1
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
2
|
Ungefroren H, von der Ohe J, Braun R, Gätje Y, Lapshyna O, Schrader J, Lehnert H, Marquardt JU, Konukiewitz B, Hass R. Characterization of Epithelial-Mesenchymal and Neuroendocrine Differentiation States in Pancreatic and Small Cell Ovarian Tumor Cells and Their Modulation by TGF-β1 and BMP-7. Cells 2024; 13:2010. [PMID: 39682758 PMCID: PMC11640004 DOI: 10.3390/cells13232010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an extremely poor prognosis, due in part to early invasion and metastasis, which in turn involves epithelial-mesenchymal transition (EMT) of the cancer cells. Prompted by the discovery that two PDAC cell lines of the quasi-mesenchymal subtype (PANC-1, MIA PaCa-2) exhibit neuroendocrine differentiation (NED), we asked whether NED is associated with EMT. Using real-time PCR and immunoblotting, we initially verified endogenous expressions of various NED markers, i.e., chromogranin A (CHGA), synaptophysin (SYP), somatostatin receptor 2 (SSTR2), and SSTR5 in PANC-1 and MIA PaCa-2 cells. By means of immunohistochemistry, the expressions of CHGA, SYP, SSTR2, and the EMT markers cytokeratin 7 (CK7) and vimentin could be allocated to the neoplastic ductal epithelial cells of pancreatic ducts in surgically resected tissues from patients with PDAC. In HPDE6c7 normal pancreatic duct epithelial cells and in epithelial subtype BxPC-3 PDAC cells, the expression of CHGA, SYP, and neuron-specific enolase 2 (NSE) was either undetectable or much lower than in PANC-1 and MIA PaCa-2 cells. Parental cultures of PANC-1 cells exhibit EM plasticity (EMP) and harbor clonal subpopulations with both M- and E-phenotypes. Of note, M-type clones were found to display more pronounced NED than E-type clones. Inducing EMT in parental cultures of PANC-1 cells by treatment with transforming growth factor-β1 (TGF-β1) repressed epithelial genes and co-induced mesenchymal and NED genes, except for SSTR5. Surprisingly, treatment with bone morphogenetic protein (BMP)-7 differentially affected gene expressions in PANC-1, MIA PaCa-2, BxPC-3, and HPDE cells. It synergized with TGF-β1 in the induction of vimentin, SNAIL, SSTR2, and NSE but antagonized it in the regulation of CHGA and SSTR5. Phospho-immunoblotting in M- and E-type PANC-1 clones revealed that both TGF-β1 and, surprisingly, also BMP-7 activated SMAD2 and SMAD3 and that in M- but not E-type clones BMP-7 was able to dramatically enhance the activation of SMAD3. From these data, we conclude that in EMT of PDAC cells mesenchymal and NED markers are co-regulated, and that mesenchymal-epithelial transition (MET) is associated with a loss of both the mesenchymal and NED phenotypes. Analyzing NED in another tumor type, small cell carcinoma of the ovary hypercalcemic type (SCCOHT), revealed that two model cell lines of this disease (SCCOHT-1, BIN-67) do express CDH1, SNAI1, VIM, CHGA, SYP, ENO2, and SSTR2, but that in contrast to BMP-7, none of these genes was transcriptionally regulated by TGF-β1. Likewise, in BIN-67 cells, BMP-7 was able to reduce proliferation, while in SCCOHT-1 cells this occurred only upon combined treatment with TGF-β and BMP-7. We conclude that in PDAC-derived tumor cells, NED is closely linked to EMT and TGF-β signaling, which may have implications for the therapeutic use of TGF-β inhibitors in PDAC management.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| | - Rüdiger Braun
- Department of Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Yola Gätje
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Olha Lapshyna
- Department of Surgery, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Jörg Schrader
- First Department of Medicine, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Hendrik Lehnert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire (UHCW), Coventry CV2 2DX, UK
| | - Jens-Uwe Marquardt
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | - Björn Konukiewitz
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
3
|
Verma P, Shukla N, Kumari S, Ansari M, Gautam NK, Patel GK. Cancer stem cell in prostate cancer progression, metastasis and therapy resistance. Biochim Biophys Acta Rev Cancer 2023; 1878:188887. [PMID: 36997008 DOI: 10.1016/j.bbcan.2023.188887] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/18/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
Prostate cancer (PCa) is the most diagnosed malignancy in the men worldwide. Cancer stem cells (CSCs) are the sub-population of cells present in the tumor which possess unique properties of self-renewal and multilineage differentiation thus thought to be major cause of therapy resistance, disease relapse, and mortality in several malignancies including PCa. CSCs have also been shown positive for the common stem cells markers such as ALDH EZH2, OCT4, SOX2, c-MYC, Nanog etc. Therefore, isolation and characterization of CSCs specific markers which may discriminate CSCs and normal stem cells are critical to selectively eliminate CSCs. Rapid advances in the field offers a theoretical explanation for many of the enduring uncertainties encompassing the etiology and an optimism for the identification of new stem-cell targets, development of reliable and efficient therapies in the future. The emerging reports have also provided unprecedented insights into CSCs plasticity, quiescence, renewal, and therapeutic response. In this review, we discuss the identification of PCa stem cells, their unique properties, stemness-driving pathways, new diagnostics, and therapeutic interventions.
Collapse
|
4
|
Hachana S, Larrivée B. TGF-β Superfamily Signaling in the Eye: Implications for Ocular Pathologies. Cells 2022; 11:2336. [PMID: 35954181 PMCID: PMC9367584 DOI: 10.3390/cells11152336] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/06/2023] Open
Abstract
The TGF-β signaling pathway plays a crucial role in several key aspects of development and tissue homeostasis. TGF-β ligands and their mediators have been shown to be important regulators of ocular physiology and their dysregulation has been described in several eye pathologies. TGF-β signaling participates in regulating several key developmental processes in the eye, including angiogenesis and neurogenesis. Inadequate TGF-β signaling has been associated with defective angiogenesis, vascular barrier function, unfavorable inflammatory responses, and tissue fibrosis. In addition, experimental models of corneal neovascularization, diabetic retinopathy, proliferative vitreoretinopathy, glaucoma, or corneal injury suggest that aberrant TGF-β signaling may contribute to the pathological features of these conditions, showing the potential of modulating TGF-β signaling to treat eye diseases. This review highlights the key roles of TGF-β family members in ocular physiology and in eye diseases, and reviews approaches targeting the TGF-β signaling as potential treatment options.
Collapse
Affiliation(s)
- Soumaya Hachana
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Bruno Larrivée
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
5
|
Sharma T, Kapoor A, Mandal CC. Duality of bone morphogenetic proteins in cancer: A comprehensive analysis. J Cell Physiol 2022; 237:3127-3163. [DOI: 10.1002/jcp.30785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Tanu Sharma
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| | - Anmol Kapoor
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| | - Chandi C. Mandal
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| |
Collapse
|
6
|
Aashaq S, Batool A, Mir SA, Beigh MA, Andrabi KI, Shah ZA. TGF-β signaling: A recap of SMAD-independent and SMAD-dependent pathways. J Cell Physiol 2021; 237:59-85. [PMID: 34286853 DOI: 10.1002/jcp.30529] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/06/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022]
Abstract
Transforming growth factor-β (TGF-β) is a proinflammatory cytokine known to control a diverse array of pathological and physiological conditions during normal development and tumorigenesis. TGF-β-mediated physiological effects are heterogeneous and vary among different types of cells and environmental conditions. TGF-β serves as an antiproliferative agent and inhibits tumor development during primary stages of tumor progression; however, during the later stages, it encourages tumor development and mediates metastatic progression and chemoresistance. The fundamental elements of TGF-β signaling have been divulged more than a decade ago; however, the process by which the signals are relayed from cell surface to nucleus is very complex with additional layers added in tumor cell niches. Although the intricate understanding of TGF-β-mediated signaling pathways and their regulation are still evolving, we tried to make an attempt to summarize the TGF-β-mediated SMAD-dependent andSMAD-independent pathways. This manuscript emphasizes the functions of TGF-β as a metastatic promoter and tumor suppressor during the later and initial phases of tumor progression respectively.
Collapse
Affiliation(s)
- Sabreena Aashaq
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Soura, Srinagar, JK, India
| | - Asiya Batool
- Division of Cancer Pharmacology, Indian Institute of Integrative Medicine, Srinagar, JK, India
| | | | | | | | - Zaffar Amin Shah
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Soura, Srinagar, JK, India
| |
Collapse
|
7
|
Yang B, Damodaran S, Khemees TA, Filon MJ, Schultz A, Gawdzik J, Etheridge T, Malin D, Richards KA, Cryns VL, Jarrard DF. Synthetic Lethal Metabolic Targeting of Androgen-Deprived Prostate Cancer Cells with Metformin. Mol Cancer Ther 2020; 19:2278-2287. [DOI: 10.1158/1535-7163.mct-19-1141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/28/2020] [Accepted: 09/01/2020] [Indexed: 11/16/2022]
|
8
|
Zhang Y, Zhang M, Xie W, Wan J, Tao X, Liu M, Zhen Y, Lin F, Wu B, Zhai Z, Wang C. Gremlin-1 is a key regulator of endothelial-to-mesenchymal transition in human pulmonary artery endothelial cells. Exp Cell Res 2020; 390:111941. [PMID: 32145252 DOI: 10.1016/j.yexcr.2020.111941] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/10/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Endothelial-to-mesenchymal transition (EndMT) has been implicated in initiation and progression of pulmonary arterial hypertension (PAH). Gremlin-1 promotes vascular remodeling of PAH and mediates epithelial-mesenchymal transition, which is similar to EndMT. In the present study we investigated the potential role of gremlin-1 plays in EndMT of pulmonary artery endothelial cells (PAECs). METHODS Immunofluorescence staining was performed to detect the expression of alpha smooth muscle actin (α-SMA) and von Willebrand factor (VWF). Migration and angiogenic responses of PAECs were determined by transwell assay and tube formation assay, respectively. Protein expression levels were determined by western blotting. RESULTS Gremlin-1 induced EndMT of PAECs in a phospho-smad2/3-dependent manner. This was characterized by the loss of platelet endothelial cell adhesion molecule 1 and an increase in protein levels of a-SMA, nerve-cadherin, and matrix metalloproteinase 2. It was also determined that gremlin-1 facilitated the migration and angiogenic responses of PAECs in a dose-dependent manner. Bone morphogenetic protein 7 (BMP-7) was found to attenuate gremlin-1-mediated EndMT, migration and angiogenesis of PAECs by inducing phosphorylation of Smad1/5/8 and suppressing phosphorylation of Smad2/3. CONCLUSION Gremlin-1 mediates EndMT in PAECs, and BMP-7 reverses gremlin-1-induced EndMT by an induction of p-Smad1/5/8 and suppression of p-Smad2/3.
Collapse
Affiliation(s)
- Yunxia Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; National Clinical Research Center for Respiratory Diseases, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Meng Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, NO 2, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Wanmu Xie
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; National Clinical Research Center for Respiratory Diseases, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Jun Wan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; National Clinical Research Center for Respiratory Diseases, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Xincao Tao
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; National Clinical Research Center for Respiratory Diseases, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Min Liu
- Department of Radiology, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Yanan Zhen
- Division of Cardiovascular Surgery, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Fan Lin
- Division of Cardiovascular Surgery, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Bo Wu
- Department of Lung Transplantation, the People's Hospital of Wuxi, 299 Qingyang Rd, Wuxi, 214023, China
| | - Zhenguo Zhai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; National Clinical Research Center for Respiratory Diseases, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China.
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; National Clinical Research Center for Respiratory Diseases, NO 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China; Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
9
|
Miura K, Hamanaka K, Uehara T, Momose M, Kanai Y, Matsuoka S, Takeda T, Agatsuma H, Hyogotani A, Ito KI. Bone morphogenetic protein-7 expression reflects the high proliferative ability and aggressiveness of thymic epithelial tumors. J Thorac Dis 2020; 12:448-456. [PMID: 32274111 PMCID: PMC7139077 DOI: 10.21037/jtd.2020.01.14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Bone morphogenetic protein-7 (BMP-7) is a transforming growth factor-β superfamily member. We examined whether BMP-7 expression in thymic epithelial tumors is associated with their clinicopathological features. Methods One hundred and thirty-two clinical specimens were analyzed in this study. The expression of BMP-7 was detected using immunohistochemistry and was scored as 0, 1, 2, or 3 according to its intensity and was then classified as negative (score 0 and 1) or positive (2 and 3). In addition, Ki-67 staining was performed in type B3 thymoma and thymic cancer. Results The positive ratio of BMP-7 was 80% in thymic cancer and 70% in thymoma type B3. In contrast, the positive ratios of BMP-7 in type B2 (29.1%), B1 (3.7%), AB (26%), and A (31%) were relatively low. The mean Ki-67 labeling index of the BMP-7 positive group (10.1%±5.9%) was significantly higher than that of the BMP-7 negative group (4.9%±5.9%) in type B3 thymoma and thymic cancer (P=0.012). The BMP-7 positive group showed significantly poorer overall survival (OS) than the BMP-7 negative group across all patients with thymic epithelial tumors and in all types of thymomas (P=0.006, P=0.018); however, no difference was observed in thymic cancers. Conclusions This study showed that high expression of BMP-7 correlated with a poor prognosis in patients with thymic epithelial tumors, and the expression of BMP-7 was higher in type B3 thymomas and thymic cancers than in other types of thymomas. BMP-7 might serve as a novel prognostic biomarker for thymic epithelial tumors.
Collapse
Affiliation(s)
- Kentaro Miura
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Kazutoshi Hamanaka
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Tsuyoshi Uehara
- Department of Laboratory Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Masanobu Momose
- Department of Laboratory Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Yukiko Kanai
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Shunichiro Matsuoka
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Tetsu Takeda
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Hiroyuki Agatsuma
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Akira Hyogotani
- Division of General Thoracic Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Ken-ichi Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| |
Collapse
|
10
|
Sun R, Guan H, Liu W, Liang J, Wang F, Li C. Expression of BMP7 in cervical cancer and inhibition of epithelial‑mesenchymal transition by BMP7 knockdown in HeLa cells. Int J Mol Med 2020; 45:1417-1424. [PMID: 32323730 PMCID: PMC7138274 DOI: 10.3892/ijmm.2020.4519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 02/11/2020] [Indexed: 12/09/2022] Open
Abstract
The aim of the present study was to investigate the expression of bone morphogenetic protein 7 (BMP7) in cervical cancer tissues, the effect of BMP7 on the proliferation, migration and epithelial-mesenchymal transition (EMT) of cervical cancer HeLa cells and the possible mechanism involved. Immunohistochemistry was used to stain the cervical cancer tissues and benign or precancerous lesions. Lentivirus containing BMP7 knockdown was transfected in HeLa cells and western blotting was performed to analyze BMP7 expression. At the same time, the influence of BMP7 knockdown on the expression of phosphorylated (p)-mothers against decapentaplegic homolog 1/5/9 and EMT-related markers [epithelial-cadherin, neural-cadherin, Vimentin, Snail and Slug] was detected. Cell Counting Kit-8 was used to detect cell proliferation. Transwell migration and invasion assays were performed to measure cell invasion and migration. The cell cycle was detected by flow cytometry. Compared with normal cervical epithelial and paracancerous cells, the positive rate of BMP7 expression in cervical cancer tissues was significantly increased. As compared with the control group, the expression of BMP7 was decreased in HeLa cells transfected with lentivirus. The knockdown of BMP7 in cervical cancer HeLa cells inhibited cell proliferation, migration and invasion, resulted in G1 cell cycle arrest and reversed the EMT process. In addition, the expression of p-Smad1/5/9 was significantly decreased in HeLa cells with BMP7 knockdown. BMP7 is expected to be a possible target for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Rui Sun
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hongwei Guan
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Wei Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Junhui Liang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Fei Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Changzhong Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
11
|
Semina EV, Rubina KA, Shmakova AA, Rysenkova KD, Klimovich PS, Aleksanrushkina NA, Sysoeva VY, Karagyaur MN, Tkachuk VA. Downregulation of uPAR promotes urokinase translocation into the nucleus and epithelial to mesenchymal transition in neuroblastoma. J Cell Physiol 2020; 235:6268-6286. [PMID: 31990070 PMCID: PMC7318179 DOI: 10.1002/jcp.29555] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/13/2020] [Indexed: 12/16/2022]
Abstract
The urokinase system is involved in a variety of physiological processes, such as fibrinolysis, matrix remodeling, wound healing, and regeneration. Upon binding to its cognate receptor urokinase‐type plasminogen activator receptor (uPAR), urokinase‐type plasminogen activator (uPA) catalyzes the conversion of plasminogen to plasmin and the activation of matrix metalloproteases. Apart from this, uPA–uPAR interaction can lead to the activation of transcription factors, mitogen‐activated protein kinase signaling pathways and RTK cascades. Elevated expression of uPA and uPAR is markedly associated with cancer progression and metastasis and correlates with a poor prognosis in clinics. Targeting the urokinase system has proved to be effective in experimental models in vitro and in vivo, however, in clinics the inhibition of the uPA/uPAR system has fallen short of expectations, suggesting that the question of the functional relevance of uPA/uPAR system is far from being moot. Recently, using CRISPR/Cas9 technology, we have shown that uPAR knockout decreases the proliferation of neuroblastoma Neuro2a cells in vitro. In the present study we demonstrate that uPAR expression is essential for maintaining the epithelial phenotype in Neuro2a cells and that uPAR silencing promotes epithelial‐mesenchymal transition (EMT) and increased cell migration. Accordingly, uPAR knockout results in the downregulation of epithelial markers (E‐cadherin, occludin, and claudin‐5) and in the increase of mesenchymal markers (N‐cadherin, α‐smooth muscle actin, and interleukin‐6). In search of the molecular mechanism underlying these changes, we identified uPA as a key component. Two key insights emerged as a result of this work: in the absence of uPAR, uPA is translocated into the nucleus where it is presumably involved in the activation of transcription factors (nuclear factor κB and Snail) resulting in EMT. In uPAR‐expressing cells, uPAR functions as a uPA “trap” that binds uPA on the cell surface and promotes controlled uPA internalization and degradation in lysosomes.
Collapse
Affiliation(s)
- Ekaterina V Semina
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| | - Kseniya A Rubina
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Morohogenesis and Tissue Reparation, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Anna A Shmakova
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| | - Karina D Rysenkova
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| | - Polina S Klimovich
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| | - Natalya A Aleksanrushkina
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Veronika Y Sysoeva
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maxim N Karagyaur
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Vsevolod A Tkachuk
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
12
|
Dituri F, Cossu C, Mancarella S, Giannelli G. The Interactivity between TGFβ and BMP Signaling in Organogenesis, Fibrosis, and Cancer. Cells 2019; 8:E1130. [PMID: 31547567 PMCID: PMC6829314 DOI: 10.3390/cells8101130] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022] Open
Abstract
The Transforming Growth Factor beta (TGFβ) and Bone Morphogenic Protein (BMP) pathways intersect at multiple signaling hubs and cooperatively or counteractively participate to bring about cellular processes which are critical not only for tissue morphogenesis and organogenesis during development, but also for adult tissue homeostasis. The proper functioning of the TGFβ/BMP pathway depends on its communication with other signaling pathways and any deregulation leads to developmental defects or diseases, including fibrosis and cancer. In this review we explore the cellular and physio-pathological contexts in which the synergism or antagonism between the TGFβ and BMP pathways are crucial determinants for the normal developmental processes, as well as the progression of fibrosis and malignancies.
Collapse
Affiliation(s)
- Francesco Dituri
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Carla Cossu
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "S. De Bellis", Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| |
Collapse
|
13
|
Ma Q, Lu Y, Lin J, Gu Y. ENKUR acts as a tumor suppressor in lung adenocarcinoma cells through PI3K/Akt and MAPK/ERK signaling pathways. J Cancer 2019; 10:3975-3984. [PMID: 31417642 PMCID: PMC6692620 DOI: 10.7150/jca.30021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 05/25/2019] [Indexed: 02/07/2023] Open
Abstract
Lung adenocarcinoma (LAD) is the most common type of lung cancer with a high possibility of tumor relapse and metastasis. ENKUR (Enkurin) was originally identified as a potential regulator or effector of TRPC channels that directly binds to several TRPC proteins and the p85 subunit of PI3K. However, the role of ENKUR in cancer development has remained unclear. In this study we analyzed the expression profile of ENKUR mRNA in clinical LAD samples and examined ENKUR mRNA expression and ENKUR protein level in LAD cells. Significant down-regulated ENKUR expression was observed in clinical tumor tissues of LAD as well as in human LAD cells. To evaluate the effects of aberrant ENKUR expression on cellular biology of LAD cells in vitro and tumor growth in vivo, ENKUR-overexpressed and -silenced LAD cell lines were constructed using lentiviral vectors. Our results showed that overexpression of ENKUR in LAD cells inhibited cell proliferation, migration and invasion, while silencing of ENKUR led to the opposite effects. Silencing of ENKUR in LAD cells also promoted tumorigenesis in nude mice model and caused epithelial to mesenchymal transition (EMT). Furthermore, using western blot and co-immunoprecipitation analyses, we demonstrated that ENKUR interacts with PI3K directly and is possibly involved in the PI3K/Akt and MAPK/ERK signaling pathways.
Collapse
Affiliation(s)
- Qing Ma
- College of Biological and Environmental Engineering, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, P.R. China
| | - Yin Lu
- College of Biological and Environmental Engineering, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, P.R. China
| | - Jie Lin
- College of Biological and Environmental Engineering, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, P.R. China
| | - Ye Gu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
14
|
Ning J, Zhao Y, Ye Y, Yu J. Opposing roles and potential antagonistic mechanism between TGF-β and BMP pathways: Implications for cancer progression. EBioMedicine 2019; 41:702-710. [PMID: 30808576 PMCID: PMC6442991 DOI: 10.1016/j.ebiom.2019.02.033] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/05/2019] [Accepted: 02/15/2019] [Indexed: 02/08/2023] Open
Abstract
The transforming growth factor β (TGF-β) superfamily participates in tumour proliferation, apoptosis, differentiation, migration, invasion, immune evasion and extracellular matrix remodelling. Genetic deficiency in distinct components of TGF-β and BMP-induced signalling pathways or their excessive activation has been reported to regulate the development and progression of some cancers. As more in-depth studies about this superfamily have been conducted, more evidence suggests that the TGF-β and BMP pathways play an opposing role. The cross-talk of these 2 pathways has been widely studied in kidney disease and bone formation, and the opposing effects have also been observed in some cancers. However, the antagonistic mechanisms are still insufficiently investigated in cancer. In this review, we aim to display more evidences and possible mechanisms accounting for the antagonism between these 2 pathways, which might provide some clues for further study in cancer. Describe the basics of TGF-β and BMP signalling Summarize the potential mechanisms accounting for the antagonism between TGF-β and BMP pathways Provide some evidence about the antagonistic effects between pathways observed in some cancers
Collapse
Affiliation(s)
- Junya Ning
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China; Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Yi Zhao
- Key Laboratory of Intelligent Information Processing, Advanced Computer Research Center, State Key Laboratory of Computer Architecture, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, PR China
| | - Yingnan Ye
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China; Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
| |
Collapse
|
15
|
Ni J, Cozzi P, Beretov J, Duan W, Bucci J, Graham P, Li Y. Epithelial cell adhesion molecule (EpCAM) is involved in prostate cancer chemotherapy/radiotherapy response in vivo. BMC Cancer 2018; 18:1092. [PMID: 30419852 PMCID: PMC6233586 DOI: 10.1186/s12885-018-5010-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022] Open
Abstract
Background Development of chemo−/radioresistance is a major challenge for the current prostate cancer (CaP) therapy. We have previously demonstrated that epithelial cell adhesion molecule (EpCAM) is associated with CaP growth and therapeutic resistance in vitro, however, the role of EpCAM in CaP in vivo is not fully elucidated. Here, we aimed to investigate how expression of EpCAM is involved in CaP growth and chemo−/radiotherapy response in NOD/SCID mouse models in vivo and to validate its role as a therapeutic target for CaP therapy. Methods EpCAM was knocked down in PC-3 CaP cell line using short hairpin RNA (shRNA). The effect of EpCAM-knockdown (KD) on tumour growth, chemo−/radiotherapy response and animal survival was evaluated on subcutaneous (s.c) and orthotopic mouse models. Results We found that KD of EpCAM significantly inhibited tumour growth, increased xenograft sensitivity to chemotherapy/radiotherapy, and prolonged the survival of tumour-bearing mice. In addition, we demonstrated that KD of EpCAM is associated with downregulation of the PI3K/Akt/mTOR pathway. Conclusions In conclusion, our data confirms that CaP growth and chemo−/radioresistance in vivo is associated with over-expression of EpCAM, which serves both a functional biomarker and promising therapeutic target. Electronic supplementary material The online version of this article (10.1186/s12885-018-5010-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Ni
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Paul Cozzi
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia.,Department of Surgery, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Julia Beretov
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia.,Anatomical Pathology, NSW Health Pathology, St George Hospital, Gray St, Kogarah, NSW, 2217, Australia
| | - Wei Duan
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, VIC, 3216, Australia
| | - Joseph Bucci
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Peter Graham
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia. .,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia. .,School of Basic Medical Sciences, Zhengzhou University, Henan, 450001, China.
| |
Collapse
|
16
|
Liu RX, Ma Y, Hu XL, Ren WY, Liao YP, Wang H, Zhu JH, Wu K, He BC, Sun WJ. Anticancer effects of oridonin on colon cancer are mediated via BMP7/p38 MAPK/p53 signaling. Int J Oncol 2018; 53:2091-2101. [PMID: 30132514 DOI: 10.3892/ijo.2018.4527] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/07/2018] [Indexed: 11/06/2022] Open
Abstract
Colon cancer is a prevalent malignancy affecting the gastrointestinal tract. Oridonin (ORI) is a promising chemotherapeutic drug used in the treatment of colon cancer. In this study, we examined the anticancer activity of ORI against colon cancer and elucidated the underlying molecular mechanisms. Cell counting kit-8, flow cytometric and western blot analyses were conducted to analyze the growth inhibitory effects of ORI on SW620 cells; we employed BMP7 and p53 recombinant adenovirus to detect the influence of ORI on the p38 MAPK signal pathway; PT-qPCR, cell immunofluorescence staining and western blot analysis were used to detect the expression of BMP7, p38 and p-p38, p53 and p-p53. A xenograft tumor model and histological evaluation were introduced to detect the effects of ORI and BMP7 in SW620 cells in vivo. ORI inhibited the proliferation of SW620 cells and induced apoptosis. ORI also increased the total and phosphorylated levels of p53. The overexpression of p53 was found to enhance the anti-proliferative effects of ORI on the SW620 cells, while the inhibition of p53 partially reversed these effects. ORI increased the expression of bone morphogenetic protein 7 (BMP7) in the SW620 cells. The overexpression of BMP7 also enhanced the antiproliferative effects of ORI on the SW620 cells and reduced the growth rate of tumors in mice. BMP7-induced immunosuppression markedly decreased the anti-proliferative effects of ORI. ORI was not found to exert any substantial effect on the phosphorylation levels of Smad1/5/8, although it increased the level of p-p38 significantly. The inhibition of p38 significantly attenuated the ORI-induced increase in the levels of p-p53. The overexpression of BMP7 enhanced the promoting effects of ORI on the p-p53 and p-p38 levels, while BMP7-induced immunosuppression reduced the effects of ORI on p-p38 and p-p53. On the whole, the findings of this study suggest that ORI may be a promising agent for use in the treatment of colon cancer, and the anticancer effects of ORI may be partially mediated through the BMP7/p38 MAPK/p53 signaling pathway.
Collapse
Affiliation(s)
- Rong-Xing Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yan Ma
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xue-Lian Hu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wen-Yan Ren
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yun-Peng Liao
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Han Wang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jia-Hui Zhu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ke Wu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Bai-Cheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wen-Juan Sun
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
17
|
Kowtharapu BS, Prakasam RK, Murín R, Koczan D, Stahnke T, Wree A, Jünemann AGM, Stachs O. Role of Bone Morphogenetic Protein 7 (BMP7) in the Modulation of Corneal Stromal and Epithelial Cell Functions. Int J Mol Sci 2018; 19:ijms19051415. [PMID: 29747422 PMCID: PMC5983782 DOI: 10.3390/ijms19051415] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/02/2018] [Accepted: 05/07/2018] [Indexed: 12/20/2022] Open
Abstract
In the cornea, healing of the wounded avascular surface is an intricate process comprising the involvement of epithelial, stromal and neuronal cell interactions. These interactions result to the release of various growth factors that play prominent roles during corneal wound healing response. Bone morphogenetic proteins (BMPs) are unique multi-functional potent growth factors of the transforming growth factor-beta (TGF-β) superfamily. Treatment of corneal epithelial cells with substance P and nerve growth factor resulted to an increase in the expression of BMP7 mRNA. Since BMP7 is known to modulate the process of corneal wound healing, in this present study, we investigated the influence of exogenous rhBMP7 on human corneal epithelial cell and stromal cell (SFs) function. To obtain a high-fidelity expression profiling of activated biomarkers and pathways, transcriptome-wide gene-level expression profiling of epithelial cells in the presence of BMP7 was performed. Gene ontology analysis shows BMP7 stimulation activated TGF-β signaling and cell cycle pathways, whereas biological processes related to cell cycle, microtubule and intermediate filament cytoskeleton organization were significantly impacted in corneal epithelial cells. Scratch wound healing assay showed increased motility and migration of BMP7 treated epithelial cells. BMP7 stimulation studies show activation of MAPK cascade proteins in epithelial cells and SFs. Similarly, a difference in the expression of claudin, Zink finger E-box-binding homeobox 1 was observed along with phosphorylation levels of cofilin in epithelial cells. Stimulation of SFs with BMP7 activated them with increased expression of α-smooth muscle actin. In addition, an elevated phosphorylation of epidermal growth factor receptor following BMP7 stimulation was also observed both in corneal epithelial cells and SFs. Based on our transcriptome analysis data on epithelial cells and the results obtained in SFs, we conclude that BMP7 contributes to epithelial-to-mesenchymal transition-like responses and plays a role equivalent to TGF-β in the course of corneal wound healing.
Collapse
Affiliation(s)
- Bhavani S Kowtharapu
- Department of Ophthalmology, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Ruby Kala Prakasam
- Department of Ophthalmology, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Radovan Murín
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia.
| | - Dirk Koczan
- Institute for Immunology, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Thomas Stahnke
- Department of Ophthalmology, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Andreas Wree
- Institute for Anatomy, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Anselm G M Jünemann
- Department of Ophthalmology, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Oliver Stachs
- Department of Ophthalmology, Rostock University Medical Center, 18057 Rostock, Germany.
| |
Collapse
|
18
|
Zhang L, Ye Y, Long X, Xiao P, Ren X, Yu J. BMP signaling and its paradoxical effects in tumorigenesis and dissemination. Oncotarget 2018; 7:78206-78218. [PMID: 27661009 PMCID: PMC5363655 DOI: 10.18632/oncotarget.12151] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/14/2016] [Indexed: 01/04/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) play important roles in embryonic and postnatal development by regulating cell differentiation, proliferation, motility, and survival, thus maintaining homeostasis during organ and tissue development. BMPs can lead to tumorigenesis and regulate cancer progression in different stages. Therefore, we summarized studies on BMP expression, the clinical significance of BMP dysfunction in various cancer types, and the molecular regulation of various BMP-related signaling pathways. We emphasized on the paradoxical effects of BMPs on various aspects of carcinogenesis, including epithelial–mesenchymal transition (EMT), cancer stem cells (CSCs), and angiogenesis. We also reviewed the molecular mechanisms by which BMPs regulate tumor generation and progression as well as potential therapeutic targets against BMPs that might be valuable in preventing tumor growth and invasion.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Yingnan Ye
- Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Xinxin Long
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Pei Xiao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Jinpu Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China.,Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin, P. R. China
| |
Collapse
|
19
|
Lo UG, Lee CF, Lee MS, Hsieh JT. The Role and Mechanism of Epithelial-to-Mesenchymal Transition in Prostate Cancer Progression. Int J Mol Sci 2017; 18:ijms18102079. [PMID: 28973968 PMCID: PMC5666761 DOI: 10.3390/ijms18102079] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/21/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
In prostate cancer (PCa), similar to many other cancers, distant organ metastasis symbolizes the beginning of the end disease, which eventually leads to cancer death. Many mechanisms have been identified in this process that can be rationalized into targeted therapy. Among them, epithelial-to-mesenchymal transition (EMT) is originally characterized as a critical step for cell trans-differentiation during embryo development and now recognized in promoting cancer cells invasiveness because of high mobility and migratory abilities of mesenchymal cells once converted from carcinoma cells. Nevertheless, the underlying pathways leading to EMT appear to be very diverse in different cancer types, which certainly represent a challenge for developing effective intervention. In this article, we have carefully reviewed the key factors involved in EMT of PCa with clinical correlation in hope to facilitate the development of new therapeutic strategy that is expected to reduce the disease mortality.
Collapse
Affiliation(s)
- U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Cheng-Fan Lee
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Ming-Shyue Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
20
|
Shirai K, Tanaka SI, Lovicu FJ, Saika S. The murine lens: A model to investigate in vivo epithelial-mesenchymal transition. Dev Dyn 2017; 247:340-345. [PMID: 28480986 PMCID: PMC5836960 DOI: 10.1002/dvdy.24518] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/09/2017] [Accepted: 04/24/2017] [Indexed: 12/21/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) produces myofibroblasts that contribute to the formation of fibrotic tissue with an impairment of tissue homeostasis and functionality. The crystalline lens of the eye is a unique transparent and isolated tissue. The lens vesicle becomes isolated from the surface ectoderm, its cells are all contained as they line the inner surface of the lens capsule. Clinically the formation of fibrotic tissue by the lens epithelial cells causes a type of cataract or opacification and contraction of the lens capsule postcataract surgery. Production of EMT in the intact animal lens by using specific gene transfer to the lens or experimental lens injury has been shown to be a powerful tool to investigate EMT processes. It is not easy to uncover whether the origin of the myofibroblast is epithelial cell‐derived or from other cell lineages in fibrotic tissues. However, myofibroblasts that appear in the crystalline lens pathology are totally derived from the lens epithelial cells for the reasons mentioned above. Here, we report on different animal models of lens EMT, using either transgenic approaches or injury to study the biological aspects of EMT. Developmental Dynamics 247:340–345, 2018. © 2017 The Authors Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists Lens, Epithelial‐mesenchymal transition, mice.
Collapse
Affiliation(s)
- Kumi Shirai
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Sai-Ichi Tanaka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Frank J Lovicu
- Save Sight Institute and Discipline of Anatomy and Histology, Bosch Institute, School of Medical Sciences, The University of Sydney, NSW, Australia
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
21
|
Abstract
Transforming growth factor β (TGF-β) and structurally related factors use several intracellular signaling pathways in addition to Smad signaling to regulate a wide array of cellular functions. These non-Smad signaling pathways are activated directly by ligand-occupied receptors to reinforce, attenuate, or otherwise modulate downstream cellular responses. This review summarizes the current knowledge of the mechanisms by which non-Smad signaling pathways are directly activated in response to ligand binding, how activation of these pathways impinges on Smads and non-Smad targets, and how final cellular responses are affected in response to these noncanonical signaling modes.
Collapse
Affiliation(s)
- Ying E Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
22
|
Hu M, Cui F, Liu F, Wang J, Wei X, Li Y. BMP signaling pathways affect differently migration and invasion of esophageal squamous cancer cells. Int J Oncol 2016; 50:193-202. [DOI: 10.3892/ijo.2016.3802] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/05/2016] [Indexed: 11/06/2022] Open
|
23
|
REN CHUNMEI, LI YANG, CHEN QIANZHAO, ZENG YUHUA, SHAO YING, WU QIUXIANG, YUAN SHUANGXUE, YANG JUNQIN, YU YU, WU KE, HE BAICHENG, SUN WENJUAN. Oridonin inhibits the proliferation of human colon cancer cells by upregulating BMP7 to activate p38 MAPK. Oncol Rep 2016; 35:2691-8. [DOI: 10.3892/or.2016.4654] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 12/17/2015] [Indexed: 11/05/2022] Open
|
24
|
Lu YJ, Fu LJ, Yang JJ, Zeng P, Jiang WM, Wu JB. Effect of siRNA mediated bone morphogenetic protein 7 knockdown on cell proliferation and migration in human hepatoma cell line HepG2. Shijie Huaren Xiaohua Zazhi 2016; 24:10-18. [DOI: 10.11569/wcjd.v24.i1.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of small interfering RNA (siRNA) mediated bone morphogenetic protein 7 (BMP7) knockdown on the proliferation and migration of human hepatoma HepG2 cells.
METHODS: Three pairs of siRNAs targeting BMP7 were transiently transfected into HepG2 cells using TransLipid HL Transfection Reagent. HepG2 cells were divided into five groups including a normal control group, a negative-siRNA group and three transfected groups (transfected with BMP7-siRNA-1, BMP7-siRNA-2, and BMP7-siRNA-3, respectively). The expression of BMP7 mRNA and protein was measured by reverse transcript-polymerase chain reaction (RT-PCR) and Western blot, respectively, and the optimal siRNA sequence for BMP7 silencing was selected. The proliferation and migration of HepG2 cells after transfection were assessed by MTT assay and transwell migration assay, respectively. The expression of apoptosis-related proteins (Bax, Bcl-2, and Caspase3) in each group was determined by Western blot, and the cell cycle was analyzed by flow cytometry.
RESULTS: BMP7-siRNA-3 group demonstrated the lowest level of BMP7 expression among the five groups (P < 0.01). Cell growth was significantly slower in the BMP7-siRNA-3 group than in the control groups 48 h and 72 h after cells were transfected (P < 0.01). The numbers of cell passing the membrane were significantly lower in the BMP7-siRNA-3 group than in the control groups 24 h after cells were transfected (P < 0.01). The expression of Bax and Caspase3 (P < 0.01) was significantly increased after BMP7 silencing, and there was no statistically significant difference in the Bcl-2 expression. Flow cytometery showed that cells were significantly blocked in G2 phase 48 h after cells were transfected with BMP7-siRNA-3 (P < 0.01).
CONCLUSION: SiRNA mediated BMP7 knockdown can inhibit HepG2 cell proliferation and migration, promote apoptosis and block cells in G2 phase.
Collapse
|
25
|
Khan MI, Hamid A, Adhami VM, Lall RK, Mukhtar H. Role of epithelial mesenchymal transition in prostate tumorigenesis. Curr Pharm Des 2015; 21:1240-8. [PMID: 25506896 DOI: 10.2174/1381612821666141211120326] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
Globally, the cancer associated deaths are generally attributed to the spread of cancerous cells or their features to the nearby or distant secondary organs by a process known as metastasis. Among other factors, the metastatic dissemination of cancer cells is attributed to the reactivation of an evolutionary conserved developmental program known as epithelial to mesenchymal transition (EMT). During EMT, fully differentiated epithelial cells undergo a series of dramatic changes in their morphology, along with loss of cell to cell contact and matrix remodeling into less differentiated and invasive mesenchymal cells. Many studies provide evidence for the existence of EMT like states in prostate cancer (PCa) and suggest its possible involvement in PCa progression and metastasis. At the same time, the lack of conclusive evidence regarding the presence of full EMT in human PCa samples has somewhat dampened the interest in the field. However, ongoing EMT research provides new perspectives and unveils the enormous potential of this field in tailoring new therapeutic regimens for PCa management. This review summarizes the role of many transcription factors and other molecules that drive EMT during prostate tumorigenesis.
Collapse
Affiliation(s)
| | | | | | | | - Hasan Mukhtar
- Department of Dermatology, University of Wisconsin, Medical Science Center, Rm B-25, 1300 University Avenue, Madison, WI 53706.
| |
Collapse
|
26
|
Ji X, Jin S, Qu X, Li K, Wang H, He H, Guo F, Dong L. Lysine-specific demethylase 5C promotes hepatocellular carcinoma cell invasion through inhibition BMP7 expression. BMC Cancer 2015; 15:801. [PMID: 26503415 PMCID: PMC4624178 DOI: 10.1186/s12885-015-1798-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/16/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common type of tumor and is associated with high morbidity and mortality rates. Patients with HCC routinely undergo surgery followed by adjuvant radiation therapy and chemotherapy. Despite such aggressive treatment approaches, median survival times remain under 1 year in most cases. KDM5C is a member of the family of JmjC domain-containing proteins that removes methyl residues from methylated lysine 4 on histone H3 lysine 4 (H3K4). KDM5C has been proposed as an oncogene in many types of tumors; however, its role and underlying mechanisms in HCC remain unclear. METHODS Expression level of KDM5C was examined by RT-PCR, and IHC. Forced expression of KDM5C was mediated by retroviruses, and KDM5C was downregulated by shRNAs expressing lentiviruses. Migration and invasion of HCC cells was measured by wound healing, Transwell and Matrigel assays respectively. RESULTS In this study, we report that KDM5C is abundantly expressed in invasive human HCC cells. Cellular depletion of KDM5C by shRNA inhibited HCC cell migration, invasion and epithelial-mesenchymal transition in vitro, and markedly decreased the metastasis capacity of invasive HCC cells in the liver and lung. Furthermore, ectopic expression of KDM5C in HCC cells promoted cell migration, invasion and epithelial-mesenchymal transition via the inactivation of BMP7. Knockdown of BMP7 significantly promotes shKDM5C-induced cell migration inhibition. CONCLUSIONS Taken together, these data suggest that KDM5C-mediated BMP7 inactivation is essential for HCC cell invasion.
Collapse
Affiliation(s)
- Xuening Ji
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, 116001, China.
| | - Shi Jin
- Department of Laparoscopic Surgery, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Street, Shahekou District, Dalian, 116001, China.
| | - Xiaotong Qu
- Department of Second Neurology, The Frist Affiliated Hospital of Dalian Medical University, No.222, Zhongshan Street, Xigang District, Dalian, 116000, China.
| | - Kejun Li
- Department of Laparoscopic Surgery, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Street, Shahekou District, Dalian, 116001, China.
| | - Hongjiang Wang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Street, Shahekou District, Dalian, 116001, China.
| | - Hui He
- Department of Laparoscopic Surgery, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Street, Shahekou District, Dalian, 116001, China.
| | - Fuchao Guo
- Department of general surgery, The first people's Hospital of jinzhou District in Dalian City, No.683, Stalin Road, Jinzhou District, Dalian, 116100, China.
| | - Lei Dong
- Department of Laparoscopic Surgery, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Street, Shahekou District, Dalian, 116001, China.
| |
Collapse
|
27
|
Zhifang M, Liang W, Wei Z, Bin H, Rui T, Nan W, Shuhai Z. The androgen receptor plays a suppressive role in epithelial- mesenchymal transition of human prostate cancer stem progenitor cells. BMC BIOCHEMISTRY 2015; 16:13. [PMID: 25943311 PMCID: PMC4430921 DOI: 10.1186/s12858-015-0042-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/24/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND To investigate the roles of androgen receptor (AR) in epithelial- mesenchymal transition (EMT) in human prostate cancer stem progenitor (S/P) cells isolated from LNCaP cell line. METHODS The S/P cells were obtained from LNCaP cell line through florescence-activated cell sorting (FACS). AR was overexpressed in S/P cells through lentivirus. Western blot assay was used to detect the EMT markers expression, such as E Cadherin, N Cadherin, Vimentin and Snail. MTT assay, soft agar colony formation assay, sphere formation assay and migration assay were used to investigate AR's roles in EMT of S/P cells. Cell signaling pathways associated with proliferation and apoptosis of S/P cells were detected simultaneously. And S/P cells were treated with in vitro combinatory use of LY 294002 (inhibitor of AKT signaling molecules) with γ-TT and/or 5-AZA. RESULTS Our data showed that S/P cells from LNCaP had high EMT markers expression, more tumorigenesis and strong migration ability. And in S/P cells overexpressed with AR, the expression of EMT markers decreased. In addition, these cells had less proliferation ability, tumorigenesis ability, self-renewal and migration ability. At the same time, targeting S/P cells with AKT signaling pathway inhibitor LY29004 and γ-TT and/or 5-AZA could inhibit S/P cell's proliferation and tumorigenesis. CONCLUSIONS Our data suggest that AR played a negative role in EMT of PCa S/P cells, by regulating AKT cell signaling pathway, which could be a new strategy to treat castration resistant prostate cancer (CRPC).
Collapse
Affiliation(s)
- Ma Zhifang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Wei Liang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Zhang Wei
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Hao Bin
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Tu Rui
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Wu Nan
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Zhang Shuhai
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
28
|
Li J, Li X, Ren S, Chen X, Zhang Y, Zhou F, Zhao M, Zhao C, Chen X, Cheng N, Zhao Y, Zhou C, Hirsch FR. miR-200c overexpression is associated with better efficacy of EGFR-TKIs in non-small cell lung cancer patients with EGFR wild-type. Oncotarget 2014; 5:7902-16. [PMID: 25277203 PMCID: PMC4202169 DOI: 10.18632/oncotarget.2302] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 07/29/2014] [Indexed: 12/31/2022] Open
Abstract
Several randomized trials have demonstrated non-small cell lung cancer (NSCLC) patients with activating epidermal growth factor receptor (EGFR) mutations can achieve favorable clinical outcomes on treatment with EGFR tyrosine kinase inhibitors (TKIs). EGFR mutation is considered as a predictive marker for efficacy of EGFR-TKIs in NSCLC. Here we show miR-200c overexpression was correlated with the epithelial phenotype and sensitivity to gefitinib in EGFR wild-type NSCLC cell lines. Up-regulated miR-200c could regain the sensitivity to gefitinib in the EGFR wild-type cell lines and miR-200c could regulate epithelial to mesenchymal transition through PI3K/AKT and MEK/ERK pathways. NSCLC patients at advanced stage (N=150) who received EGFR-TKIs (gefitinib or erlotinib) as second- or third-line therapy from September 2008 to December 2012 were included in the study. In 66 NSCLC patients with wild-type EGFR, high levels of miR-200c expression was associated with higher disease control rate (DCR), longer progression-free survival (PFS) and longer overall survival (OS) compared with low miR-200c expression subgroup. In the subgroup with EGFR mutation, the trend remained the same but not statistically significant. Overall, these findings indicated that miR-200c might be a predictive biomarker for sensitivity to EGFR-TKIs in advanced NSCLC patients with wild-type EGFR.
Collapse
Affiliation(s)
- Jiayu Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Yishi Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Mingchuan Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Xiu Chen
- Department of Respiration, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Ningning Cheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Yinmin Zhao
- Department of Central Laboratory, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University Medical School Cancer Institute, Shanghai, China
| | - Fred R. Hirsch
- Departments of Medicine and Pathology, University of Colorado Cancer Center, Aurora, Colorado, USA
| |
Collapse
|
29
|
Ni J, Cozzi PJ, Hao JL, Beretov J, Chang L, Duan W, Shigdar S, Delprado WJ, Graham PH, Bucci J, Kearsley JH, Li Y. CD44 variant 6 is associated with prostate cancer metastasis and chemo-/radioresistance. Prostate 2014; 74:602-17. [PMID: 24615685 DOI: 10.1002/pros.22775] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/23/2013] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Prostate cancer (CaP) is the second leading malignancy in older men in Western countries. The role of CD44 variant 6 (CD44v6) in CaP progression and therapeutic resistance is still uncertain. Here, we investigated the roles of CD44v6 in CaP metastasis and chemo/radioresistance. Expression of CD44v6 in metastatic CaP cell lines, human primary CaP tissues and lymph node metastases was assessed using immunofluorescence and immunohistochemistry, respectively. METHODS Knock down (KD) of CD44v6 was performed in PC-3M, DU145, and LNCaP cells using small interfering RNA (siRNA), and confirmed by confocal microscope, Western blot and quantitative real time polymerase chain reaction (qRT-PCR). Cell growth was evaluated by proliferation and colony formation assays. The adhesive ability and invasive potential were assessed using a hyaluronic acid (HA) adhesion and a matrigel chamber assay, respectively. Tumorigenesis potential and chemo-/radiosensitivity were measured by a sphere formation assay and a colony assay, respectively. RESULTS Over-expression of CD44v6 was found in primary CaP tissues and lymph node metastases including cancer cells and surrounding stromal cells. KD of CD44v6 suppressed CaP proliferative, invasive and adhesive abilities, reduced sphere formation, enhanced chemo-/radiosensitivity, and down-regulated epithelial-mesenchymal transition (EMT), PI3K/Akt/mTOR, and Wnt/β-catenin signaling pathway proteins in vitro. CONCLUSIONS Our findings demonstrate that CD44v6 is an important cancer stem cell-like marker associated with CaP proliferation, invasion, adhesion, metastasis, chemo-/radioresistance, and the induction of EMT as well as the activation PI3K/Akt/mTOR and Wnt signaling pathways, suggesting that CD44v6 is a novel therapeutic target to sensitize CaP cells to chemo/radiotherapy.
Collapse
Affiliation(s)
- Jie Ni
- Cancer Care Centre and Prostate Cancer Institute, St George Hospital, Kogarah, Australia; St George and Sutherland Clinical School, University of New South Wales (UNSW), Kensington, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ma B, Wells A. The mitogen-activated protein (MAP) kinases p38 and extracellular signal-regulated kinase (ERK) are involved in hepatocyte-mediated phenotypic switching in prostate cancer cells. J Biol Chem 2014; 289:11153-11161. [PMID: 24619413 DOI: 10.1074/jbc.m113.540237] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The greatest challenge for the seeding of cancer in metastatic sites is integration into the ectopic microenvironment despite the lack of an orthotopic supportive environment and presence of pro-death signals concomitant with a localized "foreign-body" inflammatory response. In this metastatic location, many carcinoma cells display a reversion of the epithelial-to-mesenchymal transition that marks dissemination in the primary tumor mass. This mesenchymal to epithelial reverting transition (MErT) is thought to help seeding and colonization by protecting against cell death. We have previously shown that hepatocyte coculture induces the re-expression of E-cadherin via abrogation of autocrine EGFR signaling pathway in prostate cancer (PCa) cells and that this confers a survival advantage. Herein, we show that hepatocytes educate PCa to undergo MErT by modulating the activity of p38 and ERK1/2. Hepatocytes inhibited p38 and ERK1/2 activity in prostate cancer cells, which allowed E-cadherin re-expression. Introduction of constitutively active MEK6 and MEK1 to DU145 cells cocultured with hepatocytes abrogated E-cadherin re-expression. At least a partial phenotypic reversion can be achieved by suppression of p38 and ERK1/2 activation in DU145 cells even in the absence of hepatocytes. Interestingly, these mitogen-activated protein kinase activities were also triggered by re-expressed E-cadherin leading to p38 and ERK1/2 activity in PCa cells; these signals provide protection to PCa cells upon challenge with chemotherapy and cell death-inducing cytokines. We propose that distinct p38/ERK pathways are related to E-cadherin levels and function downstream of E-cadherin allowing, respectively, for hepatocyte-mediated MErT and tumor cell survival in the face of death signals.
Collapse
Affiliation(s)
- Bo Ma
- Department of Pathology, University of Pittsburgh and Pittsburgh Veterans Affairs Medical Center, Pittsburgh Pennsylvania 15261
| | - Alan Wells
- Department of Pathology, University of Pittsburgh and Pittsburgh Veterans Affairs Medical Center, Pittsburgh Pennsylvania 15261.
| |
Collapse
|
31
|
Ha B, Ko H, Kim B, Sohn EJ, Jung JH, Kim JS, Yoon JJ, Won G, Kim JH, Jung DB, Yun M, Shim B, Kim SH. Regulation of crosstalk between epithelial to mesenchymal transition molecules and MMP-9 mediates the antimetastatic activity of anethole in DU145 prostate cancer cells. JOURNAL OF NATURAL PRODUCTS 2014; 77:63-69. [PMID: 24328151 DOI: 10.1021/np4006376] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The underlying antimetastatic mechanism of anethole (1) still remains unclear in association with the molecules of the epithelial to mesenchymal transition (EMT). Herein, the role of the EMT molecules was elucidated in terms of the antimetastatic activity of 1 using DU145 cells. Anethole significantly inhibited the adhesion of DU145 cells to vitronectin-coated plates, as well as migration in a wound-healing assay and invasion using a Boyden chamber. Also, anethole suppressed the expression of MMP-9 in DU145 cells by zymography, ELISA, and RT-PCR. Consistently, the silencing of MMP-9 enhanced the activity of 1 to upregulate the expression of E-cadherin and to attenuate the expression of Vimentin in DU145 cells. Compound 1 enhanced E-cadherin, which is an epithelial marker and attenuated the expression of Vimentin, Twist, and Snail as mesenchymal molecules at the mRNA level. Consistently, anethole upregulated E-cadherin and downregulated the expression of Vimentin, Twist and PI3K, and AKT at the protein level in DU145 cells. Conversely, the antimetastatic effects of 1 to inhibit invasion and the expression of MMP-9 and upregulate E-cadherin were reversed by the EMT inducer TGF-β in DU145 cells. Overall, the present findings suggest that anethole exerts antimetastatic activity via regulation of crosstalk between EMT molecules and MMP-9 on the basis of the in vitro data obtained.
Collapse
Affiliation(s)
- ByungChul Ha
- College of Korean Medicine, Kyung Hee University , Dongdaemun-gu, Seoul 131-701, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhang S, Zhong B, Chen M, Yang L, Yang G, Li Y, Wang H, Wang G, Li W, Cui J, Hoffman AR, Hu J. Epigenetic reprogramming reverses the malignant epigenotype of the MMP/TIMP axis genes in tumor cells. Int J Cancer 2013; 134:1583-94. [PMID: 24105737 DOI: 10.1002/ijc.28487] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 09/04/2013] [Indexed: 12/31/2022]
Abstract
Cancer progression is characterized by extensive tumor invasion into the surrounding extracellular matrix (ECM) and migration to metastatic sites. The increased proteolytic degradation of the ECM during tumor invasion is directly dependent on the activity of matrix metalloproteinases (MMPs), counter-balanced by tissue inhibitors of matrix metalloproteinases (TIMPs). In this study, we found that unbalanced expression of MMP/TIMP axis genes in tumors was correlated with aberrant epigenotypes in the various gene promoters. The malignant epigenotypes could be therapeutically corrected by a simple defined factor-mediated reprogramming approach. Correction of the abnormal epigenotypes by nuclear remodeling leads to a rebalance in the gene expression profile, an alteration in tumor cell morphology, attenuation of tumor cell migration and invasion in vitro, and reduced tumorigenicity in nude mice. We further identified the downregulation of the MKK-p38 MAPK signal pathway as an important underlying mechanism for reduced tumorigenicity in this epigenetic reprogramming model. These data demonstrate that the malignant phenotypes seen in cancer can be corrected by a nuclear remodeling mechanism, thus highlighting a novel non-chemotherapeutic, non-radiotherapeutic approach for the treatment of cancer.
Collapse
Affiliation(s)
- Shenghong Zhang
- Division of Gastroenterology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, People's Republic of China; VA Palo Alto Health Care System, Stanford University Medical School, Palo Alto, CA, 94304, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ni J, Cozzi P, Hao J, Beretov J, Chang L, Duan W, Shigdar S, Delprado W, Graham P, Bucci J, Kearsley J, Li Y. Epithelial cell adhesion molecule (EpCAM) is associated with prostate cancer metastasis and chemo/radioresistance via the PI3K/Akt/mTOR signaling pathway. Int J Biochem Cell Biol 2013; 45:2736-48. [PMID: 24076216 DOI: 10.1016/j.biocel.2013.09.008] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/06/2013] [Accepted: 09/14/2013] [Indexed: 12/21/2022]
Abstract
Prostate cancer (CaP) is the second leading malignancy in men. The role of epithelial cell adhesion molecule (EpCAM), also known as CD326, in CaP progression and therapeutic resistance is still uncertain. Here, we aimed to investigate the roles of EpCAM in CaP metastasis and chemo/radioresistance. Expression of EpCAM in CaP cell lines and human CaP tissues was assessed using immunofluorescence and immunohistochemistry, respectively. EpCAM was knocked down (KD) in PC-3, DU145 and LNCaP-C4-2B cells using small interfering RNA (siRNA), and KD results were confirmed by confocal microscope, Western blotting and quantitative real time polymerase chain reaction (qRT-PCR). Cell growth was evaluated by proliferation and colony formation assays. The invasive potential was assessed using a matrigel chamber assay. Tumorigenesis potential was measured by a sphere formation assay. Chemo-/radiosensitivity were measured using a colony formation assay. Over-expression of EpCAM was found in primary CaP tissues and lymph node metastases including cancer cells and surrounding stromal cells. KD of EpCAM suppressed CaP proliferation and invasive ability, reduced sphere formation, enhanced chemo-/radiosensitivity, and down-regulated E-cadherin, p-Akt, p-mTOR, p-4EBP1 and p-S6K expression in CaP cells. Our findings suggest that EpCAM plays an important role in CaP proliferation, invasion, metastasis and chemo-/radioresistance associated with the activation of the PI3K/Akt/mTOR signaling pathway and is a novel therapeutic target to sensitize CaP cells to chemo-/radiotherapy.
Collapse
Affiliation(s)
- Jie Ni
- Cancer Care Centre and Prostate Cancer Institute, St George Hospital, Kogarah, NSW 2217, Australia; St George Clinical School, University of New South Wales (UNSW), Kensington, NSW 2052, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
McCormack N, O'Dea S. Regulation of epithelial to mesenchymal transition by bone morphogenetic proteins. Cell Signal 2013; 25:2856-62. [PMID: 24044921 DOI: 10.1016/j.cellsig.2013.09.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 09/02/2013] [Accepted: 09/06/2013] [Indexed: 02/08/2023]
Abstract
Epithelial to mesenchymal transition (EMT) is a process in which fully differentiated epithelial cells lose many of their epithelial characteristics and adopt features typical of mesenchymal cells, thus allowing cells to become migratory and invasive. EMT is a critical process in development and its role in cancer and fibrosis is becoming increasingly recognised. It is also becoming apparent that EMT is not just restricted to embryonic development and disease in adults, but in fact may be an important process for the maintenance and regeneration of adult tissue architecture. While transforming growth factor-β (TGF-β) is considered a prototypic inducer of EMT, relatively little is known about other signalling molecules that regulate EMT. Bone morphogenic proteins (BMPs) are members of the TGF-β superfamily and 20 different human BMPs have been identified. Originally named for their effects on bone, these proteins are now considered to be key morphogenetic signals that orchestrate tissue architecture throughout the body. BMP2, -4 and -7 are the best studied to date. There are disparate reports of the roles of BMPs in EMT during development, cancer and fibrosis. Here, we present an overview of this literature as well as the emerging role of EMT in tissue regeneration and the involvement of BMPs in regulating this process.
Collapse
Affiliation(s)
- Natasha McCormack
- Institute of Immunology, National University of Ireland Maynooth, Ireland.
| | | |
Collapse
|
35
|
Behnsawy HM, Shigemura K, Meligy FY, Yamamichi F, Yamashita M, Haung WC, Li X, Miyake H, Tanaka K, Kawabata M, Shirakawa T, Fujisawa M. Possible role of sonic hedgehog and epithelial-mesenchymal transition in renal cell cancer progression. Korean J Urol 2013; 54:547-54. [PMID: 23956832 PMCID: PMC3742909 DOI: 10.4111/kju.2013.54.8.547] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/06/2013] [Indexed: 11/24/2022] Open
Abstract
Purpose Sonic hedgehog (Shh) signaling and epithelial-mesenchymal transition (EMT) are both known to relate to cancer progression. The purpose of this study was to investigate the role of Shh signaling and EMT in renal cell carcinoma (RCC). Materials and Methods Cell proliferation was assayed in RCC cell lines in the presence or absence of a Shh signaling stimulator, recombinant Shh (r-Shh) protein, or a Shh signaling inhibitor, cyclopamine. Real-time reverse transcription-polymerase chain reaction (RT-PCR) was performed to study the expression of EMT markers (E-cadherin, N-cadherin, and vimentin) and osteonectin. The expression of Ki-67, Gli-1, osteonectin, and EMT markers in nephrectomy specimens from RCC patients was also measured by immunohistochemical (IHC) staining. Results RCC cells showed enhanced cell proliferation by r-Shh protein, whereas cell proliferation was suppressed by the addition of cyclopamine in RenCa cells. Real-time RT-PCR showed that r-Shh suppressed the expression of E-cadherin and that this suppression was partly blocked by cyclopamine alone in RenCa cells. In the IHC results, osteonectin significantly correlated with vein sinus invasion (p=0.0218), and the expression of vimentin significantly correlated with lymphatic invasion (p=0.0392). Conclusions Shh signaling and EMT play roles in RCC progression, and the Shh signaling inhibitor cyclopamine might be a possible molecular targeted therapeutic strategy for RCC.
Collapse
Affiliation(s)
- Hosny M Behnsawy
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan. ; Faculty of Medicine, Assiut University, Assiut, Egypt
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Xu G, Tang S, Yang J, Chen K, Kang J, Zhao G, Feng F, Yang X, Zhao L, Lu Q, Sun L, Hong L, Gong T, Zhang H. BMP7 expression in esophageal squamous cell carcinoma and its potential role in modulating metastasis. Dig Dis Sci 2013; 58:1871-9. [PMID: 23504348 DOI: 10.1007/s10620-013-2611-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 02/18/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Our previous study showed that BMP7 revealed significantly higher levels in esophageal squamous cell carcinoma (ESCC) tissues with lymph node metastasis compared with non-lymph node metastasis, using gene expression profiling assays. The roles of BMP7 in ESCC is not fully understood. AIM The aim of this study was to investigate the effect of BMP7 on lymph node metastasis of ESCC and to explore its potential mechanism. METHODS Expression of BMP7 in ESCC tissues was evaluated by immunohistochemistry. BMP7 were down-regulated by RNA interference. The protein and mRNA levels of BMP7 were detected by western blot and RT-PCR, respectively. High content screening and transwell assay were used to identify the metastatic ability of tumor cells. RESULTS Positivity of BMP7 staining was 57.5 % in the tissues of primary carcinoma with lymph node metastasis compared to tissues without lymph node metastasis, and expression of BMP7 was significantly higher in the cell lines with highly metastatic capacity than that in the cell lines without metastatic ability. Suppression of endogenous BMP7 expression by siRNA in the highly metastatic cell lines resulted in significant reduction in ability of cell migration and invasion in both in vitro and in vivo studies. In addition, inhibition of BMP7 by siRNA also leads to up-regulation of E-cadherin and down-regulation of MMP-9 in the highly metastatic cell lines. CONCLUSIONS These findings indicate that BMP7 modulates the expression of E-cadherin and MMP-9, and by which mechanism it may regulate cell migration and metastasis of ESCCs.
Collapse
Affiliation(s)
- Guanghui Xu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, 15 Changle Western Road, Xi'an 710032, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Du F, Wu X, Liu Y, Wang T, Qi X, Mao Y, Jiang L, Zhu Y, Chen Y, Zhu R, Han X, Jin J, Ma X, Hua D. Acquisition of paclitaxel resistance via PI3K‑dependent epithelial‑mesenchymal transition in A2780 human ovarian cancer cells. Oncol Rep 2013; 30:1113-8. [PMID: 23807572 DOI: 10.3892/or.2013.2567] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 04/10/2013] [Indexed: 11/05/2022] Open
Abstract
Epithelial ovarian cancer is a major cause of mortality among women with gynecological malignancies. Paclitaxel is commonly used for chemotherapy of ovarian cancer, yet its efficacy is limited by chemoresistance. Generally, drug resistance is associated with acquisition of the epithelial-mesenchymal transition (EMT) in cancer. The aim of the present study was to determine whether the EMT is involved in acquired resistance to paclitaxel in A2780 human ovarian cancer cells. Using the paclitaxel-resistant A2780/PTX cell line, we examined the cellular morphology, molecular changes, migration and proliferation consistent with the EMT. Furthermore, we found that inhibition of phosphatidylinositol 3-kinase (PI3K) activity reduced the proliferation and migration and restored their sensitivity to paclitaxel. Our study provides new insights into EMT-like phenotypic changes that are linked to paclitaxel resistance in A2780 cells. We believe that inhibition of the PI3K signaling pathway could provide a novel therapeutic approach to overcome chemoresistance and prevent metastasis during paclitaxel chemotherapy.
Collapse
Affiliation(s)
- Fangfang Du
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, Jiangsu, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is a key signaling pathway that has been linked to both tumorigenesis and resistance to therapy in prostate cancer and other solid tumors. Given the significance of the PI3K/Akt/mTOR pathway in integrating cell survival signals and the high prevalence of activating PI3K/Akt/mTOR pathway alterations in prostate cancer, inhibitors of this pathway have great potential for clinical benefit. Here, we review the role of the PI3K/Akt/mTOR pathway in prostate cancer and discuss the potential use of pathway inhibitors as single agents or in combination in the evolving treatment landscape of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Rhonda L Bitting
- Division of Medical Oncology, Duke Cancer Institute, Duke University, DUMC Box 102002, Durham, North Carolina 27710, USA
| | | |
Collapse
|
39
|
Marked improvement of cytotoxic effects induced by docetaxel on highly metastatic and androgen-independent prostate cancer cells by downregulating macrophage inhibitory cytokine-1. Br J Cancer 2013; 108:1079-91. [PMID: 23449353 PMCID: PMC3619053 DOI: 10.1038/bjc.2012.484] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background: Overexpression of macrophage inhibitory cytokine-1 (MIC-1) frequently occurs during the progression of prostate cancer (PC) to androgen-independent (AI) and metastatic disease states and is associated with a poor outcome of patients. Methods: The gain- and loss-of-function analyses of MIC-1 were performed to establish its implications for aggressive and chemoresistant phenotypes of metastatic and AI PC cells and the benefit of its downregulation for reversing docetaxel resistance. Results: The results have indicated that an enhanced level of secreted MIC-1 protein in PC3 cells is associated with their acquisition of epithelial–mesenchymal transition features and higher invasive capacity and docetaxel resistance. Importantly, the downregulation of MIC-1 in LNCaP-LN3 and PC3M-LN4 cells significantly decreased their invasive capacity and promoted the antiproliferative, anti-invasive and mitochrondrial- and caspase-dependent apoptotic effects induced by docetaxel. The downregulation of MIC-1 in PC3M-LN4 cells was also effective in promoting the cytotoxic effects induced by docetaxel on the side population (SP) endowed with stem cell-like properties and the non-SP cell fraction from PC3M-LN4 cells. Conclusion: These data suggest that the downregulation of MIC-1 may constitute a potential therapeutic strategy for improving the efficacy of current docetaxel-based chemotherapies, eradicating the total mass of PC cells and thereby preventing disease relapse and the death of PC patients.
Collapse
|
40
|
Bi WR, Jin CX, Xu GT, Yang CQ. Effect of alendronate sodium on the expression of mesenchymal-epithelial transition markers in mice with liver fibrosis. Exp Ther Med 2012; 5:247-252. [PMID: 23251277 PMCID: PMC3524127 DOI: 10.3892/etm.2012.759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 10/11/2012] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to explore whether alendronate sodium regulates tissue remodeling by controlling the transforming growth factor (TGF)-β1-induced epithelial-mesenchymal transition (EMT) and bone morphogenetic protein (BMP)-7-induced mesenchymal-epithelial transition (MET) in CCl4-induced hepatic fibrosis in mice. A mouse model of CCl4-induced hepatic fibrosis was evaluated using the hematoxylin and eosin (HE) and Masson’s trichrome staining histological methods. The activities of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured using an automated biochemical analyzer. The expression of TGF-β1, α-smooth muscle actin (α-SMA), BMP-7 and E-cadherin in the hepatic tissue was detected using immunohistochemistry. The mRNA and protein levels of TGF-β1, α-SMA, BMP-7, fibroblast-specific protein 1 (FSP1), E-cadherin and N-cadherin were detected using RT-PCR and western blot analysis. Immunohistochemical and molecular biochemical examination revealed that alendronate sodium significantly arrested the progression of hepatic fibrosis. Alendronate sodium caused significant amelioration of liver injury and reduced the activities of serum ALT and AST (P<0.001). Furthermore, alendronate sodium markedly reduced TGF-β1 and α-SMA mRNA expression and increased BMP-7 and E-cadherin in the mouse liver tissue (P<0.001). Alendronate sodium significantly arrested the progression of hepatic fibrosis. The underlying mechanism was associated with changes in the redox state, which remains variable in liver fibrosis, and depends on the balance between TGF-β/smad- and BMP-7-modulated mechanisms which regulate EMT and MET in multifunctional progenitors.
Collapse
Affiliation(s)
- Wan-Rong Bi
- Department of Gastroenterology and Digestive Disease Institute, Tongji Hospital Branch
| | | | | | | |
Collapse
|
41
|
Xu WH, Han M, Dong Q, Fu ZX, Diao YY, Liu H, Xu J, Jiang HL, Zhang SZ, Zheng S, Gao JQ, Wei QC. Doxorubicin-mediated radiosensitivity in multicellular spheroids from a lung cancer cell line is enhanced by composite micelle encapsulation. Int J Nanomedicine 2012; 7:2661-71. [PMID: 22679376 PMCID: PMC3368509 DOI: 10.2147/ijn.s30445] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background The purpose of this study is to evaluate the efficacy of composite doxorubicinloaded micelles for enhancing doxorubicin radiosensitivity in multicellular spheroids from a non-small cell lung cancer cell line. Methods A novel composite doxorubicin-loaded micelle consisting of polyethylene glycolpolycaprolactone/Pluronic P105 was developed, and carrier-mediated doxorubicin accumulation and release from multicellular spheroids was evaluated. We used confocal laser scanning microscopy and flow cytometry to study the accumulation and efflux of doxorubicin from A549 multicellular spheroids. Doxorubicin radiosensitization and the combined effects of irradiation and doxorubicin on cell migration and proliferation were compared for the different doxorubicin delivery systems. Results Confocal laser scanning microscopy and quantitative flow cytometry studies both verified that, for equivalent doxorubicin concentrations, composite doxorubicin-loaded micelles significantly enhanced cellular doxorubicin accumulation and inhibited doxorubicin release. Colony-forming assays demonstrated that composite doxorubicin-loaded micelles are radiosensitive, as shown by significantly reduced survival of cells treated by radiation + composite micelles compared with those treated with radiation + free doxorubicin or radiation alone. The multicellular spheroid migration area and growth ability verified higher radiosensitivity for the composite micelles loaded with doxorubicin than for free doxorubicin. Conclusion Our composite doxorubicin-loaded micelle was demonstrated to have radiosensitization. Doxorubicin loading in the composite micelles significantly increased its cellular uptake, improved drug retention, and enhanced its antitumor effect relative to free doxorubicin, thereby providing a novel approach for treatment of cancer.
Collapse
Affiliation(s)
- Wen-Hong Xu
- Department of Radiation Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|