medical Center Mannheim Ruprecht‐Karl University of Heidelberg Theodor‐Kutzer‐Ufer 1–3 68167 Mannheim Germany
| | - Wasim Abuillan
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
| | - Motomu Tanaka
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- Center for Integrative Medicine and Physics Institute for Advanced Study Kyoto University Yoshida Ushinomiya‐cho Sakyo‐Ku Kyoto 606‐8501 Japan
- Center for Integrative Medicine and Physics Institute for Advanced Study, Kyoto University Kyoto 606‐8501 Japan
| | - Dirk‐Peter Herten
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- College of Medical and Dental Sciences & School of Chemistry University of Birmingham Birmingham UK
- Centre of Membrane Proteins and Receptors (COMPARE) Universities of Birmingham and Nottingham UK
| | | | - Boyan K. Garvalov
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Nicole Jung
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Stefan Bräse
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Jonathan P. Sleeman
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| |
Collapse
|
10
|
Zhao Z, Bo Z, Gong W, Guo Y. Inhibitor of Differentiation 1 (Id1) in Cancer and Cancer Therapy. Int J Med Sci 2020; 17:995-1005. [PMID: 32410828 PMCID: PMC7211148 DOI: 10.7150/ijms.42805] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The inhibitor of DNA binding (Id) proteins are regulators of cell cycle and cell differentiation. Of all Id family proteins, Id1 is mostly linked to tumorigenesis, cellular senescence as well as cell proliferation and survival. Id1 is a stem cell-like gene more than a classical oncogene. Id1 is overexpressed in numerous types of cancers and exerts its promotion effect to these tumors through different pathways. Briefly, Id1 was found significantly correlated with EMT-related proteins, K-Ras signaling, EGFR signaling, BMP signaling, PI3K/Akt signaling, WNT and SHH signaling, c-Myc signaling, STAT3 signaling, RK1/2 MAPK/Egr1 pathway and TGF-β pathway, etc. Id1 has potent effect on facilitating tumorous angiogenesis and metastasis. Moreover, high expression of Id1 plays a facilitating role in the development of drug resistance, including chemoresistance, radiation resistance and resistance to drugs targeting angiogenesis. However, controversial results were also obtained. Overall, Id1 represent a promising target of anti-tumor therapeutics based on its potent promotion effect to cancer. Numerous drugs were found exerting their anti-tumor function through Id1-related signaling pathways, such as fucoidan, berberine, tetramethylpyrazine, crizotinib, cannabidiol and vinblastine.
Collapse
Affiliation(s)
- Zhengxiao Zhao
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Zhiyuan Bo
- The Second Department of Biliary Tract Surgery, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China
| | - Weiyi Gong
- The Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, PR China
| | - Yong Guo
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
11
|
Zhu Y, Chen W, Guan W, Fang Y, Hao C. Study of As2O3 regulating proliferation and apoptosis of Tca8113 cells by inhibiting the expression of Id-1. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1932-1937. [PMID: 31079498 DOI: 10.1080/21691401.2019.1613419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Yanfeng Zhu
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Weihui Chen
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Weiqun Guan
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yihong Fang
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chunfeng Hao
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
12
|
Sun B, Dong C, Lei H, Gong Y, Li M, Zhang Y, Zhang H, Sun L. Knockdown of inhibitor of differentiation 1 suppresses proliferation and induces apoptosis by inactivating PI3K/Akt/mTOR signaling in hemangioma-derived endothelial cells. Biomed Pharmacother 2019; 111:236-243. [DOI: 10.1016/j.biopha.2018.12.072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023] Open
|
13
|
Sim MY, Huynh H, Go ML, Yuen JSP. Action of YM155 on clear cell renal cell carcinoma does not depend on survivin expression levels. PLoS One 2017; 12:e0178168. [PMID: 28582447 PMCID: PMC5459331 DOI: 10.1371/journal.pone.0178168] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 05/08/2017] [Indexed: 12/12/2022] Open
Abstract
The dioxonapthoimidazolium YM155 is a survivin suppressant which has been investigated as an anticancer agent in clinical trials. Here, we investigated its growth inhibitory properties on a panel of immortalized and patient derived renal cell carcinoma (RCC) cell lines which were either deficient in the tumour suppressor von Hippel-Lindau (VHL) protein or possessed a functional copy. Neither the VHL status nor the survivin expression levels of these cell lines influenced their susceptibility to growth inhibition by YM155. Of the various RCC lines, the papillary subtype was more resistant to YM155, suggesting that the therapeutic efficacy of YM155 may be restricted to clear cell subtypes. YM155 was equally potent in cells (RCC786.0) in which survivin expression had been stably silenced or overexpressed, implicating a limited reliance on survivin in the mode of action of YM155. A follow-up in-vitro high throughput RNA microarray identified possible targets of YM155 apart from survivin. Selected genes (ID1, FOXO1, CYLD) that were differentially expressed in YM155-sensitive RCC cells and relevant to RCC pathology were validated with real-time PCR and western immunoblotting analyses. Thus, there is corroboratory evidence that the growth inhibitory activity of YM155 in RCC cell lines is not exclusively mediated by its suppression of survivin. In view of the growing importance of combination therapy in oncology, we showed that a combination of YM155 and sorafenib at ½ x IC50 concentrations was synergistic on RCC786.0 cells. However, when tested intraperitoneally on a murine xenograft model derived from a nephrectomised patient with clear cell RCC, a combination of suboptimal doses of both drugs failed to arrest tumour progression. The absence of synergy in vivo highlighted the need to further optimize the dosing schedules of YM155 and sorafenib, as well as their routes of administration. It also implied that the expression of other oncogenic proteins which YM155 may target is either low or absent in this clear cell RCC.
Collapse
Affiliation(s)
- Mei Yi Sim
- Department of Urology, Singapore General Hospital, Republic of Singapore
- * E-mail:
| | - Hung Huynh
- Laboratory of Molecular Endocrinology, Division of Molecular and Cellular Research, National Cancer Centre, Republic of Singapore
| | - Mei Lin Go
- Department of Pharmacy, National University of Singapore, Republic of Singapore
| | | |
Collapse
|
14
|
Li L, Wei X, Wu B, Xiao Y, Yin M, Yang Q. siRNA-mediated knockdown of ID1 disrupts Nanog- and Oct-4-mediated cancer stem cell-likeness and resistance to chemotherapy in gastric cancer cells. Oncol Lett 2017; 13:3014-3024. [PMID: 28529558 PMCID: PMC5431526 DOI: 10.3892/ol.2017.5828] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/07/2016] [Indexed: 12/15/2022] Open
Abstract
DNA-binding protein inhibitor ID-1 (ID1) serves an essential role in tumor progression, and the self-renewal and pluripotency of embryonic stem cells. However, the effect of ID1 on the stemness and cancer stem cell (CSC)-like properties of gastric adenocarcinoma cells remains to be elucidated. In the present study, effective ID1 knockdown was achieved in gastric cancer (GC) cells using small interfering RNA, and the self-renewal ability and cisplatin (DDP) sensitivity of GC cells was subsequently examined. ID1 knockdown in the MKN-28 and MGC-803 cell lines was demonstrated to significantly suppress colony formation (P=0.005 in MKN-28 and P=0.001 in MGC-803), tumor spheroid formation (P=0.021 in MKN-28 and P=0.037 in MGC-803), cell proliferation (P=0.028 in MKN-28 and P=0.001 in MGC-803) and migration (P=0.002 in MKN-28 and P=0.015 in MGC-803). To the best of our knowledge, the present study revealed for the first time that ID1 knockdown suppresses the expression of the key CSC-associated factors Nanog and octamer-binding protein 4 (Oct-4). It was further demonstrated that ID1 knockdown sensitized GC cells to DDP. In conclusion, knockdown of ID1 attenuates the stem cell like-properties of self-renewal in normal GC cells, potentially through the targeting of Nanog and Oct-4, and subsequently decreases cell proliferation and resistance to DDP. The results of the present study suggest that ID1 functions as an oncogene in GC and regulates the stem cell like-properties of gastric cancer cells by targeting Nanog and Oct-4.
Collapse
Affiliation(s)
- Linlin Li
- Cancer Research Institute, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaoyong Wei
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Baofeng Wu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Yuanli Xiao
- Department of Gastroenterology, Pingdingshan Second People's Hospital, Pingdingshan, Henan 467000, P.R. China
| | - Mingzhu Yin
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Qiaohong Yang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
15
|
Baik IH, Jo GH, Seo D, Ko MJ, Cho CH, Lee MG, Lee YH. Knockdown of RPL9 expression inhibits colorectal carcinoma growth via the inactivation of Id-1/NF-κB signaling axis. Int J Oncol 2016; 49:1953-1962. [PMID: 27633352 DOI: 10.3892/ijo.2016.3688] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/25/2016] [Indexed: 11/06/2022] Open
Abstract
Ribosomal protein L9 (RPL9), a component of the 60S subunit for protein synthesis, is upregulated in human colorectal cancer. In the present study, we investigated whether RPL9 gained extraribosomal function during tumorigenesis and whether targeting of RPL9 with small interfering (si) RNA could alter the course of colorectal cancer progression. Our results showed that siRNA knockdown of RPL9 suppresses colorectal cancer (CRC) cell growth and long-term colony formation through an increase in sub-G1 cell population and a strong induction of apoptotic cell death. To obtain insights into the molecular changes in response to RPL9 knockdown, global changes in gene expression were examined using RNA sequencing. It revealed that RPL9-specific knockdown led to dysregulation of 918 genes in HCT116 and 3178 genes in HT29 cells. Among these, 296 genes showed same directional regulation (128 upregulated and 168 downregulated genes) and were considered as a common RPL9 knockdown signature. Particularly, we found through a network analysis that Id-1, which is functionally associated with activation of NF-κB and cell survival, was commonly downregulated. Subsequent western blot analysis affirmed that RPL9 silencing induced the decrease in the levels of Id-1 and phosphorylated IκBα in both HCT116 and HT29 cells. Also, the same condition decreased the levels of PARP-1 and pro-caspase-3, accelerating apoptosis. Furthermore, inhibition of RPL9 expression significantly suppressed the growth of human CRC xenografts in nude mice. These findings indicate that the function of RPL9 is correlated with Id-1/NF-κB signaling axis and suggest that targeting RPL9 could be an attractive option for molecular therapy of colorectal cancer.
Collapse
Affiliation(s)
- In Hye Baik
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Guk-Heui Jo
- Myunggok Eye Research Institute, Kim's Eye Hospital, Konyang University College of Medicine, Seoul, Republic of Korea
| | - Daekwan Seo
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Ji Ko
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Chi Heum Cho
- Department of Obstetrics and Gynecology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Min Goo Lee
- Department of Pharmacology and Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yun-Han Lee
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
16
|
Lin G, Huang C, Su G, Hu H, Xu H, Huang C. [Effect of TRAF6 Downregulation on Malignant Biological Behavior of
Lung Cancer Cell Lines]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 18:661-7. [PMID: 26582220 PMCID: PMC6000311 DOI: 10.3779/j.issn.1009-3419.2015.11.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
背景与目的 已有的研究提示肿瘤坏死因子受体相关因子6(tumor necrosis factor receptor-associated factor 6, TRAF6)在肺癌中常常扩增,可能扮演癌基因角色,但TRAF6的确切作用尚未充分阐明。本研究探索TRAF6表达对肺癌细胞株的增殖、凋亡、细胞周期、迁移及侵袭能力的影响以及可能作用机制。 方法 选用A549、H1650、SPC-A-1以及Calu-3等四种肺癌细胞株,应用蛋白印迹、qRT-PCR检测其TRAF6蛋白及mRNA表达。SPC-A-1、Calu-3细胞转染TRAF6 siRNA,以EMSA方法检测不同处理组核因子-κB的DNA结合活性,MTS法检测细胞增殖,流式细胞仪PI染色检测细胞凋亡,流式细胞仪进行细胞周期测定,划痕实验及Transwell小室法检测细胞迁移及侵袭能力,并应用蛋白印迹检测泛素化抗体、p65、CD24、CXCR4等蛋白表达。SPC-A-1细胞提取DNA后,应用二代测序法进行全基因组测序。 结果 在四种细胞株中,SPC-A-1和Calu-3细胞TRAF6相对高表达,TRAF6发生自身K63-泛素化,但仅在SPC-A-1细胞中观察到核因子-κB组成性活化。转染TRAF6 siRNA后,SPC-A-1、Calu-3细胞TRAF6表达明显下调,与空白组及对照组相比,下调TRAF6表达可抑制SPC-A-1细胞核因子-κB活性、降低迁移及侵袭能力以及促进细胞凋亡,CD24和CXCR4的表达也明显下调,但对细胞增殖及细胞周期无明显影响。下调TRAF6表达对Calu-3细胞株的核因子-κB活性、细胞增殖、凋亡、细胞周期、迁移及侵袭能力等均无明显影响。未发现SPC-A-1细胞株TRAF6基因突变或拷贝数改变。 结论 下调TRAF6表达可抑制SPC-A-1细胞迁移及侵袭能力,促进细胞凋亡,并且TRAF6可能是通过调控核因子-κB-CD24/CXCR4信号通路参与调控肺癌侵袭、细胞凋亡。
Collapse
Affiliation(s)
- Gen Lin
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Teaching Hospital, Fuzhou 350014, China
| | - Chuangzhong Huang
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Teaching Hospital, Fuzhou 350014, China
| | - Guangjian Su
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Teaching Hospital, Fuzhou 350014, China
| | - Huihua Hu
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Teaching Hospital, Fuzhou 350014, China
| | - Haipeng Xu
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Teaching Hospital, Fuzhou 350014, China
| | - Cheng Huang
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Teaching Hospital, Fuzhou 350014, China
| |
Collapse
|
17
|
Narayanan NK, Kunimasa K, Yamori Y, Mori M, Mori H, Nakamura K, Miller G, Manne U, Tiwari AK, Narayanan B. Antitumor activity of melinjo (Gnetum gnemon L.) seed extract in human and murine tumor models in vitro and in a colon-26 tumor-bearing mouse model in vivo. Cancer Med 2015; 4:1767-80. [PMID: 26408414 PMCID: PMC4674003 DOI: 10.1002/cam4.520] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 08/02/2015] [Accepted: 08/04/2015] [Indexed: 01/05/2023] Open
Abstract
Melinjo (Gnetum gnemon L.) seed extract (MSE) and its active ingredient gnetin C (GC), a resveratrol dimer, have been shown to possess a broad spectrum of pharmacological activities. In this study, we investigated the antitumor activity of MSE and GC using human and murine tumor cell culture models in vitro. The antitumor activity of GC was compared with trans-resveratrol (tRV), a stilbenoid polyphenol. Our results show that MSE and GC at clinically achievable concentrations significantly inhibited the proliferation of pancreatic, prostate, breast, and colon cancer cell types (P < 0.05), without affecting normal cells. Interestingly, GC exerts enhanced antitumor activity than that of tRV (P < 0.05). MSE and GC significantly induced apoptosis in all the cancer cells, indicating MSE and GC inhibit tumor cell growth by inducing apoptosis (P < 0.001). Our findings provide evidence that MSE might induce apoptosis in cancer cells via caspase-3/7-dependent and -independent mechanisms. However, GC might trigger both early and late stage apoptosis in cancer cells, at least in part by activating caspase 3/7-dependent mechanisms. Furthermore, the antitumor efficacy of MSE observed in vitro was also validated in a widely used colon-26 tumor-bearing mouse model. Oral administration of MSE at 50 and 100 mg/kg per day significantly inhibited tumor growth, intratumoral angiogenesis, and liver metastases in BALB/c mice bearing colon-26 tumors (P < 0.05). In conclusion, our findings provide evidence that MSE and GC have potent antitumor activity. Most importantly, we provide the first evidence that MSE inhibits tumor growth, intratumoral angiogenesis, and liver metastasis in a colon-26 tumor-bearing mice.
Collapse
Affiliation(s)
- Narayanan K Narayanan
- Department of Environmental Medicine, New York University School of Medicine, New York, New York
| | - Kazuhiro Kunimasa
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan.,Institution for World Health Development, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Yukio Yamori
- Institution for World Health Development, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Mari Mori
- Institution for World Health Development, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Hideki Mori
- Institution for World Health Development, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Kazuki Nakamura
- School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - George Miller
- Departments of Surgery and Cell Biology, New York University School of Medicine, New York, New York
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, Ohio
| | - Bhagavathi Narayanan
- Department of Environmental Medicine, New York University School of Medicine, New York, New York
| |
Collapse
|
18
|
Yan H, Wang W, Dou C, Tian F, Qi S. Securin promotes migration and invasion via matrix metalloproteinases in glioma cells. Oncol Lett 2015; 9:2895-2901. [PMID: 26137166 DOI: 10.3892/ol.2015.3074] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 03/10/2015] [Indexed: 12/27/2022] Open
Abstract
Human securin, encoded by pituitary tumor transforming gene 1, is implicated in several oncogenic processes in the pathogenesis of brain tumors, including glioma. The aim of the present study was to examine the effect of securin on the migration and invasion of glioma cells. The results revealed that the overexpression of securin in glioma LN-229 cells significantly increased the invasion and transmigration abilities. By contrast, these abilities were significantly reduced by the downregulation of securin in glioma U373 cells. Furthermore, the results demonstrated that securin overexpression and downregulation significantly increased and decreased the levels of matrix metalloproteinase 2 and 9, respectively. These findings indicate a promotive role for securin in glioma migration and invasion, which may involve the action of matrix metalloproteinases.
Collapse
Affiliation(s)
- Haicheng Yan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wei Wang
- Department of Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia 010017, P.R. China
| | - Changwu Dou
- Department of Neurosurgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010051, P.R. China
| | - Fuming Tian
- Department of Neurosurgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010051, P.R. China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|