1
|
Al-U’datt DGF, Tranchant CC, Al-Husein B, Hiram R, Al-Dwairi A, AlQudah M, Al-shboul O, Jaradat S, Alqbelat J, Almajwal A. Involvement and possible role of transglutaminases 1 and 2 in mediating fibrotic signalling, collagen cross-linking and cell proliferation in neonatal rat ventricular fibroblasts. PLoS One 2023; 18:e0281320. [PMID: 36848364 PMCID: PMC9970086 DOI: 10.1371/journal.pone.0281320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/19/2023] [Indexed: 03/01/2023] Open
Abstract
Transglutaminase (TG) isoforms control diverse normal and pathophysiologic processes through their capacity to cross-link extracellular matrix (ECM) proteins. Their functional and signalling roles in cardiac fibrosis remain poorly understood, despite some evidence of TG2 involvement in abnormal ECM remodelling in heart diseases. In this study, we investigated the role of TG1 and TG2 in mediating fibrotic signalling, collagen cross-linking, and cell proliferation in healthy fibroblasts by siRNA-mediated knockdown. siRNA for TG1, TG2 or negative control was transfected into cultured neonatal rat ventricular fibroblasts and cardiomyocytes. mRNA expression of TGs and profibrotic, proliferation and apoptotic markers was assessed by qPCR. Cell proliferation and soluble and insoluble collagen were determined by ELISA and LC-MS/MS, respectively. TG1 and TG2 were both expressed in neonatal rat cardiomyocytes and fibroblasts before transfection. Other TGs were not detected before and after transfection. TG2 was predominantly expressed and more effectively silenced than TG1. Knocking down TG1 or TG2 significantly modified profibrotic markers mRNA expression in fibroblasts, decreasing connective tissue growth factor (CTGF) and increasing transforming growth factor-β1 compared to the negative siRNA control. Reduced expression of collagen 3A1 was found upon TG1 knockdown, while TG2 knockdown raised α-smooth muscle actin expression. TG2 knockdown further increased fibroblast proliferation and the expression of proliferation marker cyclin D1. Lower insoluble collagen content and collagen cross-linking were evidenced upon silencing TG1 or TG2. Transcript levels of collagen 1A1, fibronectin 1, matrix metalloproteinase-2, cyclin E2, and BCL-2-associated X protein/B-cell lymphoma 2 ratio were strongly correlated with TG1 mRNA expression, whereas TG2 expression correlated strongly with CTGF mRNA abundance. These findings support a functional and signalling role for TG1 and TG2 from fibroblasts in regulating key processes underlying myocardial ECM homeostasis and dysregulation, suggesting that these isoforms could be potential and promising targets for the development of cardiac fibrosis therapies.
Collapse
Affiliation(s)
- Doa’a G. F. Al-U’datt
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Carole C. Tranchant
- School of Food Science, Nutrition and Family Studies, Faculty of Health Sciences and Community Services, Université de Moncton, New Brunswick, Canada
| | - Belal Al-Husein
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Roddy Hiram
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Ahmed Al-Dwairi
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammad AlQudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
- Physiology Department, Arabian Gulf University, Manama, Bahrain
| | - Othman Al-shboul
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Saied Jaradat
- Princess Haya Biotechnology Center, Jordan University of Science and Technology, Irbid, Jordan
| | - Jenan Alqbelat
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Zaykov V, Chaqour B. The CCN2/CTGF interactome: an approach to understanding the versatility of CCN2/CTGF molecular activities. J Cell Commun Signal 2021; 15:567-580. [PMID: 34613590 DOI: 10.1007/s12079-021-00650-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/23/2021] [Indexed: 01/16/2023] Open
Abstract
Cellular communication network 2 (CCN2), also known as connective tissue growth factor (CTGF) regulates diverse cellular processes, some at odds with others, including adhesion, proliferation, apoptosis, and extracellular matrix (ECM) protein synthesis. Although a cause-and-effect relationship between CCN2/CTGF expression and local fibrotic reactions has initially been established, CCN2/CTGF manifests cell-, tissue-, and context-specific functions and differentially affects developmental and pathological processes ranging from progenitor cell fate decisions and angiogenesis to inflammation and tumorigenesis. CCN2/CTGF multimodular structure, binding to and activation or inhibition of multiple cell surface receptors, growth factors and ECM proteins, and susceptibility for proteolytic cleavage highlight the complexity to CCN2/CTGF biochemical attributes. CCN2/CTGF expression and dosage in the local environment affects a defined community of its interacting partners, and this results in sequestration of growth factors, interference with or potentiation of ligand-receptor binding, cellular internalization of CCN2/CTGF, inhibition or activation of proteases, and generation of CCN2/CTGF degradome products that add molecular diversity and expand the repertoire of functional modules in the cells and their microenvironment. Through these interactions, different intracellular signals and cellular responses are elicited culminating into physiological or pathological reactions. Thus, the CCN2/CTGF interactome is a defining factor of its tissue- and context-specific effects. Mapping of new CCN2/CTGF binding partners might shed light on yet unknown roles of CCN2/CTGF and provide a solid basis for tissue-specific targeting this molecule or its interacting partners in a therapeutic context.
Collapse
Affiliation(s)
- Viktor Zaykov
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA
| | - Brahim Chaqour
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA. .,Department of Ophthalmology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
3
|
Chen Z, Zhang N, Chu HY, Yu Y, Zhang ZK, Zhang G, Zhang BT. Connective Tissue Growth Factor: From Molecular Understandings to Drug Discovery. Front Cell Dev Biol 2020; 8:593269. [PMID: 33195264 PMCID: PMC7658337 DOI: 10.3389/fcell.2020.593269] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/09/2020] [Indexed: 01/18/2023] Open
Abstract
Connective tissue growth factor (CTGF) is a key signaling and regulatory molecule involved in different biological processes, such as cell proliferation, angiogenesis, and wound healing, as well as multiple pathologies, such as tumor development and tissue fibrosis. Although the underlying mechanisms of CTGF remain incompletely understood, a commonly accepted theory is that the interactions between different protein domains in CTGF and other various regulatory proteins and ligands contribute to its variety of functions. Here, we highlight the structure of each domain of CTGF and its biology functions in physiological conditions. We further summarized main diseases that are deeply influenced by CTGF domains and the potential targets of these diseases. Finally, we address the advantages and disadvantages of current drugs targeting CTGF and provide the perspective for the drug discovery of the next generation of CTGF inhibitors based on aptamers.
Collapse
Affiliation(s)
- Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ning Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hang Yin Chu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zong-Kang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
4
|
Zhang Y, Sun X. Role of Focal Adhesion Kinase in Head and Neck Squamous Cell Carcinoma and Its Therapeutic Prospect. Onco Targets Ther 2020; 13:10207-10220. [PMID: 33116602 PMCID: PMC7553669 DOI: 10.2147/ott.s270342] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Head and neck cancers are one of the most prevalent cancers globally. Among them, head and neck squamous cell carcinoma (HNSCC) accounts for approximately 90% of head and neck cancers, which occurs in the oral cavity, oral pharynx, hypopharynx and larynx. The 5-year survival rate of HNSCC patients is only 63%, mainly because about 80–90% of patients with advanced HNSCC tend to suffer from local recurrence or even distant metastasis. Despite the more in-depth understanding of the molecular mechanisms underlying the occurrence and progression of HNSCC in recent years, effective targeted therapies are unavailable for HNSCC, which emphasize the urgent demand for studies in this area. Focal adhesion kinase (FAK) is an intracellular non-receptor tyrosine kinase that contributes to oncogenesis and tumor progression by its significant function in cell survival, proliferation, adhesion, invasion and migration. In addition, FAK exerts an effect on the tumor microenvironment, epithelial–mesenchymal transition, radiation (chemotherapy) resistance, tumor stem cells and regulation of inflammatory factors. Moreover, the overexpression and activation of FAK are detected in multiple types of tumors, including HNSCC. FAK inhibition can induce cell cycle arrest and apoptosis, significantly decrease cell growth, invasion and migration in HNSCC cell lines. In this article, we mainly review the research progress of FAK in the occurrence, development and metastasis of HNSCC, and put forward the prospects for the therapeutic targets of HNSCC.
Collapse
Affiliation(s)
- Yuxi Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
5
|
Expression of concern: Reduced CTGF expression promotes cell growth, migration, and invasion in nasopharyngeal carcinoma. PLoS One 2020; 15:e0231520. [PMID: 32240269 PMCID: PMC7117724 DOI: 10.1371/journal.pone.0231520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
6
|
Lin Q, Wang H, Lin X, Zhang W, Huang S, Zheng Y. PTPN12 Affects Nasopharyngeal Carcinoma Cell Proliferation and Migration Through Regulating EGFR. Cancer Biother Radiopharm 2018; 33:60-64. [PMID: 29634414 DOI: 10.1089/cbr.2017.2254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Nasopharyngeal carcinoma (NPC) shows the leading morbidity in otorhinolaryngological malignant tumor. It is a common malignancy in China with obvious reginal distribution. NPC is a polygenic disease that is affected by numerous factors. Protein tyrosine phosphatase nonreceptor type 12 (PTPN12) regulates multiple tumor proliferation and development, including breast cancer and colon cancer. However, the role of PTPN12 in NPC occurrence and development has not been elucidated. PATIENTS AND METHODS NPC cell line CNE2 was cultured in vitro and divided into three groups, including control, empty plasmid, and PTPN12 groups. PTPN12 mRNA and protein expressions were tested by real-time polymerase chain reaction and Western blot. CNE2 cell proliferation was detected by MTT assay. Cell migration was determined by wound healing assay. Cell apoptosis was evaluated by caspase 3 activity detection. Epidermal growth factor receptor (EGFR) expression was assessed by Western blot. RESULTS PTPN12 plasmid transfection increased PTPN12 mRNA and protein expressions, suppressed cell proliferation and migration, reduced EGFR level, and enhanced caspase 3 activity compared with control and empty plasmid groups (p < 0.05). CONCLUSIONS PTPN12 regulates NPC proliferation and migration through negative regulating EGFR. It could be treated as a molecular target for NPC diagnosis and prognosis analysis.
Collapse
Affiliation(s)
- Qinghai Lin
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Huige Wang
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Xinqiang Lin
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Wenrui Zhang
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Shuhua Huang
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Yandan Zheng
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| |
Collapse
|
7
|
Peng Y, He G, Tang D, Xiong L, Wen Y, Miao X, Hong Z, Yao H, Chen C, Yan S, Lu L, Yang Y, Li Q, Deng X. Lovastatin Inhibits Cancer Stem Cells and Sensitizes to Chemo- and Photodynamic Therapy in Nasopharyngeal Carcinoma. J Cancer 2017; 8:1655-1664. [PMID: 28775785 PMCID: PMC5535721 DOI: 10.7150/jca.19100] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus-associated malignancy occurring at high incidence in Southeast Asia and southern China. In spite of the good response to radio- and chemo-therapy at the early stage, resistance and recurrence develop in NPC patients in the advanced setting. Cancer stem cells (CSCs) play an important role in drug resistance and cancer recurrence. Here we report that lovastatin, a natural compound and a lipophilic statin that has already been used in the clinic to treat hypercholesterolemia, inhibited the CSC properties and induced apoptosis and cell cycle arrest in sphere-forming cells derived from the 5-8F and 6-10B NPC cell lines. Furthermore, lovastatin conferred enhanced sensitivity to the chemotherapeutic and photodynamic agents in NPC CSCs. Together our findings suggest that targeting CSCs by lovastatin in combination with routine chemotherapeutic drugs or photodynamic therapy might be a promising approach to the treatment of NPC.
Collapse
Affiliation(s)
- Yikun Peng
- Department of Otorhinolaryngology-Head and Neck Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Guangchun He
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Da Tang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiongying Miao
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hui Yao
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Chao Chen
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Shichao Yan
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Lu Lu
- Medical College, Hunan Normal University, Changsha, Hunan, China
| | - Yingke Yang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Qinglong Li
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiyun Deng
- Medical College, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
8
|
Deng J, Zhang W, Liu S, An H, Tan L, Ma L. LATS1 suppresses proliferation and invasion of cervical cancer. Mol Med Rep 2017; 15:1654-1660. [PMID: 28259899 PMCID: PMC5364969 DOI: 10.3892/mmr.2017.6180] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/03/2016] [Indexed: 01/15/2023] Open
Abstract
Loss of large tumor suppressor kinase 1 (LATS1)Y has been implicated in numerous types of human cancer. However, its involvement in human cervical cancer remains to be elucidated. The present study aimed to investigate the clinical significance and biological characteristics of LATS1 in human cervical cancer. The present study investigated the protein expression levels of LATS1 in tissues from 80 cases of cervical cancer using immunohistochemistry and demonstrated that LATS1 was downregulated in 45% (36/80) of cervical cancers. Transfection of LATS1 was performed in the SiHa cell line and LATS1 siRNA knockdown was performed in the Caski cell line. MTT assay and Matrigel invasion assay indicated that LATS1 overexpression inhibited cell proliferation and invasion. LATS1 overexpression upregulated p27 expression, and downregulated the expression of cyclin E and matrix metalloproteinase 9. In addition, LATS1 overexpression stimulated yes-associated protein 1 (YAP) phosphorylation. Depletion of LATS1 in Caski cells resulted in the opposite effects. The current study demonstrated that LATS1 was downregulated in cervical cancer and may suppress cell growth and invasion through regulating the expression of cyclin E, p27, MMP9 and YAP.
Collapse
Affiliation(s)
- Jihong Deng
- Department of Gynecology, Kunming Maternity and Child Care Hospital, Kunming, Yunnan 650000, P.R. China
| | - Wen Zhang
- Department of Gynecology, Kunming Maternity and Child Care Hospital, Kunming, Yunnan 650000, P.R. China
| | - Shuangyue Liu
- Department of Gynecology, Kunming Maternity and Child Care Hospital, Kunming, Yunnan 650000, P.R. China
| | - Hongmei An
- Department of Gynecology, Kunming Maternity and Child Care Hospital, Kunming, Yunnan 650000, P.R. China
| | - Lu Tan
- Department of Gynecology, Kunming Maternity and Child Care Hospital, Kunming, Yunnan 650000, P.R. China
| | - Lisha Ma
- Department of Gynecology, Kunming Maternity and Child Care Hospital, Kunming, Yunnan 650000, P.R. China
| |
Collapse
|
9
|
Zhan P, Zhao S, Yan H, Yin C, Xiao Y, Wang Y, Ni R, Chen W, Wei G, Zhang P. α-enolase promotes tumorigenesis and metastasis via regulating AMPK/mTOR pathway in colorectal cancer. Mol Carcinog 2017; 56:1427-1437. [PMID: 27996156 DOI: 10.1002/mc.22603] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/07/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022]
Abstract
The α-enolase (ENO1) plays pivotal roles in several types of cancer, but its clinical significance, functional role, and possible mechanism in colorectal cancer (CRC) have remained unclear. Expression level of ENO1 in CRC tissues was examined by qRT-PCR, Western blot, and immunohistochemistry. The effects of ENO1 on cell growth were investigated by MTT, colony formation, flow cytometry assays, and in vivo tumorigenic capacity analysis. The impacts of ENO1 on cell migration and invasion were also explored by scratch-healing, Transwell or Matrigel chamber assays, and in vivo metastatic capacity analysis. Our results showed that the expression level of ENO1 was significantly elevated in CRC tissues. High expression level of ENO1 was associated with disease progression in CRC patients. Overexpression of ENO1 in HCT116 cell line promoted cell proliferation, migration, and invasion in vitro as well as tumorigenesis and metastasis in vivo. In other hand, ablation of ENO1 in HCT116 cells led to totally reverse effects. Mechanistically, we revealed ENO1 could regulate AMPK/mTOR signaling pathway. AMPK pathway activation or mTOR pathway suppression blocked these ENO1 induced alterations. Together, our results demonstrated that ENO1 is a potent promoter of CRC genesis and metastasis at least in part though regulating AMPK/mTOR pathway. These findings also suggested that ENO1 may be a promising therapeutic target in CRC patients.
Collapse
Affiliation(s)
- Panpan Zhan
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, P.R. China.,Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center/Cancer Hospital, Beijing, P.R. China
| | - Shihu Zhao
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Hua Yan
- Department of Spleen and Stomach Diseases, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| | - Chunli Yin
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Yi Xiao
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Yunshan Wang
- Department of Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Ruoxuan Ni
- Department of Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Guangwei Wei
- Department of Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Pengju Zhang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, P.R. China
| |
Collapse
|
10
|
The pivotal role of CCN2 in mammalian palatogenesis. J Cell Commun Signal 2016; 11:25-37. [PMID: 27761803 DOI: 10.1007/s12079-016-0360-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/15/2016] [Indexed: 01/25/2023] Open
Abstract
Mammalian palatogenesis is a complex process involving a temporally and spatially regulated myriad of factors. Together these factors control the 3 vital processes of proliferation, elevation and fusion of the developing palate. In this study, we show for the first time the unequivocally vital role of CCN2 in development of the mammalian palate. We utilized CCN2 knockout (KO) mice and cranial neural crest derived mesenchymal cells from these CCN2 KO mice to investigate the 3 processes crucial to normal palatogenesis. Similar to previously published reports, the absence of CCN2 inhibits proliferation of cells in the palate specifically at the G1/S transition. Absence of CCN2 also inhibited palatal shelf elevation from the vertical to horizontal position. CCN2 KO mesenchymal cells demonstrated deficiencies in adhesion and spreading owing to an inability to activate Rac1 and RhoA. On the contrary, CCN2 KO mesenchymal cells exhibited increased rates of migration compared to WT cells. The addition of exogenous CCN2 to KO mesenchymal cells restored their ability to spread normally on fibronectin. Finally, utilizing an organ culture model we show that the palatal shelves of the CCN2 KO mice demonstrate an inability to fuse when apposed. Together, these data signify that CCN2 plays an indispensible role in normal development of the mammalian palate and warrants additional studies to determine the precise mechanism(s) responsible for these effects.
Collapse
|
11
|
Chen S, Wu H, Lv N, Wang H, Wang Y, Tang Q, Shao H, Sun C. LncRNA CCAT2 predicts poor prognosis and regulates growth and metastasis in small cell lung cancer. Biomed Pharmacother 2016; 82:583-8. [DOI: 10.1016/j.biopha.2016.05.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 01/29/2023] Open
|
12
|
Drzewiecka H, Gałęcki B, Jarmołowska-Jurczyszyn D, Kluk A, Dyszkiewicz W, Jagodziński PP. Decreased expression of connective tissue growth factor in non-small cell lung cancer is associated with clinicopathological variables and can be restored by epigenetic modifiers. J Cancer Res Clin Oncol 2016; 142:1927-46. [PMID: 27393180 PMCID: PMC4978771 DOI: 10.1007/s00432-016-2195-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 06/18/2016] [Indexed: 01/27/2023]
Abstract
Purpose Recent studies indicated undisputed contribution of connective tissue growth factor (CTGF) in the development of many cancers, including non-small cell lung cancer (NSCLC). However, the functional role and regulation of CTGF expression during tumorigenesis remain elusive. Our goal was to determine CTGF transcript and protein levels in tumoral and matched control tissues from 98 NSCLC patients, to correlate the results with clinicopathological features and to investigate whether the CTGF expression can be epigenetically regulated in NSCLC. Methods We used quantitative PCR, Western blotting and immunohistochemistry to evaluate CTGF expression in lung cancerous and histopathologically unchanged tissues. We tested the impact of 5-Aza-2′-deoxycytidine (5-dAzaC) and trichostatin A (TSA) on CTGF transcript and protein levels in NSCLC cells (A549, Calu-1). DNA methylation status of the CTGF regulatory region was evaluated by bisulfite sequencing. The influence of 5-dAzaC and TSA on NSCLC cells viability and proliferation was monitored by the trypan blue assay. Results We found significantly decreased levels of CTGF mRNA and protein (both p < 0.0000001) in cancerous tissues of NSCLC patients. Down-regulation of CTGF occurred regardless of gender in all histological subtypes of NSCLC. Moreover, we showed that 5-dAzaC and TSA were able to restore CTGF mRNA and protein contents in NSCLC cells. However, no methylation within CTGF regulatory region was detected. Both compounds significantly reduced NSCLC cells proliferation. Conclusions Decreased expression of CTGF is a common feature in NSCLC; however, it can be restored by the chromatin-modifying agents such as 5-dAzaC or TSA and consequently restrain cancer development. Electronic supplementary material The online version of this article (doi:10.1007/s00432-016-2195-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hanna Drzewiecka
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznan, Poland.
| | - Bartłomiej Gałęcki
- Department of Thoracic Surgery, Poznan University of Medical Sciences, Szamarzewskiego 62 Street, 60-569, Poznan, Poland
| | - Donata Jarmołowska-Jurczyszyn
- Department of Clinical Pathomorphology, Poznan University of Medical Sciences, Przybyszewskiego 49 Street, 60-355, Poznan, Poland
| | - Andrzej Kluk
- Department of Clinical Pathomorphology, Poznan University of Medical Sciences, Przybyszewskiego 49 Street, 60-355, Poznan, Poland
| | - Wojciech Dyszkiewicz
- Department of Thoracic Surgery, Poznan University of Medical Sciences, Szamarzewskiego 62 Street, 60-569, Poznan, Poland
| | - Paweł P Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznan, Poland
| |
Collapse
|
13
|
Talar B, Gajos-Michniewicz A, Talar M, Chouaib S, Czyz M. Pentoxifylline Inhibits WNT Signalling in β-Cateninhigh Patient-Derived Melanoma Cell Populations. PLoS One 2016; 11:e0158275. [PMID: 27351373 PMCID: PMC4924839 DOI: 10.1371/journal.pone.0158275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/13/2016] [Indexed: 11/18/2022] Open
Abstract
Background The heterogeneity of melanoma needs to be addressed and combination therapies seem to be necessary to overcome intrinsic and acquired resistance to newly developed immunotherapies and targeted therapies. Although the role of WNT/β-catenin pathway in melanoma was early demonstrated, its contribution to the lack of the melanoma patient response to treatment was only recently recognized. Using patient-derived melanoma cell populations, we investigated the influence of pentoxifylline on melanoma cells with either high or low expression of β-catenin. Findings Our results indicate that pentoxifylline inhibits the activity of the canonical WNT pathway in melanoma cell populations with high basal activity of this signalling. This is supported by lowered overall activity of transcription factors TCF/LEF and reduced nuclear localisation of active β-catenin. Moreover, treatment of β-cateninhigh melanoma cell populations with pentoxifylline induces downregulation of genes that are targets of the WNT/β-catenin pathway including connective tissue growth factor (CTGF) and microphthalmia-associated transcription factor (MITF-M), a melanocyte- and melanoma cell-specific regulator. Conclusions These results suggest that pentoxifylline, a drug approved by the FDA in the treatment of peripheral arterial disease, might be tested in a subset of melanoma patients with elevated activity of β-catenin. This pharmaceutical might be tested as an adjuvant drug in combination therapies when the response to immunotherapy is prevented by high activity of the WNT/β-catenin pathway.
Collapse
Affiliation(s)
- Beata Talar
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
| | | | - Marcin Talar
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | - Salem Chouaib
- Unité INSERM U1186, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
- * E-mail:
| |
Collapse
|
14
|
Dikshit A, Gao C, Small C, Hales K, Hales DB. Flaxseed and its components differentially affect estrogen targets in pre-neoplastic hen ovaries. J Steroid Biochem Mol Biol 2016; 159:73-85. [PMID: 26925929 PMCID: PMC4821676 DOI: 10.1016/j.jsbmb.2016.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/12/2016] [Accepted: 02/24/2016] [Indexed: 02/07/2023]
Abstract
Flaxseed has been studied for decades for its health benefits that include anti-cancer, cardio-protective, anti-diabetic, anti-inflammatory properties. The biologically active components that mediate these effects are the omega-3 fatty acids and the lignan, secoisolariciresinol diglucoside. We have previously shown that whole flaxseed supplemented diet decreases the severity and incidence of ovarian cancer while a 15% dose of flaxseed is most protective against inflammation and estrogen-induced chemical and genotoxicity. The objective of this study was to dissect the independent effects of the two flaxseed components on estrogen signaling and metabolism. Two and half year old hens were fed either a control diet, 15% whole flaxseed diet, defatted flax meal diet or 5% flax oil diet for 3 months after which the animals were sacrificed and blood and tissues were harvested. Whole flaxseed diet caused a decrease in expression of ERα. ERα target gene expression was assessed using RT(2) profiler PCR array. Some targets involved in the IGF/insulin signaling pathway (IRS1, IGFBP4, IGFBP5) were downregulated by flaxseed and its components. Flaxseed diet also downregulated AKT expression. A number of targets related to NF-kB signaling were altered by flaxseed diet including a series of targets implicated in cancer. Whole flaxseed diet also affected E2 metabolism by increasing CYP1A1 expression with a corresponding increase in the onco-protective E2 metabolite, 2-methoxyestradiol. The weak anti-estrogens, enterolactone, enterodiol and 2-methoxyestradiol, might be working synergistically to generate a protective effect on the ovaries from hens on whole flaxseed diet by altering the estrogen signaling and metabolism.
Collapse
Affiliation(s)
- Anushka Dikshit
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Chunqi Gao
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Carrie Small
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Karen Hales
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | - Dale Buchanan Hales
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA.
| |
Collapse
|
15
|
Huang X, Li Y, Xu J, Liu K, Yu X, Cheng X, Xu D, Li Z. Restoration of murine femoral segmental defect using CTGF-overexpressing MC3T3-E1 cells. Am J Transl Res 2016; 8:1530-1540. [PMID: 27186279 PMCID: PMC4859638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 02/17/2016] [Indexed: 06/05/2023]
Abstract
Connective tissue growth factor (CTGF) is a member of the CCN super family and is reported to widely participate in bone development and regeneration. This study aimed to restore murine femoral segmental defect using CTGF-overexpressing MC3T3-E1 cells. MC3T3-E1 cells were transinfected by lenti-CTGF (LvCTGF) and lenti-negative control (LvNC) virus to obtain stably transinfected cells. Real-time PCR, Western blot, alkaline phosphatase activity assay, and alizarin red staining demonstrated that the overexpression of CTGF enhanced osteogenesis in vitro. Cell migration assay results showed that LvCTGF cells expressed higher migration ability than LvNC cells, while CCK-8 assay revealed no significant difference in cell proliferation. The LvCTGF and LvNC cells were then seeded into a chitosan/β-TCP scaffold and were used to restore a murine femoral segmental defect. Samples were harvested by the end of 2 and 5 weeks respectively. Micro-CT analysis and Masson's trichrome staining results showed that the LvCTGF-scaffold group expressed better bone healing compared with the LvNC-scaffold and scaffold-only groups. CTGF-overexpressed cells serve as an efficient source of seeding cells for bone regeneration.
Collapse
Affiliation(s)
- Xiangyu Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan UniversityWuhan, China
| | - Yanqiu Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan UniversityWuhan, China
| | - Jiantao Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan UniversityWuhan, China
| | - Kai Liu
- Lishui UniversityLishui, China
| | - Xin Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan UniversityWuhan, China
| | - Xin Cheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan UniversityWuhan, China
| | - Dongdong Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan UniversityWuhan, China
| | - Zubing Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan UniversityWuhan, China
- Department of Oral and Maxillofacial Trauma and Plastic Aesthetic Surgery, School & Hospital of Stomatology, Wuhan UniversityWuhan, China
| |
Collapse
|
16
|
Abstract
High-mobility group nucleosome-binding domain 5 (HMGN5) is a new member of the high-mobility group N (HMGN) protein family that is involved in nucleosomal binding and transcriptional activation. It was first discovered in mouse, and recent studies found that the expressions of HMGN5 in many human cancers were also highly regulated, such as prostate, bladder, breast, and lung and clear cell renal cell carcinoma. Numerous reports have demonstrated that HMGN5 plays significant roles in many biological and pathological conditions, such as in developmental defects, hypersensitivity to stress, embryonic stem cell differentiation, and tumor progression. Importantly, deficiency of HMGN5 has been shown to be linked to cancer cell growth, cell cycle regulation, migration, invasion, and clinical outcomes, and it represents a promising therapeutic target for many malignant tumors. In the present review, we provide an overview of the current knowledge concerning the role of HMGN5 in cancer development and progression.
Collapse
|
17
|
Yang S, Long M, Tachado SD, Seng S. Cigarette smoke modulates PC3 prostate cancer cell migration by altering adhesion molecules and the extracellular matrix. Mol Med Rep 2015; 12:6990-6. [PMID: 26351771 PMCID: PMC4626126 DOI: 10.3892/mmr.2015.4302] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 01/07/2015] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related mortality among American males. Studies suggest that cigarette smoking is associated with the progression of PCa; however, the molecular mechanisms underlying this process have not been extensively investigated. PCa progression is characterized by increased cell migration and alterations in extracellular matrix (ECM)- and cell adhesion molecule (CAM)-related gene expression. In the present study, the influence of cigarette smoke medium (SM) on cell migration and on the expression of ECM- and CAM-related genes in PC3 prostate adenocarcinoma cells was investigated. According to a wound-healing assay, SM treatment promoted PC3 cell migration. RNA expression levels from SM-treated and control cells were analyzed using a polymerase chain reaction (PCR) array. Of 84 genes analyzed, 27.38% (23/84) exhibited a ≥2-fold change in threshold cycle in PC3 cells following 0.5% SM treatment. Functional gene grouping analysis demonstrated that SM treatment modulated the RNA transcription of approximately 18.4% of CAMs and 33.93% of ECM-related genes. Quantitative PCR analysis showed that SM treatment led to a significant decrease in transcription levels of the following genes: Collagen 5 α-1(V), connective tissue growth factor, integrin β-2, kallmann syndrome 1, laminin α 3, matrix metallopeptidase 7 (MMP7), MMP13, secreted protein acidic cysteine-rich, thrombospondin-2 and versican; and that SM significantly increased the transcription levels of MMP2 and MMP12. Furthermore, MMP2 knockdown significantly reduced the migration of SM-treated PC3 cells. The present study provides novel insights into the association of cigarette smoking with PCa progression, via the alteration of ECM/CAM interactions.
Collapse
Affiliation(s)
- Suping Yang
- Division of Experimental Medicine, Critical Care and Sleep Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Minica Long
- Division of Experimental Medicine, Critical Care and Sleep Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Souvenir D Tachado
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Seyha Seng
- Division of Experimental Medicine, Critical Care and Sleep Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
18
|
Tsai HC, Su HL, Huang CY, Fong YC, Hsu CJ, Tang CH. CTGF increases matrix metalloproteinases expression and subsequently promotes tumor metastasis in human osteosarcoma through down-regulating miR-519d. Oncotarget 2015; 5:3800-12. [PMID: 25003330 PMCID: PMC4116521 DOI: 10.18632/oncotarget.1998] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Osteosarcoma, the most common primary malignant bone tumor, shows potent capacity for local invasion and distant metastasis. Connective tissue growth factor (CTGF/CCN2), a secreted protein, binds to integrins, modulates invasive behavior of certain human cancer cells. Effect of CTGF in metastasis of human osteosarcoma is unknown. We found overexpression of CTGF increasing matrix metalloproteinases (MMPs)-2 and MMP-3 expression as well as promoting cell migration. MicroRNA (miRNA) analysis of CTGF-overexpressed osteosarcoma versus control cells probed mechanisms of CTGF-mediated promotion of migration. Among miRNAs regulated by CTGF, miR-519d was most downregulated after CTGF treatment. Co-transfection with miR-519d mimic reversed CTGF-mediated MMPs expression and cell migration. Also, MEK and ERK inhibitors or mutants reduced CTGF-increased cell migration and miR-519d suppression. By contrast, knockdown of CTGF diminished lung metastasis in vivo. Clinical samples indicate CTGF expression as linked with clinical stage and tumor metastasis. Taken together, data show CTGF elevating MMPs expression and subsequently promoting tumor metastasis in human osteosarcoma, down-regulating miR-519d via MEK and ERK pathways, making CTGF a new molecular therapeutic target in osteosarcoma metastasis.
Collapse
Affiliation(s)
- Hsiao-Chi Tsai
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | | | | | | | | | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
19
|
Wang J, Hu JL, Cao RB, Ding Q, Peng G, Fei SJ, Jiang Y, Li PC, Yang KY, Zhang WJ, Wu G, Wang RZ, Li PD. Small hairpin RNA-mediated Krüppel-like factor 8 gene knockdown inhibits invasion of nasopharyngeal carcinoma. Oncol Lett 2015; 9:2515-2519. [PMID: 26137099 DOI: 10.3892/ol.2015.3099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 01/29/2015] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to characterize the expression of Krüppel-like factor 8 (KLF8) in nasopahryngeal carcinoma (NPC) cell lines and determine its effect on tumor development and invasion following KLF8 gene knockdown by small hairpin RNA (shRNA). KLF8 expression in four NPC cell lines was examined by quantitative polymerase chain reaction (qPCR) and western blotting. KLF8 was knocked down in the SUNE1-5-8F/Sh-KLF8 cell line using shRNA, and the resulting stable cell line SUNE1-5-8F-sh-KLF8 was transplanted into nude mice in order to observe tumor formation and invasion. The results obtained from qPCR and western blotting revealed that, of the four NPC cell lines, KLF8 expression was lowest in the CNE-1 cells and highest in the SUNE1-5-8F cells. The tumor xenograft mouse models revealed that SUNE1-5-8F/Sh-KLF8 cells had a reduced ability for tumor formation and invasion compared with the control group. These results demonstrated for the first time that KLF8 modulates the formation and invasive ability of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Jing Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Jian Li Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Ru Bo Cao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Qian Ding
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Gang Peng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Shi Jiang Fei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Yao Jiang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Peng Cheng Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Kun Yu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Wen Jie Zhang
- Department of Pathology, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| | - Ruo Zheng Wang
- Department of Radiation Oncology, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Pin Dong Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430023, P.R. China
| |
Collapse
|
20
|
Fu QF, Liu Y, Fan Y, Hua SN, Qu HY, Dong SW, Li RL, Zhao MY, Zhen Y, Yu XL, Chen YY, Luo RC, Li R, Li LB, Deng XJ, Fang WY, Liu Z, Song X. Alpha-enolase promotes cell glycolysis, growth, migration, and invasion in non-small cell lung cancer through FAK-mediated PI3K/AKT pathway. J Hematol Oncol 2015; 8:22. [PMID: 25887760 PMCID: PMC4359783 DOI: 10.1186/s13045-015-0117-5] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/09/2015] [Indexed: 02/04/2023] Open
Abstract
Background During tumor formation and expansion, increasing glucose metabolism is necessary for unrestricted growth of tumor cells. Expression of key glycolytic enzyme alpha-enolase (ENO1) is controversial and its modulatory mechanisms are still unclear in non-small cell lung cancer (NSCLC). Methods The expression of ENO1 was examined in NSCLC and non-cancerous lung tissues, NSCLC cell lines, and immortalized human bronchial epithelial cell (HBE) by quantitative real-time reverse transcription PCR (qRT-PCR), immunohistochemistry, and Western blot, respectively. The effects and modulatory mechanisms of ENO1 on cell glycolysis, growth, migration, invasion, and in vivo tumorigenesis and metastasis in nude mice were also analyzed. Results ENO1 expression was increased in NSCLC tissues in comparison to non-cancerous lung tissues. Similarly, NSCLC cell lines A549 and SPCA-1 also express higher ENO1 than HBE cell line in both mRNA and protein levels. Overexpressed ENO1 significantly elevated NSCLC cell glycolysis, proliferation, clone formation, migration, and invasion in vitro, as well as tumorigenesis and metastasis in vivo by regulating the expression of glycolysis, cell cycle, and epithelial-mesenchymal transition (EMT)-associated genes. Conversely, ENO1 knockdown reversed these effects. More importantly, our further study revealed that stably upregulated ENO1 activated FAK/PI3K/AKT and its downstream signals to regulate the glycolysis, cell cycle, and EMT-associated genes. Conclusion This study showed that ENO1 is responsible for NSCLC proliferation and metastasis; thus, ENO1 might serve as a potential molecular therapeutic target for NSCLC treatment. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0117-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiao-Fen Fu
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Yan Liu
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Yue Fan
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Sheng-Ni Hua
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Hong-Ying Qu
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Su-Wei Dong
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Rui-Lei Li
- Department of Cancer Biotherapy Center, Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, People's Republic China.
| | - Meng-Yang Zhao
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Yan Zhen
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Xiao-Li Yu
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Yi-Yu Chen
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Rong-Cheng Luo
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Rong Li
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Li-Bo Li
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Xiao-Jie Deng
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Wei-Yi Fang
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Zhen Liu
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Department of Pathology, Basic School of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Xin Song
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Department of Cancer Biotherapy Center, Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, People's Republic China.
| |
Collapse
|
21
|
Kubota S, Maeda-Uematsu A, Nishida T, Takigawa M. New functional aspects of CCN2 revealed by trans-omic approaches. J Oral Biosci 2015. [DOI: 10.1016/j.job.2014.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Zeng H, Zhang Y, Yi Q, Wu Y, Wan R, Tang L. CRIM1, a newfound cancer-related player, regulates the adhesion and migration of lung cancer cells. Growth Factors 2015; 33:384-92. [PMID: 26653968 DOI: 10.3109/08977194.2015.1119132] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
CRIM1 is a member of the bone morphogenetic protein (BMP) antagonists; however, the role of CRIM1 in controlling cancer cell behavior remains unknown. This study investigated its function in the A549 cell line in vitro. The results show that treating cells with CRIM1 peptide could increase the migration and adhesion of A549. Consistently, silencing the CRIM1 expression decreased the migration and adhesion of A549. Furthermore, the CRIM1 protein expression was increased in A549 which were treated with transforming growth factor beta 1 to induced EMT. However, CRIM1 peptide treatment could increase the expression of N-CAD and E-CAD expression. Finally, overexpression of the oncogene YAP1 resulted in an up-regulation of the CRIM1 expression in A549, suggesting that CRIM1 was a target of the Hippo pathway. These observations provide evidence for the first time that CRIM1 plays a role in cancer cells by enhancing the migration and adhesion and increasing the expression of N-CAD and E-CAD.
Collapse
Affiliation(s)
- Hui Zeng
- a Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University , Chongqing , China
| | - Yi Zhang
- a Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University , Chongqing , China
| | - Qian Yi
- a Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University , Chongqing , China
| | - Yanjiao Wu
- a Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University , Chongqing , China
| | - Rongxue Wan
- a Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University , Chongqing , China
| | - Liling Tang
- a Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University , Chongqing , China
| |
Collapse
|
23
|
Ahmad A, Askari S, Befekadu R, Hahn-Strömberg V. Investigating the association between polymorphisms in connective tissue growth factor and susceptibility to colon carcinoma. Mol Med Rep 2014; 11:2493-503. [PMID: 25502877 PMCID: PMC4337474 DOI: 10.3892/mmr.2014.3083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/25/2014] [Indexed: 01/01/2023] Open
Abstract
There have been numerous studies on the gene expression of connective tissue growth factor (CTGF) in colorectal cancer, however very few have investigated polymorphisms in this gene. The present study aimed to determine whether single nucleotide polymorphisms (SNPs) in the CTGF gene are associated with a higher susceptibility to colon cancer and/or an invasive tumor growth pattern. The CTGF gene was genotyped for seven SNPs (rs6918698, rs1931002, rs9493150, rs12526196, rs12527705, rs9399005 and rs12527379) by pyrosequencing. Formalin-fixed paraffin-embedded tissue samples (n=112) from patients diagnosed with colon carcinoma, and an equal number of blood samples from healthy controls, were selected for genomic DNA extraction. The complexity index was measured using images of tumor samples (n=64) stained for cytokeratin-8. The images were analyzed and correlated with the identified CTGF SNPs and clinicopathological parameters of the patients, including age, gender, tumor penetration, lymph node metastasis, systemic metastasis, differentiation and localization of tumor. It was demonstrated that the frequency of the SNP rs6918698 GG genotype was significantly associated (P=0.05) with an increased risk of colon cancer, as compared with the GC and CC genotypes. The other six SNPs (rs1931002, rs9493150, rs12526196, rs12527705, rs9399005 and rs12527379) exhibited no significant difference in the genotype and allele frequencies between patients diagnosed with colon carcinoma and the normal healthy population. A trend was observed between genotype variation at rs6918698 and the complexity index (P=0.052). The complexity index and genotypes for any of the studied SNPs were not significantly correlated with clinical or pathological parameters of the patients. These results indicate that the rs6918698 GG genotype is associated with an increased risk of developing colon carcinoma, and genetic variations at the rs6918698 are associated with the growth pattern of the tumor. The present results may facilitate the identification of potential biomarkers of the disease in addition to drug targets.
Collapse
Affiliation(s)
- Abrar Ahmad
- Department of Clinical Medicine, Örebro University, Örebro 701 81, Sweden
| | - Shlear Askari
- Department of Clinical Medicine, Örebro University, Örebro 701 81, Sweden
| | - Rahel Befekadu
- Department of Laboratory Medicine, Section for Transfusion Medicine, Örebro University Hospital, Örebro 701 85, Sweden
| | | |
Collapse
|
24
|
Su FR, Ding JH, Bo L, Liu XG. Chromodomain helicase/ATPase DNA binding protein 1-like protein expression predicts poor prognosis in nasopharyngeal carcinoma. Exp Ther Med 2014; 8:1745-1750. [PMID: 25371726 PMCID: PMC4217779 DOI: 10.3892/etm.2014.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 09/11/2014] [Indexed: 12/22/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignancy with a high metastatic ability. Recent studies have implicated the role of chromodomain helicase/ATPase DNA binding protein 1-like (CHD1L) gene as a novel oncogene; however, the functional role of CHD1L in NPC remains unknown. The aim of this study was to evaluate the clinical significance of CHD1L positivity in NPC. CHD1L protein expression was examined by performing western blot analysis of 30 fresh NPC tissues and conducting immunohistochemistry tests of 133 NPC samples between December 1, 2005 and December 1, 2009. The correlations of CHD1L expression status with clinicopathological features and prognosis were investigated. Immunohistochemical analysis showed that 88 of 133 (66.2%) paraffin-embedded NPC biopsies exhibited positive expression of CHD1L, but all non-cancerous nasopharyngeal specimens were negative for CHD1L expression. In addition, positive CHD1L expression was strongly associated with an advanced clinical stage (P=0.016), recurrence (P=0.002) and the metastasis status (P=0.031) of NPC. Kaplan-Meier survival analysis demonstrated that patients with CHD1L-positive NPC had significantly shorter overall survival (P<0.001). Furthermore, the multivariate analysis indicated that CHD1L protein expression was an independent prognostic factor for overall survival (hazard ratio, 7.916; 95% confidence interval, 2.067-16.034; P=0.003) in patients with NPC. These results indicate that CHD1L is a prognostic marker for NPC.
Collapse
Affiliation(s)
- Fa-Ren Su
- Department of Otorhinolaryngology and Head and Neck Surgery, Shandong People's Armed Police Corps Hospital, Jinan, Shandong 250014, P.R. China
| | - Jing-Hua Ding
- Department of Otorhinolaryngology and Head and Neck Surgery, Shandong People's Armed Police Corps Hospital, Jinan, Shandong 250014, P.R. China
| | - Lin Bo
- Department of Otorhinolaryngology and Head and Neck Surgery, Shandong People's Armed Police Corps Hospital, Jinan, Shandong 250014, P.R. China
| | - Xin-Gang Liu
- Department of Otorhinolaryngology and Head and Neck Surgery, Shandong People's Armed Police Corps Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
25
|
Cellular and molecular actions of CCN2/CTGF and its role under physiological and pathological conditions. Clin Sci (Lond) 2014; 128:181-96. [DOI: 10.1042/cs20140264] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CCN family protein 2 (CCN2), also widely known as connective tissue growth factor (CTGF), is one of the founding members of the CCN family of matricellular proteins. Extensive investigation on CCN2 over decades has revealed the novel molecular action and functional properties of this unique signalling modulator. By its interaction with multiple molecular counterparts, CCN2 yields highly diverse and context-dependent biological outcomes in a variety of microenvironments. Nowadays, CCN2 is recognized to conduct the harmonized development of relevant tissues, such as cartilage and bone, in the skeletal system, by manipulating extracellular signalling molecules involved therein by acting as a hub through a web. However, on the other hand, CCN2 occasionally plays profound roles in major human biological disorders, including fibrosis and malignancies in major organs and tissues, by modulating the actions of key molecules involved in these clinical entities. In this review, the physiological and pathological roles of this unique protein are comprehensively summarized from a molecular network-based viewpoint of CCN2 functionalities.
Collapse
|
26
|
A complex mechanism for HDGF-mediated cell growth, migration, invasion, and TMZ chemosensitivity in glioma. J Neurooncol 2014; 119:285-95. [PMID: 24986090 DOI: 10.1007/s11060-014-1512-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 06/17/2014] [Indexed: 01/15/2023]
Abstract
HDGF is overexpressed in gliomas as compared to normal brain. We therefore analyzed the molecular mechanisms of HDGF action in gliomas. HDGF was downregulated in normal brain tissue as compared to glioma specimens at both the mRNA and the protein levels. In glioma samples, increased HDGF expression was associated with disease progression. Knocking down HDGF expression not only significantly decreased cellular proliferation, migration, invasion, and tumorigenesis, but also markedly enhanced TMZ-induced cytotoxicity and apoptosis in glioma cells. Mechanistic analyses revealed that CCND1, c-myc, and TGF-β were downregulated after stable HDGF knockdown in the U251 and U87 glioma cells. HDGF knockdown restored E-cadherin expression and suppressed mesenchymal cell markers such as vimentin, β-catenin, and N-cadherin. The expression of cleaved caspase-3 increased, while Bcl-2 decreased in each cell line following treatment with shHDGF and TMZ, as compared to TMZ alone. Furthermore, RNAi-based knockdown study revealed that HDGF is probably involved in the activation of both the PI3K/Akt and the TGF-β signaling pathways. Together, our data suggested that HDGF regulates glioma cell growth, apoptosis and epithelial-mesenchymal transition (EMT) probably through the Akt and the TGF-β signaling pathways. These results provide evidence that targeting HDGF or its downstream targets may lead to novel therapies for gliomas.
Collapse
|
27
|
Chen S, Ma Z, Chen X, Zhang J. Prognostic significance of nemo-like kinase in nasopharyngeal carcinoma. Mol Med Rep 2014; 10:131-6. [PMID: 24789020 DOI: 10.3892/mmr.2014.2190] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 03/06/2014] [Indexed: 11/05/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a highly metastatic malignancy, which is highly prevalent in Southeast Asia and North Africa. Recent studies implicated the critical role of nemo‑like kinase (NLK) in tumor biology. However, the functional role of NLK in NPC has yet to be elucidated. In the present study, the significance of NLK positivity in NPC was examined. NLK expression was evaluated by immunohistochemistry in a relatively large sample of patients with NPC (n=352) from December 1, 2002 to December 1, 2009. The correlation between the NLK expression status and clinicopathological features and prognosis was investigated. Univariate and multivariate Cox regression models were developed to evaluate the association between the NLK status and the relative risks for relapse and mortality. In total, 54% (190/352) of NPC samples were identified as positive for NLK. By contrast, all 176 specimens of adjacent normal tissue were negative for NLK. NLK positivity was associated with tumor extent, regional lymph node status and distant metastases. A Kaplan‑Meier survival analysis demonstrated that patients with NLK‑positive NPC exhibited significantly shorter disease‑free survival (DFS) and overall survival (OS). Furthermore, Cox regression analysis revealed that NLK positivity was an unfavorable prognostic indicator of DFS and OS in NPC, independent of other features. Additionally, NLK‑positive patients with NPC without distant metastases were more likely to relapse compared with NLK‑negative patients with NPC without distant metastases. The present study indicates that NLK is a good prognostic marker for NPC.
Collapse
Affiliation(s)
- Size Chen
- Department of Oncology, The Affiliated Second Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Zhijian Ma
- Department of Radiation Oncology, Cancer Center of Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Xuemei Chen
- School of Public Health and Tropic Medicine, South Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jiren Zhang
- Department of Oncology, The Affiliated Second Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
28
|
Song Y, Luo Q, Long H, Hu Z, Que T, Zhang X, Li Z, Wang G, Yi L, Liu Z, Fang W, Qi S. Alpha-enolase as a potential cancer prognostic marker promotes cell growth, migration, and invasion in glioma. Mol Cancer 2014; 13:65. [PMID: 24650096 PMCID: PMC3994408 DOI: 10.1186/1476-4598-13-65] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 03/13/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The success of using glycolytic inhibitors for cancer treatment relies on better understanding the roles of each frequently deregulated glycolytic genes in cancer. This report analyzed the involvement of a key glycolytic enzyme, alpha-enolase (ENO1), in tumor progression and prognosis of human glioma. METHODS ENO1 expression levels were examined in glioma tissues and normal brain (NB) tissues. The molecular mechanisms of ENO1 expression and its effects on cell growth, migration and invasion were also explored by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay, Transwell chamber assay, Boyden chamber assay, Western blot and in vivo tumorigenesis in nude mice. RESULTS ENO1 mRNA and protein levels were upregulated in glioma tissues compared to NB. In addition, increased ENO1 was associated disease progression in glioma samples. Knocking down ENO1 expression not only significantly decreased cell proliferation, but also markedly inhibited cell migration and invasion as well as in vivo tumorigenesis. Mechanistic analyses revealed that Cyclin D1, Cyclin E1, pRb, and NF-κB were downregulated after stable ENO1 knockdown in glioma U251 and U87 cells. Conversely, knockdown of ENO1 resulted in restoration of E-cadherin expression and suppression of mesenchymal cell markers, such as Vimentin, Snail, N-Cadherin, β-Catenin and Slug. Furthermore, ENO1 suppression inactivated PI3K/Akt pathway regulating the cell growth and epithelial-mesenchymal transition (EMT) progression. CONCLUSION Overexpression of ENO1 is associated with glioma progression. Knockdown of ENO1 expression led to suppressed cell growth, migration and invasion progression by inactivating the PI3K/Akt pathway in glioma cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zhen Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| | | | | |
Collapse
|