1
|
Yang Y, Li S, Sun X, Zhang L, Chen M, Fu H. CCN1 secreted by human adipose-derived stem cells enhances wound healing and promotes angiogenesis through activating the AKT signalling pathway. Int Wound J 2023; 20:1667-1677. [PMID: 36541685 PMCID: PMC10088821 DOI: 10.1111/iwj.14028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
The study aimed to explore the role of cellular communication network factor 1 (CCN1) an extracellular matrix protein in hADSC-treated wound healing. Immunofluorescence and enzyme-linked immunosorbent assays (ELISA) were used to demonstrate the secretion of CCN1 by hADSCs, isolated from human fat tissue. We investigated the role of CCN1 in wound healing by knockdown of CCN1 expression in hADSCs using CCN1 siRNA. Conditioned medium of hADSCs or hADSCs with CCN1 knocked down (hADSC-CMCCN1↓ ) was collected. After treatment with plain DMEM/F12, hADSC-CM, hADSC-CMCCN1↓ , or recombinant human CCN1 (rhCCN1), the wound healing abilities of human umbilical vascular endothelial cells (HUVECs) were assayed, and the AKT, also known as protein kinase B (PKB), signalling pathway was detected using western blotting. Next, we created full-thickness skin wounds on the backs of the mice and different treatments were applied to the wound surface. Wound size was measured using a digital camera on days 0-10, and evaluated. H&E and immunohistochemical staining were performed, and laser Doppler perfusion imaging was used to evaluate blood perfusion. The wound model and wound-healing assay showed that the hADSCs-CM and rhCCN1 groups had enhanced wound healing compared to the hADSCs-CMCCN1↓ group. Further, CCN1 and hADSCs-CM promoted the proliferation and migration of HUVECs through the AKT signalling pathway. We concluded that CCN1 secreted by hADSCs enhances wound healing and promotes angiogenesis by activating the AKT signalling pathway. CCN1 plays a vital role in the regulation of hADSCs-CM during wound healing.
Collapse
Affiliation(s)
- Yi Yang
- Senior Department of Burns and Plastic SurgeryThe Fourth Medical Center of PLA General HospitalBeijingChina
- Chinese PLA Medical SchoolBeijingChina
| | - Shiyi Li
- Senior Department of Burns and Plastic SurgeryThe Fourth Medical Center of PLA General HospitalBeijingChina
- Chinese PLA Medical SchoolBeijingChina
| | - Xuer Sun
- Senior Department of Burns and Plastic SurgeryThe Fourth Medical Center of PLA General HospitalBeijingChina
- Chinese PLA Medical SchoolBeijingChina
| | - Lixia Zhang
- Senior Department of Burns and Plastic SurgeryThe Fourth Medical Center of PLA General HospitalBeijingChina
- Chinese PLA Medical SchoolBeijingChina
| | - Minliang Chen
- Senior Department of Burns and Plastic SurgeryThe Fourth Medical Center of PLA General HospitalBeijingChina
| | - Huijuan Fu
- Senior Department of Burns and Plastic SurgeryThe Fourth Medical Center of PLA General HospitalBeijingChina
| |
Collapse
|
2
|
Ketchen SE, Gamboa-Esteves FO, Lawler SE, Nowicki MO, Rohwedder A, Knipp S, Prior S, Short SC, Ladbury JE, Brüning-Richardson A. Drug Resistance in Glioma Cells Induced by a Mesenchymal-Amoeboid Migratory Switch. Biomedicines 2021; 10:9. [PMID: 35052688 PMCID: PMC8773151 DOI: 10.3390/biomedicines10010009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 11/27/2022] Open
Abstract
Cancer cell invasion is a precondition for tumour metastasis and represents one of the most devastating characteristics of cancer. The development of drugs targeting cell migration, known as migrastatics, may improve the treatment of highly invasive tumours such as glioblastoma (GBM). In this study, investigations into the role of the cell adhesion protein Cellular communication network factor 1 (CCN1, also known as CYR61) in GBM cell migration uncovered a drug resistance mechanism adopted by cells when treated with the small molecule inhibitor CCG-1423. This inhibitor binds to importin α/β inhibiting the nuclear translocation of the transcriptional co-activator MKL1, thus preventing downstream effects including migration. Despite this reported role as an inhibitor of cell migration, we found that CCG-1423 treatment did not inhibit GBM cell migration. However, we could observe cells now migrating by mesenchymal-amoeboid transition (MAT). Furthermore, we present evidence that CCN1 plays a critical role in the progression of GBM with increased expression in higher-grade tumours and matched blood samples. These findings support a potential role for CCN1 as a biomarker for the monitoring and potentially early prediction of GBM recurrence, therefore as such could help to improve treatment of and increase survival rates of this devastating disease.
Collapse
Affiliation(s)
- Sophie E. Ketchen
- Light Laboratories, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; (S.E.K.); (A.R.); (J.E.L.)
| | - Filomena O. Gamboa-Esteves
- Leeds Institute of Medical Research at St. James’s, University of Leeds, Leeds LS9 7TF, UK; (F.O.G.-E.); (S.C.S.)
| | - Sean E. Lawler
- Brown University Cancer Center, Pathology & Laboratory Medicine, Brown University, Providence, RI 02903, USA;
| | - Michal O. Nowicki
- Harvey Cushing Neuro-Oncology Research Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Arndt Rohwedder
- Light Laboratories, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; (S.E.K.); (A.R.); (J.E.L.)
| | - Sabine Knipp
- School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK; (S.K.); (S.P.)
| | - Sally Prior
- School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK; (S.K.); (S.P.)
| | - Susan C. Short
- Leeds Institute of Medical Research at St. James’s, University of Leeds, Leeds LS9 7TF, UK; (F.O.G.-E.); (S.C.S.)
| | - John E. Ladbury
- Light Laboratories, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; (S.E.K.); (A.R.); (J.E.L.)
| | - Anke Brüning-Richardson
- School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK; (S.K.); (S.P.)
| |
Collapse
|
3
|
Li D, Zhang Z, Xia C, Niu C, Zhou W. Non-Coding RNAs in Glioma Microenvironment and Angiogenesis. Front Mol Neurosci 2021; 14:763610. [PMID: 34803608 PMCID: PMC8595242 DOI: 10.3389/fnmol.2021.763610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/11/2021] [Indexed: 12/17/2022] Open
Abstract
Glioma, especially glioblastoma, is the most common and lethal brain tumor. In line with the complicated vascularization processes and the strong intratumoral heterogeneity, tumor-associated blood vessels in glioma are regulated by multiple types of cells through a variety of molecular mechanisms. Components of the tumor microenvironment, including tumor cells and tumor-associated stromata, produce various types of molecular mediators to regulate glioma angiogenesis. As critical regulatory molecules, non-coding RNAs (ncRNAs) inside cells or secreted to the tumor microenvironment play essential roles in glioma angiogenesis. In this review, we briefly summarize recent studies about the production, delivery, and functions of ncRNAs in the tumor microenvironment, as well as the molecular mechanisms underlying the regulation of angiogenesis by ncRNAs. We also discuss the ncRNA-based therapeutic strategies in the anti-angiogenic therapy for glioma treatment.
Collapse
Affiliation(s)
- Dongxue Li
- Intelligent Pathology Institute, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Department of Neurosurgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhe Zhang
- Basic Medical College, Qingdao University, Qingdao, China
| | - Chengyu Xia
- Department of Neurosurgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chaoshi Niu
- Department of Neurosurgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenchao Zhou
- Intelligent Pathology Institute, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Department of Pathology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
4
|
Li Y, Fan S, Xia W, Qiao B, Huang K, Zhou J, Liang M. MiR-181b suppresses angiogenesis by directly targeting cellular communication network factor 1. J Transl Med 2021; 101:1026-1035. [PMID: 33875791 DOI: 10.1038/s41374-021-00596-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 11/08/2022] Open
Abstract
Angiogenesis is essential for various physiological and pathological processes. Previous studies have shown that miRNAs play an important role in blood vessel development and angiogenesis. Recent studies have suggested that miR-181b might be involved in the regulation of angiogenesis in tumors. However, whether miR-181b plays a role in angiogenesis in nontumor diseases is unclear. We found that miR-181b expression was downregulated in hypoxia-stimulated primary human umbilical vein endothelial cells (HUVECs) and a mouse hindlimb ischemia (HLI) model. Gain- and loss-of-function studies showed that a miR-181b mimic inhibited HUVEC migration and tube formation in vitro, and a miR-181b inhibitor had the opposite effects. In vivo, agomir-181b suppressed perfusion recovery in the HLI model and capillary density in a Matrigel plug assay, while perfusion recovery and capillary density were increased by injection of antagomir-181b. Mechanistically, we showed with a reporter assay that cellular communication network factor 1 (CCN1) was a direct target of miR-181b. Moreover, miR-181b suppressed angiogenesis at least in part by targeting CCN1 to inhibit the AMPK signaling pathway. Our research suggests that miR-181b suppresses angiogenesis by directly targeting CCN1, which provides new clues for pro-angiogenic treatment strategies.
Collapse
Affiliation(s)
- Yue Li
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siyuan Fan
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weichang Xia
- Affiliated Renhe Hospital to China Three Gorges University, Yichang City, China
| | - Baoru Qiao
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jingqun Zhou
- Affiliated Renhe Hospital to China Three Gorges University, Yichang City, China.
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Abou-Kheir W, Mukherji D, Hadadeh O, Saleh E, Bahmad HF, Kanso M, Khalifeh M, Shamseddine A, Tamraz S, Jaafar R, Dagher C, Khalifeh I, Faraj W. CYR61/CCN1 expression in resected pancreatic ductal adenocarcinoma: A retrospective pilot study of the interaction between the tumors and their surrounding microenvironment. Heliyon 2020; 6:e03842. [PMID: 32395647 PMCID: PMC7205742 DOI: 10.1016/j.heliyon.2020.e03842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/10/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND CCN1 is an extracellular matrix-associated protein thought to be implicated in tumor-stromal interaction in several solid tumors. The aim of our pilot study was to evaluate the correlation between CCN1 expression in stromal cells, pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma cells in resected pancreatic ductal adenocarcinoma (PDAC) specimens, and correlate that clinically. METHODS A total of 42 paraffin-embedded PDAC tumor specimens were stained for CCN1 and evaluated via immunohistochemical (IHC) analysis. Statistical analysis was performed to correlate between CCN1 expression profiles in tumor tissues and clinicopathological parameters of patients. RESULTS Our results showed CCN1 (CYR61) gene was highly expressed in PDAC tissues relative to other organ specific tumor tissues. Also, moderate and overexpression of CCN1 in PanIN was associated with PanIN grade 3 tissues. A statistically significant association was found between PanIN CCN1 scores on one hand and cancer stage, cancer grade, and CCN1 expression among ductal tumor cells and adjacent stromal cells on the other hand. DISCUSSION The associations demonstrated suggest that CCN1 might be contributing to a substantial role in the interaction between the pancreatic tumors on one hand and their surrounding microenvironment and their precursors on the other hand; hence, it might serve as a potential therapeutic target for PDAC.
Collapse
Affiliation(s)
- Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Deborah Mukherji
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ola Hadadeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Eman Saleh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hisham F. Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mariam Kanso
- Department of Surgery, Division of General Surgery, Liver Transplantation and Hepatopancreaticobiliary (HPB) Unit, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohamad Khalifeh
- Department of Surgery, Division of General Surgery, Liver Transplantation and Hepatopancreaticobiliary (HPB) Unit, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Sally Tamraz
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rola Jaafar
- Department of Surgery, Division of General Surgery, Liver Transplantation and Hepatopancreaticobiliary (HPB) Unit, American University of Beirut Medical Center, Beirut, Lebanon
| | - Christelle Dagher
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ibrahim Khalifeh
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Walid Faraj
- Department of Surgery, Division of General Surgery, Liver Transplantation and Hepatopancreaticobiliary (HPB) Unit, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
6
|
Li SZ, Hu YY, Zhao JL, Zang J, Fei Z, Han H, Qin HY. Downregulation of FHL1 protein in glioma inhibits tumor growth through PI3K/AKT signaling. Oncol Lett 2020; 19:3781-3788. [PMID: 32382330 PMCID: PMC7202308 DOI: 10.3892/ol.2020.11476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 01/27/2020] [Indexed: 01/29/2023] Open
Abstract
Human four-and-a-half LIM domains protein 1 (FHL1) is a member of the FHL protein family, which serves an important role in multiple cellular events by interacting with transcription factors using its cysteine-rich zinc finger motifs. A previous study indicated that FHL1 was downregulated in several types of human cancer and served a role as a tumor suppressive gene. The overexpression of FHL1 inhibited tumor cell proliferation. However, to the best of our knowledge, there is no evidence to confirm whether FHL1 affected glioma growth, and the molecular mechanisms through which FHL1 represses tumor development remain unclear. In the present study, the expression level of FHL1 was determined using immunohistochemical staining in 114 tumor specimens from patients with glioma. The results indicated that FHL1 expression was negatively associated with the pathological grade of gliomas. Furthermore, Kaplan-Meier survival curves demonstrated that the patients with an increased FHL1 expression exhibited a significantly longer survival time, suggesting that FHL1 may be a prognostic marker for glioma. The protein level of FHL1 was relatively increased in the U251 glioma cell line compared with that in the U87 cell line. Therefore, FHL1 was knocked down in U251 by siRNA and overexpressed in U87, and it was identified that FHL1 significantly decreased the activation of PI3K/AKT signaling by interacting with AKT. Further experiments verified that FHL1 inhibited the growth of gliomas in vivo by modulating PI3K/AKT signaling. In conclusion, the results of the present study demonstrated that FHL1 suppressed glioma development through PI3K/AKT signaling.
Collapse
Affiliation(s)
- San-Zhong Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yi-Yang Hu
- Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jun-Long Zhao
- Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China.,Department of Biochemistry and Molecular Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jian Zang
- Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China.,Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hua Han
- Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China.,Department of Biochemistry and Molecular Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hong-Yan Qin
- Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
7
|
Gerarduzzi C, Hartmann U, Leask A, Drobetsky E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res 2020; 80:2705-2717. [PMID: 32193287 DOI: 10.1158/0008-5472.can-18-2098] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2019] [Accepted: 03/17/2020] [Indexed: 11/16/2022]
Abstract
The extracellular matrix (ECM) surrounding cells is indispensable for regulating their behavior. The dynamics of ECM signaling are tightly controlled throughout growth and development. During tissue remodeling, matricellular proteins (MCP) are secreted into the ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins and cell-matrix interactions to influence normal cellular functions. In the tumor microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can support multiple hallmarks of carcinogenesis by interacting with various cellular components that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This realization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer, as well as effective and selective therapeutic targets.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada. .,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Elliot Drobetsky
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
8
|
Pu N, Gao S, Yin H, Li JA, Wu W, Fang Y, Zhang L, Rong Y, Xu X, Wang D, Kuang T, Jin D, Yu J, Lou W. Cell-intrinsic PD-1 promotes proliferation in pancreatic cancer by targeting CYR61/CTGF via the hippo pathway. Cancer Lett 2019; 460:42-53. [PMID: 31233838 DOI: 10.1016/j.canlet.2019.06.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/27/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a refractory disease. Programmed cell death protein-1 (PD-1) monotherapy has shown strong performance in targeting several malignancies. However, the effect and mechanism of intrinsic PD-1 in pancreatic cancer cells is still unknown. In this study, associations between clinicopathological characteristics and stained tissue microarrays of PDAC specimens were analyzed along with profiling and functional analyses. The results showed that cell-intrinsic PD-1 was significantly correlated with overall survival (OS). Independently of adaptive immunity, intrinsic PD-1 promoted tumor growth in PDAC. Concomitantly, the overexpression of intrinsic PD-1 enhanced cancer proliferation and inhibited cell apoptosis in vitro and in vivo. Mechanistically, PD-1 binds to the downstream MOB1, thereby inhibiting its phosphorylation. Moreover, greater synergistic tumor suppression in vitro resulted from combining Hippo inhibitors with anti-PD-1 treatment compared with the suppression achieved by either single agent alone. Additionally, Hippo downstream targets, CYR61 (CCN1) and CTGF (CCN2), were directly affected by PD-1 mediated Hippo signaling activation in concert with survival outcomes. Finally, the formulated nomogram showed superior predictive accuracy for OS in comparison with the TNM stage alone. Therefore, PD-1 immunotherapy in combination with Hippo pathway inhibitors may optimize the anti-tumor efficacy in PDAC patients via targeting cell-intrinsic PD-1.
Collapse
Affiliation(s)
- Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Shanshan Gao
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hanlin Yin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jian-Ang Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuan Fang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lei Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yefei Rong
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xuefeng Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dansong Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tiantao Kuang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dayong Jin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Yu
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Tomita Y, Kurozumi K, Yoo JY, Fujii K, Ichikawa T, Matsumoto Y, Uneda A, Hattori Y, Shimizu T, Otani Y, Oka T, Kaur B, Date I. Oncolytic Herpes Virus Armed with Vasculostatin in Combination with Bevacizumab Abrogates Glioma Invasion via the CCN1 and AKT Signaling Pathways. Mol Cancer Ther 2019; 18:1418-1429. [PMID: 31092561 DOI: 10.1158/1535-7163.mct-18-0799] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/30/2019] [Accepted: 05/10/2019] [Indexed: 11/16/2022]
Abstract
Anti-VEGF treatments such as bevacizumab have demonstrated convincing therapeutic advantage in patients with glioblastoma. However, bevacizumab has also been reported to induce invasiveness of glioma. In this study, we examined the effects of rapid antiangiogenesis mediated by oncolytic virus (RAMBO), an oncolytic herpes simplex virus-1 expressing vasculostatin, on bevacizumab-induced glioma invasion. The effect of the combination of RAMBO and bevacizumab in vitro was assessed by cytotoxicity, migration, and invasion assays. For in vivo experiments, glioma cells were stereotactically inoculated into the brain of mice. RAMBO was intratumorally injected 7 days after tumor inoculation, and bevacizumab was administered intraperitoneally twice a week. RAMBO significantly decreased both the migration and invasion of glioma cells treated with bevacizumab. In mice treated with bevacizumab and RAMBO combination, the survival time was significantly longer and the depth of tumor invasion was significantly smaller than those treated with bevacizumab monotherapy. Interestingly, RAMBO decreased the expression of cysteine-rich protein 61 and phosphorylation of AKT, which were increased by bevacizumab. These results suggest that RAMBO suppresses bevacizumab-induced glioma invasion, which could be a promising approach to glioma therapy.
Collapse
Affiliation(s)
- Yusuke Tomita
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiko Kurozumi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Ji Young Yoo
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Kentaro Fujii
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tomotsugu Ichikawa
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuji Matsumoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Atsuhito Uneda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuhiko Hattori
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshihiko Shimizu
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshihiro Otani
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Tetsuo Oka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Balveen Kaur
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
10
|
Yan J, Yang B, Lin S, Xing R, Lu Y. Downregulation of miR-142-5p promotes tumor metastasis through directly regulating CYR61 expression in gastric cancer. Gastric Cancer 2019; 22:302-313. [PMID: 30178386 DOI: 10.1007/s10120-018-0872-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 08/18/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recurrence is a primary cause of gastric cancer (GC)-related deaths. We reported previously that low expression of miR-142-5p could predict recurrence in GC. The present study aimed to investigate the function and mechanism of miR-142-5p in metastasis of GC. METHODS MiR-142-5p expression was detected in 101 GC samples by qRT-PCR. Its clinical significance was statistically analyzed. The roles of miR-142-5p and its candidate target gene CYR61 in metastasis were determined both in vivo and in vitro. RESULTS MiR-142-5p downregulation was significantly associated with the recurrence (P = 0.031) and poor prognosis of GC (P = 0.043). MiR-142-5p inhibited cancer cell migration and invasion both in vitro and in vivo. CYR61 was identified as a novel direct target of miR-142-5p by bioinformatics analysis of target prediction and luciferase reporter assay. The re-expression and knockdown of CYR61 could, respectively, rescue the effects induced by miR-142-5p overexpression and knockdown. MiR-142-5p attenuated GC cell migration and invasion, at least partially, by inactivation of the canonical Wnt/β-catenin signaling pathway through CYR61. CONCLUSIONS The newly identified miR-142-5p-CYR61-Wnt/β-catenin axis partially illustrates the molecular mechanism of GC recurrence and represents a novel prognosis biomarker for GC.
Collapse
Affiliation(s)
- Jing Yan
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Bing Yang
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Shuye Lin
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Rui Xing
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Youyong Lu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
11
|
Wei S, Wang J, Oyinlade O, Ma D, Wang S, Kratz L, Lal B, Xu Q, Liu S, Shah SR, Zhang H, Li Y, Quiñones-Hinojosa A, Zhu H, Huang ZY, Cheng L, Qian J, Xia S. Heterozygous IDH1 R132H/WT created by "single base editing" inhibits human astroglial cell growth by downregulating YAP. Oncogene 2018; 37:5160-5174. [PMID: 29849122 PMCID: PMC6590918 DOI: 10.1038/s41388-018-0334-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/06/2018] [Accepted: 05/04/2018] [Indexed: 11/09/2022]
Abstract
Mutations in the isocitrate dehydrogenase 1 (IDH1) gene have been identified in a number of cancer types, including brain cancer. The Cancer Genome Atlas project has revealed that IDH1 mutations occur in 70-80% of grade II and grade III gliomas. Until recently, most of the functional studies of IDH1 mutations in cellular models have been conducted in overexpression systems with the IDH1 wild type background. In this study, we employed a modified CRISPR/Cas9 genome editing technique called "single base editing", and efficiently introduced heterozygous IDH1 R132H mutation (IDH1R132H/WT) in human astroglial cells. Global DNA methylation profiling revealed hypermethylation as well as hypomethylation induced by IDH1R132H/WT. Global gene expression analysis identified molecular targets and pathways altered by IDH1R132H/WT, including cell proliferation, extracellular matrix (ECM), and cell migration. Our phenotype analysis indicated that compared with IDH1 wild type cells, IDH1R132H/WT promoted cell migration by upregulating integrin β4 (ITGB4); and significantly inhibited cell proliferation. Using our mutated IDH1 models generated by "single base editing", we identified novel molecular targets of IDH1R132H/WT, namely Yes-associated protein (YAP) and its downstream signaling pathway Notch, to mediate the cell growth-inhibiting effect of IDH1R132H/WT. In summary, the "single base editing" strategy has successfully created heterozygous IDH1 R132H mutation that recapitulates the naturally occurring IDH1 mutation. Our isogenic cellular systems that differ in a single nucleotide in one allele of the IDH1 gene provide a valuable model for novel discoveries of IDH1R132H/WT-driven biological events.
Collapse
Affiliation(s)
- Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, 430030, Wuhan, China.,Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jie Wang
- Department of Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Olutobi Oyinlade
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Ding Ma
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Shuyan Wang
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Lisa Kratz
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA
| | - Bachchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Qingfu Xu
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Senquan Liu
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Sagar R Shah
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA.,Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Yunqing Li
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA.,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | | | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.,Center for High Throughput Biology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Zhi-Yong Huang
- Department of General Surgery, Tongji Hospital, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Linzhao Cheng
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jiang Qian
- Department of Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Shuli Xia
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA. .,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
12
|
Goodwin CR, Rath P, Oyinlade O, Lopez H, Mughal S, Xia S, Li Y, Kaur H, Zhou X, Ahmed AK, Ho S, Olivi A, Lal B. Crizotinib and erlotinib inhibits growth of c-Met +/EGFRvIII + primary human glioblastoma xenografts. Clin Neurol Neurosurg 2018; 171:26-33. [PMID: 29803091 DOI: 10.1016/j.clineuro.2018.02.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Receptor tyrosine kinases (RTK), such as c-Met and epidermal growth factor receptor (EGFR), are implicated in the malignant progression of glioblastoma. Studies show that RTK systems can co-modulate distinct and overlapping oncogenic downstream signaling pathways. EGFRvIII, a constitutively activated EGFR deletion mutant variant, leads to increased tumor growth and diminishes the tumor growth response to HGF: c-Met pathway inhibitor therapy. Conversely, activation of the c-Met pathway diminishes the tumor growth response to EGFR pathway inhibitors. Previously we reported that EGFRvIII and c-Met pathway inhibitors synergize to inhibit tumor growth in isogenic GBM cell lines engineered to express EGFRvIII. More recently, studies suggest that despite targeting RTK signaling in glioblastoma multiforme, a subpopulation of stem-like tumor-propagating cells can persist to replenish the tumor cell population leading to tumor recurrence. PATIENTS AND METHODS Mayo 39 and Mayo 59 xenograft lines were cultured and xenografts were maintained. Subcutaneous xenograft lines were serially passaged in nude mice to generate subcutaneous xenografts. Xenografts were implanted in 6-8 week old nude mice. Once tumors reached a substantial size (150 mm3), mice were randomly divided into 4 groups: 1) control vehicle, 2) Crizotinib (crizo), 3) Erlotinib (erlot), or 4) Crizotinib + Erlotinib, (n = 5 per group). RESULTS Crizotinib (c-Met pathway inhibitor) and Erlotinib (EGFR pathway inhibitor) in combination significantly inhibited tumor growth, phospho-EGFRvIII, phospho-Met, phospho-AKT, phospho-MAPK, and neurosphere growth in Mayo 39 and Mayo 59 primary GBM subcutaneous xenografts. The expression of the stem cell markers Nestin, Musashi, Olig 2 and Sox2 were also significantly down-regulated by c-Met inhibition, but no additive down-regulation was seen by co-treatment with Erlotinib. CONCLUSIONS These results are consistent with and corroborate our previous findings demonstrating that targeting these two parallel pathways with c-Met and EGFR inhibitor therapy provides substantial anti-tumor activity in glioblastoma models.
Collapse
Affiliation(s)
- C Rory Goodwin
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States; Department of Neurosurgery, The Johns Hopkins University, School of Medicine, Baltimore, MD, United States; Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.
| | - Prakash Rath
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States; Department of Neurosurgery, The Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Olutobi Oyinlade
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States
| | - Hernando Lopez
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States; Department of Neurology, The Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Salman Mughal
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States
| | - Shuli Xia
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States; Department of Neurosurgery, The Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Yunqing Li
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States
| | - Harsharan Kaur
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States
| | - Xin Zhou
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States
| | - A Karim Ahmed
- Department of Neurosurgery, The Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Sandra Ho
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States
| | - Alessandro Olivi
- Department of Neurosurgery, The Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Bachchu Lal
- Department of Neurology, The Hugo W. Moser Research Institute at Kennedy Krieger Inc., United States; Department of Neurosurgery, The Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| |
Collapse
|
13
|
Tan Z, Zhao J, Jiang Y. MiR-634 sensitizes glioma cells to temozolomide by targeting CYR61 through Raf-ERK signaling pathway. Cancer Med 2018; 7:913-921. [PMID: 29473317 PMCID: PMC5852346 DOI: 10.1002/cam4.1351] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/19/2017] [Accepted: 12/30/2017] [Indexed: 12/11/2022] Open
Abstract
Glioma is the most common intracranial malignant tumors, accounting for about 40% of intracranial tumors. Primary or secondary drug resistance is one of the main reasons for the failure of treatment. The oncogenic or tumor-suppressive roles of miR-634 have been revealed in different types of cancer. However, the role of miR-634 in glioma remains unknown and whether miR-634 could sensitize glioma cells to temozolomide also is unclear. Here, we aim to investigate the biological function of miR-634 and the possible mechanisms in glioma. In this study, we found that miR-634 was downregulated in glioma tissues compared with normal brain tissues, and its expression was associated with tumor size and WHO grade. Importantly, glioma patients with low miR-634 expression showed a shorter survival time than patients which had high expression of miR-634. This study also showed that miR-634 was decreased in temozolomide-resistant glioma cells, and restoration of miR-634 could sensitize the resistant cells to temozolomide by targeting CYR61 through Raf-ERK signaling. Our study provides a potential target for overcome drug resistance in glioma.
Collapse
Affiliation(s)
- Zhigang Tan
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Jizong Zhao
- Department of Neurosurgery, Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| |
Collapse
|
14
|
Long non-coding RNA MT1DP shunts the cellular defense to cytotoxicity through crosstalk with MT1H and RhoC in cadmium stress. Cell Discov 2018; 4:5. [PMID: 29507753 PMCID: PMC5824791 DOI: 10.1038/s41421-017-0005-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Metallothioneins (MTs) are known to protect cells against oxidative stress, especially providing protection against cadmium (Cd) toxicity in hepatocytes. There are various gene variants and pseudogenes for MTs; however, there is little understanding on the functions of those non-coding MT members that are known to be expressed as long non-coding RNAs (lncRNAs) nowadays. Different from most protein-coding MT members, MT1DP was here found that remarkably induced to provoke cytotoxicity in hepatocytes in response to Cd treatment. MT1DP exerted such a pro-apoptotic function in Cd-treated hepatocytes through interacting with two partners: RhoC and MT1H. On one hand, MT1DP interacted with RhoC protein to increase the latter’s stability by preventing lysosome-dependent protein degradation. Therefore, upon Cd stress, MT1DP/RhoC complex was quickly reinforced to activate RhoC-CCN1/2-AKT signaling and potentiate Ca2+ influx, leading to enhanced Cd uptake and elevated Cd toxicity. On the other hand, MT1H, a protein-coding member of the MT family with little known function, was found to quickly respond to Cd exposure along with MT1DP. Mechanistically, MT1H and MT1DP were uncovered to mutually protect each other through a reciprocal ceRNA mechanism, building up a positive feedback loop to enforce MT1DP-conducted signaling upon Cd exposure. Moreover, MT1DP was found to contribute much more to the activation of RhoC-CCN1/2-AKT signaling than MT1H. Considered together, we here unveiled a mystery whether a pseudogene within the MT family, MT1DP, has actual biological functions in regulating Cd-induced cellular defense. Our findings unearthed an important role of pseudogene MT1DP in calibrating the cellular machinery to switch the cellular defense to cytotoxicity through crosslinking an interplay between its two partners, namely MT1H and RhoC, under cadmium stress.
Collapse
|
15
|
Wang R, Wei B, Wei J, Tian Y, Du C. Cysteine-rich 61-associated gene expression profile alterations in human glioma cells. Mol Med Rep 2017; 16:5561-5567. [PMID: 28849002 DOI: 10.3892/mmr.2017.7216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 02/20/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate gene expression profile alterations associated with cysteine‑rich 61 (CYR61) expression in human glioma cells. The GSE29384 dataset, downloaded from the Gene Expression Omnibus, includes three LN229 human glioma cell samples expressing CYR61 induced by doxycycline (Dox group), and three control samples not exposed to doxycycline (Nodox group). Differentially expressed genes (DEGs) between the Dox and Nodox groups were identified with cutoffs of |log2 fold change (FC)|>0.5 and P<0.05. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses for DEGs were performed. Protein‑protein interaction (PPI) network and module analyses were performed to identify the most important genes. Transcription factors (TFs) were obtained by detecting the TF binding sites of DEGs using a Whole Genome rVISTA online tool. A total of 258 DEGs, including 230 (89%) upregulated and 28 (11%) downregulated DEGs were identified in glioma cells expressing CYR61 compared to cells without CYR61 expression. The majority of upregulated DEGs, including interferon (IFN)B1, interferon‑induced (IFI)44 and interferon regulatory factor (IRF)7, were associated with immune, defense and virus responses, and cytokine‑cytokine receptor interaction signaling pathways. Signal transducer and activator of transcription 1 (STAT1) and DEAD‑box helicase 58 (DDX58) were observed to have high connection degrees in the PPI network. A total of seven TFs of the DEGs, including interferon consensus sequence‑binding protein and IFN‑stimulated gene factor‑3 were additionally detected. In conclusion, IFNB1, genes encoding IFN‑induced proteins (IFI16, IFI27, IFI44 and IFITM1), IRFs (IRF1, IRF7 and IRF9), STAT1 and DDX58 were demonstrated to be associated with CYR61 expression in glioma cells; thus, they may be critical for maintaining the role of CYR61 during cancer progression.
Collapse
Affiliation(s)
- Rui Wang
- Department of Radiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Bo Wei
- Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Jun Wei
- Department of Science and Education Section, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yu Tian
- Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Chao Du
- Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
16
|
Wan J, Su Y, Song Q, Tung B, Oyinlade O, Liu S, Ying M, Ming GL, Song H, Qian J, Zhu H, Xia S. Methylated cis-regulatory elements mediate KLF4-dependent gene transactivation and cell migration. eLife 2017; 6:e20068. [PMID: 28553926 PMCID: PMC5466421 DOI: 10.7554/elife.20068] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 05/24/2017] [Indexed: 12/13/2022] Open
Abstract
Altered DNA methylation status is associated with human diseases and cancer; however, the underlying molecular mechanisms remain elusive. We previously identified many human transcription factors, including Krüppel-like factor 4 (KLF4), as sequence-specific DNA methylation readers that preferentially recognize methylated CpG (mCpG), here we report the biological function of mCpG-dependent gene regulation by KLF4 in glioblastoma cells. We show that KLF4 promotes cell adhesion, migration, and morphological changes, all of which are abolished by R458A mutation. Surprisingly, 116 genes are directly activated via mCpG-dependent KLF4 binding activity. In-depth mechanistic studies reveal that recruitment of KLF4 to the methylated cis-regulatory elements of these genes result in chromatin remodeling and transcription activation. Our study demonstrates a new paradigm of DNA methylation-mediated gene activation and chromatin remodeling, and provides a general framework to dissect the biological functions of DNA methylation readers and effectors.
Collapse
Affiliation(s)
- Jun Wan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Yijing Su
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Qifeng Song
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, United States
- Center for High-Throughput Biology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Brian Tung
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, United States
| | - Olutobi Oyinlade
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Sheng Liu
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Mingyao Ying
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, United States
| | - Guo-li Ming
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Hongjun Song
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jiang Qian
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, United States
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Heng Zhu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, United States
- Center for High-Throughput Biology, Johns Hopkins University School of Medicine, Baltimore, United States
- Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, United States
| | - Shuli Xia
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
17
|
Kiwanuka E, Junker JP, Eriksson E. Transforming growth factor β1 regulates the expression of CCN2 in human keratinocytes via Smad-ERK signalling. Int Wound J 2017; 14:1006-1018. [PMID: 28371159 DOI: 10.1111/iwj.12749] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/04/2017] [Indexed: 01/22/2023] Open
Abstract
Connective tissue growth factor (CCN2/CTGF) and transforming growth factor β1 (TGF-β1) are important regulators of skin wound healing, but controversy remains regarding their expression in epithelial cell lineages. Here, we investigate the expression of CCN2 in keratinocytes during reepithelialisation and its regulation by TGF-β1. CCN2 was detected in the epidermis of healing full-thickness porcine wounds. Human keratinocytes were incubated with or without 10 ng/ml TGF-β1, and signalling pathways were blocked with 10-μM SIS3 or 20-μM PD98059. Semi-quantitative real-time PCR was used to study CCN2 mRNA expression, and western blot was used to measure CCN2, phosphorylated-ERK1/2, ERK1/2, phosphorylated-Smad3 and Smad2/3 proteins. CCN2 was transiently expressed in neoepidermis at the leading edge of the wound in vivo. In vitro, CCN2 expression was induced by TGF-β1 at 2 hours (7·5 ± 1·9-fold mRNA increase and 3·0 ± 0·6-fold protein increase) and 12 hours (5·4 ± 1·9-fold mRNA increase and 3·3 ± 0·6-fold protein increase). Compared with inhibiting the SMAD pathway, inhibiting the mitogen-activated protein kinase (MAPK) pathway was more effective in reducing TGF-β1-induced CCN2 mRNA and protein expression. Inhibition of the MAPK pathway had minimal impact on the activity of the SMAD pathway. CCN2 is expressed in keratinocytes in response to tissue injury or TGF-β1. In addition, TGF-β1 induces CCN2 expression in keratinocytes through the ras/MEK/ERK pathway. A complete understanding of CCN2 expression in keratinocytes is critical to developing novel therapies for wound healing and cutaneous malignancy.
Collapse
Affiliation(s)
- Elizabeth Kiwanuka
- Department of Plastic Surgery, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, USA
| | - Johan Pe Junker
- Center for Disaster Medicine and Traumatology, Department of Clinical and Experimental Medicine, Linköping University Hospital, Linköping, Sweden
| | - Elof Eriksson
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Zhu X, Song Y, Wu C, Pan C, Lu P, Wang M, Zheng P, Huo R, Zhang C, Li W, Lin Y, Cao Y, Li N. Cyr61 participates in the pathogenesis of acute lymphoblastic leukemia by enhancing cellular survival via the AKT/NF-κB signaling pathway. Sci Rep 2016; 6:34018. [PMID: 27725691 PMCID: PMC5057070 DOI: 10.1038/srep34018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/06/2016] [Indexed: 12/25/2022] Open
Abstract
Cyr61 (CCN1) is the product of a growth factor–inducible immediate early gene and is involved in cell adhesion, survival, proliferation, and differentiation. Cyr61 is overexpressed in human tumors and is involved in the development of tumors. However, the role that Cyr61 plays in acute lymphoblastic leukemia (ALL) cells remains undetermined. The aim of this study was to identify the role of Cyr61 in regulating ALL cell survival. Here, we found that the level of Cyr61 was increased in the plasma and bone marrow (BM) from ALL patients compared with samples from normal control patients. Furthermore, we observed that Cyr61 could effectively stimulate Jurkat (T ALL cell lines), Nalm-6 (B ALL cell lines), and primary ALL cell survival. Mechanistically, we showed that Cyr61 stimulated ALL cell survival via the AKT/NF-κB signaling pathways and the consequent up-regulation of Bcl-2. Taken together, our study is the first to reveal that Cyr61 is elevated in ALL and promotes cell survival through the AKT/NF-κB pathway by up-regulating Bcl-2. Our findings suggest that Cyr61 plays an important role in the pathogenesis of ALL.
Collapse
Affiliation(s)
- Xianjin Zhu
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Yanfang Song
- Affiliated Renmin Hospital of Fujian University of Traditional Chinese Medicine, 602 Bayiqi Road, Fuzhou 350001, China
| | - Conglian Wu
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Chuxi Pan
- University of Toronto, 27 King's College Circle, Toronto M5S1A1, Canada
| | - Pingxia Lu
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Meihua Wang
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Peizheng Zheng
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Rongfen Huo
- Shanghai Institute of Immunology, Institute of medical sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Chenqing Zhang
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Wanting Li
- Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Yulin Lin
- Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Yingping Cao
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China
| | - Ningli Li
- Shanghai Institute of Immunology, Institute of medical sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
19
|
Di Y, Zhang Y, Hui L, Yang H, Yang Y, Wang A, Chen X. Cysteine‑rich 61 RNA interference inhibits pathological angiogenesis via the phosphatidylinositol 3‑kinase/Akt‑vascular endothelial growth factor signaling pathway in endothelial cells. Mol Med Rep 2016; 14:4321-4327. [PMID: 27666419 DOI: 10.3892/mmr.2016.5772] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 11/18/2015] [Indexed: 11/05/2022] Open
Abstract
Hypoxia is a key factor in the pathogenesis of angiogenesis, and cysteine‑rich 61 (CCN1), an angiogenic factor, is involved in the development of pathological angiogenesis. The aim of the present study was to investigate the mechanism of CCN1 RNA interference (RNAi)‑induced inhibition of hypoxia‑induced pathological angiogenesis in endothelial cells. Human umbilical vein endothelial cells (HUVECs) were cultured under hypoxic conditions in vitro. The effects of inhibiting phosphoinositide 3‑kinase (PI3K)/Akt signaling using LY294002 were investigated in hypoxic HUVECs. The proliferation and apoptosis of HUVECs under hypoxia were assessed using CCN1 RNAi. The CCN1‑PI3K/Akt‑vascular endothelial growth factor (VEGF) pathway was analyzed under hypoxic conditions using reverse transcription‑quantitative polymerase chain reaction and western blotting. CCN1 RNAi inhibited the proliferation and induced the apoptosis of the HUVECs under hypoxia, with hypoxia significantly increasing the mRNA and protein expression levels of CCN1, Akt and VEGF. By contrast, CCN1 RNAi reduced the mRNA and protein expression levels of CCN1, Akt and VEGF in the HUVECs (P<0.05). Furthermore, LY294002 reduced the mRNA and protein expression levels of CCN1 in the hypoxic cells (P<0.05). These data indicated that CCN1 inhibits apoptosis and promotes proliferation in HUVECs. Therefore, CCN1 RNAi may offer a novel therapeutic strategy, which may aid in the treatment of pathological angiogenesis via inhibition of the PI3K/Akt‑VEGF pathway.
Collapse
Affiliation(s)
- Yu Di
- Department of Ophthalmology, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yiou Zhang
- Graduate School, China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Linping Hui
- Laboratory Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hongwei Yang
- Department of Ophthalmology, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yang Yang
- Department of Ophthalmology, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Aiyuan Wang
- Department of Ophthalmology, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xiaolong Chen
- Department of Ophthalmology, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
20
|
Gründker C, Bauerschmitz G, Schubert A, Emons G. Invasion and increased expression of S100A4 and CYR61 in mesenchymal transformed breast cancer cells is downregulated by GnRH. Int J Oncol 2016; 48:2713-21. [PMID: 27098123 DOI: 10.3892/ijo.2016.3491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/17/2016] [Indexed: 11/05/2022] Open
Abstract
S100 calcium binding protein A4 (S100A4) and cysteine-rich angiogenic inducer 61 (CYR61) play important roles in epithelial-mesenchymal-transition (EMT), invasion and metastasis by promoting cancer cell motility. Recently we were able to show that invasion of GnRH receptor-positive breast cancer cells is time- and dose-dependently reduced by GnRH analogs. We have now analyzed whether GnRH treatment affects S100A4 and CYR61 in mesenchymal transformed breast cancer cells. S100A4 and CYR61 expression was analyzed using RT-PCR. Invasion was quantified by assessment of breast cancer cell migration rate through an artificial basement membrane. The role of S100A4 and CYR61 in invasion of breast cancer cells was analyzed by neutralizing their biological activity. Expression of S100A4, CYR61 and GnRH receptor in human breast cancers, normal and other non-malignant breast tissues was analyzed by immuno-histochemistry. Invasion and expression of S100A4 and CYR61 in MDA-MB-231 breast cancer cells were significant higher as compared with MCF-7 breast cancer cells. Invasion and expression of S100A4 and CYR61 were significantly increased in mesenchymal transformed MCF-7 cells (MCF-7-EMT). The increased invasion of MCF-7-EMT cells could be reduced by anti-S100A4 and anti-CYR61 antibodies. In addition, invasion of MDA-MB-231 cells was decreased by anti-S100A4 and anti-CYR61 antibodies. Treatment of MCF-7-EMT and MDA-MB-231 cells with GnRH agonist Triptorelin resulted in a significant decrease of invasion and expression of S100A4 and CYR61. Both, S100A4 and CYR61 were found highly expressed in biopsy specimens of breast hyperplasia and malignant breast cancers. GnRH receptor expression was detectable in approximately 71% of malignant breast cancers. Our findings suggest that S100A4 and CYR61 play major roles in breast cancer invasion. Both, invasion and expression of S100A4 and CYR61 can be inhibited by GnRH treatment.
Collapse
Affiliation(s)
- Carsten Gründker
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Gerd Bauerschmitz
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Antje Schubert
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| |
Collapse
|
21
|
Microarray-Based Phospho-Proteomic Profiling of Complex Biological Systems. Transl Oncol 2016; 9:124-129. [PMID: 27084428 PMCID: PMC4833893 DOI: 10.1016/j.tranon.2016.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/16/2016] [Accepted: 02/17/2016] [Indexed: 01/29/2023] Open
Abstract
Protein microarray technology has been successfully used for identifying substrates of purified activated kinases. We used protein microarrays to globally interrogate the effects of PTEN and Akt activity on the phospho-kinome of in vitro and in vivo glioma models and validated results in clinical pathological specimens. Whole cell lysates extracted from tumor samples can be applied to human kinome chip microarrays to profile the global kinase phosphorylation patterns in a high-throughput manner and identify novel substrates inherent to the tumor cell and the interactions with tumor microenvironment. Our findings identify a novel microarray-based method for assessing intracellular signaling events applicable to human oncogenesis and other pathophysiologic states.
Collapse
|
22
|
Sawyer AJ, Kyriakides TR. Matricellular proteins in drug delivery: Therapeutic targets, active agents, and therapeutic localization. Adv Drug Deliv Rev 2016; 97:56-68. [PMID: 26763408 DOI: 10.1016/j.addr.2015.12.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
Extracellular matrix is composed of a complex array of molecules that together provide structural and functional support to cells. These properties are mainly mediated by the activity of collagenous and elastic fibers, proteoglycans, and proteins such as fibronectin and laminin. ECM composition is tissue-specific and could include matricellular proteins whose primary role is to modulate cell-matrix interactions. In adults, matricellular proteins are primarily expressed during injury, inflammation and disease. Particularly, they are closely associated with the progression and prognosis of cardiovascular and fibrotic diseases, and cancer. This review aims to provide an overview of the potential use of matricellular proteins in drug delivery including the generation of therapeutic agents based on the properties and structures of these proteins as well as their utility as biomarkers for specific diseases.
Collapse
|
23
|
Li J, Ye L, Owen S, Weeks HP, Zhang Z, Jiang WG. Emerging role of CCN family proteins in tumorigenesis and cancer metastasis (Review). Int J Mol Med 2015; 36:1451-63. [PMID: 26498181 PMCID: PMC4678164 DOI: 10.3892/ijmm.2015.2390] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/07/2015] [Indexed: 12/28/2022] Open
Abstract
The CCN family of proteins comprises the members CCN1, CCN2, CCN3, CCN4, CCN5 and CCN6. They share four evolutionarily conserved functional domains, and usually interact with various cytokines to elicit different biological functions including cell proliferation, adhesion, invasion, migration, embryonic development, angiogenesis, wound healing, fibrosis and inflammation through a variety of signalling pathways. In the past two decades, emerging functions for the CCN proteins (CCNs) have been identified in various types of cancer. Perturbed expression of CCNs has been observed in a variety of malignancies. The aberrant expression of certain CCNs is associated with disease progression and poor prognosis. Insight into the detailed mechanisms involved in CCN-mediated regulation may be useful in understanding their roles and functions in tumorigenesis and cancer metastasis. In this review, we briefly introduced the functions of CCNs, especially in cancer.
Collapse
Affiliation(s)
- Jun Li
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Institute of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Sioned Owen
- Cardiff China Medical Research Collaborative, Institute of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Hoi Ping Weeks
- Cardiff China Medical Research Collaborative, Institute of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Institute of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| |
Collapse
|
24
|
Liu Z, Wei P, Yang Y, Cui W, Cao B, Tan C, Yu B, Bi R, Xia K, Chen W, Wang Y, Zhang Y, Du X, Zhou X. BATF2 Deficiency Promotes Progression in Human Colorectal Cancer via Activation of HGF/MET Signaling: A Potential Rationale for Combining MET Inhibitors with IFNs. Clin Cancer Res 2015; 21:1752-63. [PMID: 25762344 DOI: 10.1158/1078-0432.ccr-14-1564] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 01/13/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE BATF2, a novel IFN-stimulated gene, inhibits tumor cell proliferation, invasion, and migration. The objectives of this study were to determine how BATF2 expression is associated with colorectal cancer progression and patient outcome, to investigate how BATF2 overexpression inhibits hepatocyte growth factor (HGF)/MET signaling, and to elucidate the rationale for combining MET inhibitors with IFN. EXPERIMENTAL DESIGN BATF2 expression in colorectal cancer tissues was determined and correlated with colorectal cancer patient prognosis. Cultured colorectal cancer cells were used to investigate the effects of BATF2 overexpression on the malignant phenotype of colorectal cancer cells and HGF/MET signaling. Tumor xenograft models were used to validate the effects of BATF2 on colorectal cancer xenograft growth and assess the efficacy of the combination of MET inhibitors with IFNs in colorectal cancer. RESULTS In colorectal cancer tissues, BATF2 was found to be significantly downregulated, and its expression negatively correlated with MET expression. Decreased BATF2 expression was associated with progression and shorter patient survival in colorectal cancer. BATF2 overexpression promoted apoptosis and inhibited proliferation, migration, and invasion in colorectal cancer cells, as well as dramatically blunted tumor xenograft growth. In addition, MET inhibitors in combination with IFNβ produced synergistic cytotoxicity both in vitro and in vivo. CONCLUSIONS Together, these novel findings suggest that BATF2, a tumor suppressor gene, is a potent negative regulator of HGF/MET signaling in colorectal cancer and may serve as a prognostic tumor marker. Furthermore, these results provide a rationale for combining MET inhibitors with IFNs in preclinical trials.
Collapse
Affiliation(s)
- Zebing Liu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Department of Pathology, Jinshan Hospital, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Yu Yang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wenli Cui
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Bing Cao
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Cong Tan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Baohua Yu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Rui Bi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Kaiqin Xia
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Weixiang Chen
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Department of Pathology, Shanghai Gongli Hospital, Shanghai, China
| | - Yiqin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Youyuan Zhang
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China. Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. Institute of Pathology, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Park YS, Hwang S, Jin YM, Yu Y, Jung SA, Jung SC, Ryu KH, Kim HS, Jo I. CCN1 secreted by tonsil-derived mesenchymal stem cells promotes endothelial cell angiogenesis via integrin αv β3 and AMPK. J Cell Physiol 2015; 230:140-9. [PMID: 24909560 DOI: 10.1002/jcp.24690] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 05/21/2014] [Indexed: 11/12/2022]
Abstract
CCN1 is highly expressed in cancer cells and has been identified in the secretome of bone marrow-derived mesenchymal stem cells (BM-MSC). Although secreted CCN1 is known to promote angiogenesis, its underlying mechanism remains unclear. Here, we examined whether our recently-established tonsil-derived MSC (T-MSC) secrete CCN1 and, if any, how CCN1 promotes the angiogenesis of human umbilical vein endothelial cells (HUVEC). Compared with untreated control T-MSC, a higher level of CCN1 was secreted by T-MSC treated with activin A and sonic hedgehog, drugs known to induce endodermal differentiation. Expectedly, conditioned medium collected from differentiated T-MSC (DCM) significantly increased HUVEC migration and tube formation compared with that from control T-MSC (CCM), and these stimulatory effects were reversed by neutralization with anti-CCN1 antibody. Treatment with recombinant human CCN1 (rh-CCN1) alone also mimicked the stimulatory effects of DCM. Furthermore, treatment with either DCM or rh-CCN1 increased the phosphorylation of AMP kinase (AMPK), and ectopic expression of siRNA of the AMPK gene inhibited all observed effects of both DCM and rh-CCN1. However, no alteration of intracellular ATP levels or phosphorylation of LKB1, a well-known upstream factor of AMPK activation, was observed under our conditions. Finally, the neutralization of integrin α(v) β(3) with anti-integrin α(v) β(3) antibody almost completely reversed the effects of CCN1 on AMPK phosphorylation, and EC migration and tube formation. Taken together, we demonstrated that T-MSC increase the secretion of CCN1 in response to endodermal differentiation and that integrin α(v) β(3) and AMPK mediate CCN1-induced EC migration and tube formation independent of intracellular ATP levels alteration.
Collapse
Affiliation(s)
- Yoon Shin Park
- Department of Molecular Medicine, School of Medicine, and Global Top 5 Research Program, Ewha Womans University, Mok-6-dong, Yangcheon-gu, Seoul 158-710, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Cheng G, Zhang H, Zhang L, Zhang J. Cyr61 promotes growth of glioblastoma in vitro and in vivo. Tumour Biol 2014; 36:2869-73. [DOI: 10.1007/s13277-014-2915-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 11/27/2014] [Indexed: 01/09/2023] Open
|
27
|
Thorne AH, Meisen WH, Russell L, Yoo JY, Bolyard CM, Lathia JD, Rich J, Puduvalli VK, Mao H, Yu J, Caligiuri MA, Tridandapani S, Kaur B. Role of cysteine-rich 61 protein (CCN1) in macrophage-mediated oncolytic herpes simplex virus clearance. Mol Ther 2014; 22:1678-87. [PMID: 24895995 DOI: 10.1038/mt.2014.101] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 05/23/2014] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma is a devastating disease, and there is an urgent need to develop novel therapies, such as oncolytic HSV1 (OV) to effectively target tumor cells. OV therapy depends on tumor-specific replication leading to destruction of neoplastic tissues. Host responses that curtail virus replication limit its efficacy in vivo. We have previously shown that cysteine-rich 61 protein (CCN1) activates a type 1 IFN antiviral defense response in glioblastoma cells. Incorporating TCGA data, we found CCN1 expression to be a negative prognostic factor for glioblastoma patients. Based on this, we used neutralizing antibodies against CCN1 to investigate its effect on OV therapy. Use of an anti-CCN1 antibody in mice bearing glioblastomas treated with OV led to enhanced virus expression along with reduced immune cell infiltration. OV-induced CCN1 increases macrophage migration toward infected glioblastoma cells by directly binding macrophages and also by enhancing the proinflammatory activation of macrophages inducing MCP-1 expression in glioblastoma cells. Activation of macrophages by CCN1 also increases viral clearance. Neutralization of integrin αMβ2 reversed CCN1-induced macrophage activation and migration, and reduced MCP-1 expression by glioblastoma cells. Our findings reveal that CCN1 plays a novel role in pathogen clearance; increasing macrophage infiltration and activation resulting in increased virus clearance in tumors.
Collapse
Affiliation(s)
- Amy Haseley Thorne
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Walter H Meisen
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Luke Russell
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ji Young Yoo
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Chelsea M Bolyard
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jeremy Rich
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Vinay K Puduvalli
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Hsiaoyin Mao
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Michael A Caligiuri
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Susheela Tridandapani
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Balveen Kaur
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
28
|
Tilghman J, Wu H, Sang Y, Shi X, Guerrero-Cazares H, Quinones-Hinojosa A, Eberhart CG, Laterra J, Ying M. HMMR maintains the stemness and tumorigenicity of glioblastoma stem-like cells. Cancer Res 2014; 74:3168-79. [PMID: 24710409 DOI: 10.1158/0008-5472.can-13-2103] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Glioblastoma (GBM) stem cells (GSC) are a subpopulation of tumor cells that display stem-like characteristics (stemness) and play unique roles in tumor propagation, therapeutic resistance, and tumor recurrence. Therapeutic targets in GSCs are a focus of increasing interest to improve GBM therapy. Here we report that the hyaluronan-mediated motility receptor (HMMR) is highly expressed in GBM tumors, where it supports the self-renewal and tumorigenic potential of GSCs. HMMR silencing impairs GSC self-renewal and inhibits the expression of GSC markers and regulators. Furthermore, HMMR silencing suppresses GSC-derived tumor growth and extends the survival of mice bearing GSC xenografts. Conversely, HMMR overexpression promotes GSC self-renewal and intracranial tumor propagation. In human GBM tumor specimens, HMMR expression is correlated positively with the expression of stemness-associated markers and regulators. Our findings identify HMMR as a candidate therapeutic target to GSCs as a GBM treatment strategy.
Collapse
Affiliation(s)
- Jessica Tilghman
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GeorgiaAuthors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Hao Wu
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Yingying Sang
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Xiaohai Shi
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Hugo Guerrero-Cazares
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Alfredo Quinones-Hinojosa
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GeorgiaAuthors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Charles G Eberhart
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GeorgiaAuthors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - John Laterra
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GeorgiaAuthors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GeorgiaAuthors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GeorgiaAuthors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Mingyao Ying
- Authors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GeorgiaAuthors' Affiliations: Hugo W. Moser Research Institute at Kennedy Krieger; Departments of Neurology, Neuroscience, Oncology, Neurosurgery, and Pathology, Johns Hopkins School of Medicine; Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; and Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| |
Collapse
|
29
|
Jeong D, Heo S, Sung Ahn T, Lee S, Park S, Kim H, Park D, Byung Bae S, Lee SS, Soo Lee M, Kim CJ, Jun Baek M. Cyr61 expression is associated with prognosis in patients with colorectal cancer. BMC Cancer 2014; 14:164. [PMID: 24606730 PMCID: PMC3975645 DOI: 10.1186/1471-2407-14-164] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 02/28/2014] [Indexed: 01/22/2023] Open
Abstract
Background Cysteine-rich 61 (Cyr61), a member of the CCN protein family, possesses diverse functionality in cellular processes such as adhesion, migration, proliferation, and survival. Cyr61 can also function as an oncogene or a tumour suppressor, depending on the origin of the cancer. Only a few studies have reported Cyr61 expression in colorectal cancer. In this study, we assessed the Cyr61 expression in 251 colorectal cancers with clinical follow up. Methods We examined Cyr61 expression in 6 colorectal cancer cell lines (HT29, Colo205, Lovo, HCT116, SW480, SW620) and 20 sets of paired normal and colorectal cancer tissues by western blot. To validate the association of Cyr61 expression with clinicopathological parameters, we assessed Cyr61 expression using tissue microarray analysis of primary colorectal cancer by immunohistochemical analysis. Results We verified that all of the cancer cell lines expressed Cyr61; 2 cell lines (HT29 and Colo205) demonstrated Cyr61 expression to a slight extent, while 4 cell lines (Lovo, HCT116, SW480, SW620) demonstrated greater Cyr61 expression than HT29 and Colo205 cell lines. Among the 20 cases of paired normal and tumour tissues, greater Cyr61 expression was observed in 16 (80%) tumour tissues than in normal tissues. Furthermore, 157 out of 251 cases (62.5%) of colorectal cancer examined in this study displayed strong Cyr61 expression. Cyr61 expression was found to be associated with pN (p = 0.018). Moreover, Cyr61 expression was associated with statistically significant cancer-specific mortality (p = 0.029). The duration of survival was significantly lesser in patients with Cyr61 high expression than in patients with Cyr61 low expression (p = 0.001). These results suggest that Cyr61 expression plays several important roles in carcinogenesis and may also be a good prognostic marker for colorectal cancer. Conclusions Our data confirmed that Cyr61 was expressed in colorectal cancers and the expression was correlated with worse prognosis of colorectal cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Moo Jun Baek
- Department of Surgery, College of Medicine, Soonchunhyang University, 31 soonchunhyang 6 gil, Dongnam-gu, Cheonan, Chungcheongnam-do 330-722, Republic of Korea.
| |
Collapse
|
30
|
Huang J, Gao K, Lin J, Wang Q. MicroRNA-100 inhibits osteosarcoma cell proliferation by targeting Cyr61. Tumour Biol 2013; 35:1095-100. [PMID: 24317814 DOI: 10.1007/s13277-013-1146-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 08/26/2013] [Indexed: 12/22/2022] Open
Abstract
Increasing evidence indicates that microRNAs (miRNAs) participate in almost every step of cellular processes and are often aberrantly expressed in human cancer. Therefore, the discovery of miRNAs may provide a new and powerful tool for understanding the mechanism and treatment of carcinogenesis. The aim of this study was to investigate the functional significance of miR-100 and to identify its possible target genes in osteosarcoma (OS) cells. Here, we found that expression level of miR-100 was significantly decreased in osteosarcoma tissues in comparison with the adjacent normal tissues. The enforced expression of miR-100 was able to inhibit cell proliferation in Saos-2 and MG63 cells, while its antisense oligonucleotides (antisense miR-100) promoted cell proliferation. Moreover, our results further revealed that expression of Cyr61, an extracellular matrix-associated growth factor, was negatively regulated by miR-100. Therefore, we consider that miR-100 acts as a tumor suppressor for osteosarcoma. It may provide novel diagnostic and therapeutic options for human osteosarcoma in the future.
Collapse
Affiliation(s)
- Jianhua Huang
- Department of Orthopaedics, Affiliated People's First Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | | | | | | |
Collapse
|
31
|
Shen L, Chen XD, Zhang YH. MicroRNA-128 promotes proliferation in osteosarcoma cells by downregulating PTEN. Tumour Biol 2013; 35:2069-74. [PMID: 24132591 DOI: 10.1007/s13277-013-1274-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 09/25/2013] [Indexed: 01/10/2023] Open
Abstract
MicroRNAs are a class of small noncoding RNAs that function as critical gene regulators through targeting mRNAs for translational repression or degradation. Several studies have indicated that abnormal expression of miRNAs occurs frequently in human osteosarcoma. In the present study, we found that miR-128 expression was significantly increased in osteosarcoma tissues compared to adjacent normal tissues. Ectopic overexpression of miR-128 significantly promoted while suppression of miR-128 by its antisense inhibited the proliferation of MG63 and U2OS cells. At the molecular level, our results demonstrated that miR-128 overexpression could repress expression of PTEN by directly targeting PTEN 3'-untranslated region. Consistently, downstream AKT signaling was altered by miR-128 overexpression or knockdown. Therefore, our results suggest that miR-128 plays an important role in the proliferation of human osteosarcoma cells by directly regulation of PTEN/AKT signaling.
Collapse
Affiliation(s)
- Lei Shen
- Department of Orthopaedic Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, No.1665 kongjiang Rd., Shanghai, 200092, People's Republic of China
| | | | | |
Collapse
|
32
|
Newly synthesized quinazolinone HMJ-38 suppresses angiogenetic responses and triggers human umbilical vein endothelial cell apoptosis through p53-modulated Fas/death receptor signaling. Toxicol Appl Pharmacol 2013; 269:150-62. [DOI: 10.1016/j.taap.2013.03.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Revised: 03/09/2013] [Accepted: 03/12/2013] [Indexed: 01/28/2023]
|
33
|
Xie H, Zhao Y, Caramuta S, Larsson C, Lui WO. miR-205 expression promotes cell proliferation and migration of human cervical cancer cells. PLoS One 2012; 7:e46990. [PMID: 23056551 PMCID: PMC3463520 DOI: 10.1371/journal.pone.0046990] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 09/07/2012] [Indexed: 01/04/2023] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNA regulators that control gene expression mainly through post-transcriptional silencing. We previously identified miR-205 in a signature for human cervical cancer using a deep sequencing approach. In this study, we confirmed that miR-205 expression was frequently higher in human cervical cancer than their matched normal tissue samples. Functionally, we demonstrate that miR-205 promotes cell proliferation and migration in human cervical cancer cells. To further understand the biological roles of miR-205, we performed in vivo crosslinking and Argonaute 2 immunoprecipitation of miRNA ribonucleoprotein complexes followed by microarray analysis (CLIP-Chip) to identify its potential mRNA targets. Applying CLIP-Chip on gain- and loss-of-function experiments, we identified a set of transcripts as potential targets of miR-205. Several targets are functionally involved in cellular proliferation and migration. Two of them, CYR61 and CTGF, were further validated by Western blot analysis and quantification of mRNA enrichment in the Ago2 immunoprecipitates using qRT-PCR. Furthermore, both CYR61 and CTGF were downregulated in cervical cancer tissues. In summary, our findings reveal novel functional roles and targets of miR-205 in human cervical cancer, which may provide new insights about its role in cervical carcinogenesis and its potential value for clinical diagnosis.
Collapse
Affiliation(s)
- Hong Xie
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
- * E-mail: (HX); (WL)
| | - Yungang Zhao
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin, China
| | - Stefano Caramuta
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
| | - Catharina Larsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
| | - Weng-Onn Lui
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
- * E-mail: (HX); (WL)
| |
Collapse
|
34
|
Abstract
While most gene transcription yields RNA transcripts that code for proteins, a sizable proportion of the genome generates RNA transcripts that do not code for proteins, but may have important regulatory functions. The brain-derived neurotrophic factor (BDNF) gene, a key regulator of neuronal activity, is overlapped by a primate-specific, antisense long noncoding RNA (lncRNA) called BDNFOS. We demonstrate reciprocal patterns of BDNF and BDNFOS transcription in highly active regions of human neocortex removed as a treatment for intractable seizures. A genome-wide analysis of activity-dependent coding and noncoding human transcription using a custom lncRNA microarray identified 1288 differentially expressed lncRNAs, of which 26 had expression profiles that matched activity-dependent coding genes and an additional 8 were adjacent to or overlapping with differentially expressed protein-coding genes. The functions of most of these protein-coding partner genes, such as ARC, include long-term potentiation, synaptic activity, and memory. The nuclear lncRNAs NEAT1, MALAT1, and RPPH1, composing an RNAse P-dependent lncRNA-maturation pathway, were also upregulated. As a means to replicate human neuronal activity, repeated depolarization of SY5Y cells resulted in sustained CREB activation and produced an inverse pattern of BDNF-BDNFOS co-expression that was not achieved with a single depolarization. RNAi-mediated knockdown of BDNFOS in human SY5Y cells increased BDNF expression, suggesting that BDNFOS directly downregulates BDNF. Temporal expression patterns of other lncRNA-messenger RNA pairs validated the effect of chronic neuronal activity on the transcriptome and implied various lncRNA regulatory mechanisms. lncRNAs, some of which are unique to primates, thus appear to have potentially important regulatory roles in activity-dependent human brain plasticity.
Collapse
|
35
|
Wouters J, Stas M, Govaere O, Van den Eynde K, Vankelecom H, van den Oord JJ. Gene expression changes in melanoma metastases in response to high-dose chemotherapy during isolated limb perfusion. Pigment Cell Melanoma Res 2012; 25:454-65. [PMID: 22486811 DOI: 10.1111/j.1755-148x.2012.01004.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite recent advances in melanoma therapy, disseminated melanoma still lacks effective treatment, and recurrence of the tumor frequently occurs, even after high-dose chemotherapy. The mechanisms responsible for this chemoresistance or for the formation of new relapses remain poorly understood. Using a human 'model', in which the isolated limb is perfused with high doses of the chemotherapeutic melphalan (ILP), we identified a five-gene set (ATF3, CYR61, IER5, IL6, and PTGS2) of stress-induced genes that was consistently upregulated after ILP in all in-transit metastatic melanoma samples as well as in three melphalan-treated melanoma cell lines. Early post-ILP relapses retained these elevated expressions, whereas the expression of these genes returned to their original levels in late post-ILP recurrences. In addition, we identified upregulation of these genes in the A375 cell line's side population (SP) and melanospheres, established methods to enrich for candidate cancer stem cells (CSCs), which are considered chemoresistant and tumorigenic, and thus proposed to be responsible for tumor relapse. Our data identify an immediate and short-term upregulation of early stress-responsive genes that are potentially linked to chemoresistance and CSCs.
Collapse
Affiliation(s)
- Jasper Wouters
- Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
36
|
Lin J, Huo R, Wang L, Zhou Z, Sun Y, Shen B, Wang R, Li N. A novel anti-Cyr61 antibody inhibits breast cancer growth and metastasis in vivo. Cancer Immunol Immunother 2012; 61:677-87. [PMID: 22048717 PMCID: PMC11028480 DOI: 10.1007/s00262-011-1135-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 10/18/2011] [Indexed: 01/16/2023]
Abstract
Cysteine-rich protein 61(CCN1/Cyr61) has been implicated as an important mediator in proliferation and metastasis of breast cancer, which indicated that blockage of Cyr61 might be a potent target for breast cancer treatment. However, the antitumor effect of anti-Cyr61 antibodies on breast cancer in vivo has not been reported so far. In this study, we reported the effect and likely mechanism of generated anti-human Cyr61 monoclonal antibodies (mAb) on Cyr61 high expression line MDA-MB-231, known as a highly malignant and invasive human breast cancer cell line, at aspects of proliferation and migration in vitro and in vivo. We found the mAb, denoted as 093G9, revealed inhibitory effects on MDA-MB-231 cell proliferation, migration, and invasion through downregulation of both AKT and ERK phosphorylation in vitro compared with its isotype control. 093G9 also showed significant efficacy on suppressing primary tumor growth and spontaneous lymph node metastasis in in vivo mouse model. The specific epitope recognized by 093G9 was identified to be (140)LPNLGCP(146), adjacent to the VWC domain of Cyr61 by Ph.D.-C7C phage library display system. Our study provides direct evidence that Cyr61 can be a potent therapeutic target for patients who bear high Cyr61 expression breast cancer. Furthermore, the mAb, 093G9 developed in our laboratory, has shown a promising therapeutic characteristic in breast cancer.
Collapse
Affiliation(s)
- Jinpiao Lin
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Rongfen Huo
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Li Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhou Zhou
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Yue Sun
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Baihua Shen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| | - Rongfang Wang
- Diasys Diagnostic Systems (Shanghai) Co., Ltd, Shanghai International Medical Zone Building 16, Lane 588, Tianxiong Road, Shanghai, 201318 China
| | - Ningli Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 China
| |
Collapse
|
37
|
EphB2 receptor controls proliferation/migration dichotomy of glioblastoma by interacting with focal adhesion kinase. Oncogene 2012; 31:5132-43. [PMID: 22310282 PMCID: PMC3349801 DOI: 10.1038/onc.2012.16] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Glioblastoma multiforme (GBM) are the most frequent and aggressive primary brain tumors in adults. Uncontrolled proliferation and abnormal cell migration are two prominent spatially and temporally disassociated characteristics of GBMs. In this study, we investigated the role of the receptor tyrosine kinase EphB2 in controlling the proliferation/migration dichotomy of GBM. We studied EphB2 gain-of-function and loss-of function in glioblastoma-derived stem-like neurospheres (GBM-SCs), whose in vivo growth pattern closely replicates human GBM. EphB2 expression stimulated GBM neurosphere cell migration and invasion, and inhibited neurosphere cell proliferation in vitro. In parallel, EphB2 silencing increased tumor cell proliferation and decreased tumor cell migration. EphB2 was found to increase tumor cell invasion in vivo using an internally controlled dual-fluorescent xenograft model. Xenografts derived from EphB2 overexpressing GBM neurospheres also showed decreased cellular proliferation. The non-receptor tyrosine kinase focal adhesion kinase (FAK) was found to be co-associated with and highly activated by EphB2 expression and FAK activation facilitated focal adhesion formation, cytoskeleton structure change and cell migration in EphB2-expression GBM neurosphere cells. Taken together, our findings indicate that EphB2 has pro-invasive and anti-proliferative actions in GBM stem-like neurospheres mediated, in part, by interactions between EphB2 receptors and FAK. These novel findings suggest that tumor cell invasion can be therapeutically targeted by inhibiting EphB2 signaling and that optimal anti-tumor responses to EphB2 targeting may require the concurrent use of anti-proliferative agents.
Collapse
|
38
|
Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nat Rev Drug Discov 2011; 10:945-63. [PMID: 22129992 DOI: 10.1038/nrd3599] [Citation(s) in RCA: 513] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Members of the CCN family of matricellular proteins are crucial for embryonic development and have important roles in inflammation, wound healing and injury repair in adulthood. Deregulation of CCN protein expression or activities contributes to the pathobiology of various diseases - many of which may arise when inflammation or tissue injury becomes chronic - including fibrosis, atherosclerosis, arthritis and cancer, as well as diabetic nephropathy and retinopathy. Emerging studies indicate that targeting CCN protein expression or signalling pathways holds promise in the development of diagnostics and therapeutics for such diseases. This Review summarizes the biology of CCN proteins, their roles in various pathologies and their potential as therapeutic targets.
Collapse
|
39
|
Uddin S, Hussain AR, Ahmed M, Al-Sanea N, Abduljabbar A, Ashari LH, Alhomoud S, Al-Dayel F, Bavi P, Al-Kuraya KS. Coexpression of activated c-Met and death receptor 5 predicts better survival in colorectal carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:3032-3044. [PMID: 21978492 PMCID: PMC3260832 DOI: 10.1016/j.ajpath.2011.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2011] [Revised: 08/03/2011] [Accepted: 08/09/2011] [Indexed: 02/04/2023]
Abstract
Dysregulated overexpression of hepatocyte growth factor and its receptor, c-Met, has been reported in various cancers, but its role in colorectal carcinoma (CRC) has not been elucidated. Therefore, we investigated the role of phosphorylated Met (p-Met) in Middle Eastern CRC patient samples and cell lines. The p-Met was overexpressed in 80.8% of CRCs and strongly associated with the expression of p-AKT, DR5, and Ki-67 by immunohistochemistry. Coexpression of p-Met and DR5 was seen in 53.1% of CRC cases and was associated with a less aggressive phenotype, characterized by a histological subtype of adenocarcinomas, well-differentiated tumors, and was an independent prognostic marker for better overall survival. PHA665752, a selective p-Met inhibitor, induced apoptosis in CRC cells via inactivation of c-Met and AKT. PHA665752 treatment also caused increased expression of DR5 via generation of reactive oxygen species, and combination treatment with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and PHA665752 induced significant apoptosis. In vivo, cotreatment of a CRC xenograft with PHA665752 and TRAIL significantly reduced tumor volume and weight. These data demonstrate a significant correlation between p-Met and DR5 in patients with CRC. Furthermore, inhibition of p-Met signaling by PHA665752 in combination with TRAIL significantly inhibited cell growth and induced apoptosis in CRC cell lines, suggesting that this may have significant clinical implications as a therapeutic target in the treatment of CRC.
Collapse
Affiliation(s)
- Shahab Uddin
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Azhar R. Hussain
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Maqbool Ahmed
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nasser Al-Sanea
- Colorectal Unit, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Alaa Abduljabbar
- Colorectal Unit, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Luai H. Ashari
- Colorectal Unit, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Samar Alhomoud
- Colorectal Unit, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fouad Al-Dayel
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Prashant Bavi
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Khawla S. Al-Kuraya
- Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
40
|
Hepatocyte growth factor promoting the proliferation of human eccrine sweat gland epithelial cells is relative to AKT signal channel and β-catenin. Arch Dermatol Res 2011; 304:23-9. [DOI: 10.1007/s00403-011-1167-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 08/01/2011] [Indexed: 11/25/2022]
|
41
|
Lau LF. CCN1/CYR61: the very model of a modern matricellular protein. Cell Mol Life Sci 2011; 68:3149-63. [PMID: 21805345 DOI: 10.1007/s00018-011-0778-3] [Citation(s) in RCA: 252] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 07/19/2011] [Accepted: 07/19/2011] [Indexed: 02/08/2023]
Abstract
CCN1 (CYR61) is a dynamically expressed, multifunctional matricellular protein that plays essential roles in cardiovascular development during embryogenesis, and regulates inflammation, wound healing and fibrogenesis in the adult. Aberrant CCN1 expression is associated with myriad pathologies, including various cancers and diseases associated with chronic inflammation. CCN1 promotes diverse and sometimes opposing cellular responses, which can be ascribed, as least in part, to disparate activities mediated through its direct binding to distinct integrins in different cell types and contexts. Accordingly, CCN1 promotes cell proliferation, survival and angiogenesis by binding to integrin α(v)β(3), and induces apoptosis and senescence through integrin α(6)β(1) and heparan sulfate proteoglycans. The ability of CCN1 to trigger the accumulation of a robust and sustained level of reactive oxygen species underlies some of its unique activities as a matrix cell-adhesion molecule. Emerging studies suggest that CCN1 might be useful as a biomarker or therapeutic target in certain diseases.
Collapse
Affiliation(s)
- Lester F Lau
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, 900 S. Ashland Avenue, Chicago, IL 60607, USA.
| |
Collapse
|
42
|
Regulation of glioblastoma stem cells by retinoic acid: role for Notch pathway inhibition. Oncogene 2011; 30:3454-67. [PMID: 21383690 DOI: 10.1038/onc.2011.58] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is necessary to understand mechanisms by which differentiating agents influence tumor-initiating cancer stem cells. Toward this end, we investigated the cellular and molecular responses of glioblastoma stem-like cells (GBM-SCs) to all-trans retinoic acid (RA). GBM-SCs were grown as non-adherent neurospheres in growth factor supplemented serum-free medium. RA treatment rapidly induced morphology changes, induced growth arrest at G1/G0 to S transition, decreased cyclin D1 expression and increased p27 expression. Immunofluorescence and western blot analysis indicated that RA induced the expression of lineage-specific differentiation markers Tuj1 and GFAP and reduced the expression of neural stem cell markers such as CD133, Msi-1, nestin and Sox-2. RA treatment dramatically decreased neurosphere-forming capacity, inhibited the ability of neurospheres to form colonies in soft agar and inhibited their capacity to propagate subcutaneous and intracranial xenografts. Expression microarray analysis identified ∼350 genes that were altered within 48 h of RA treatment. Affected pathways included retinoid signaling and metabolism, cell-cycle regulation, lineage determination, cell adhesion, cell-matrix interaction and cytoskeleton remodeling. Notch signaling was the most prominent of these RA-responsive pathways. Notch pathway downregulation was confirmed based on the downregulation of HES and HEY family members. Constitutive activation of Notch signaling with the Notch intracellular domain rescued GBM neurospheres from the RA-induced differentiation and stem cell depletion. Our findings identify mechanisms by which RA targets GBM-derived stem-like tumor-initiating cells and novel targets applicable to differentiation therapies for glioblastoma.
Collapse
|