51
|
Li D, Zhang Y, Zhang H, Zhan C, Li X, Ba T, Qiu Z, E F, Lv G, Zou C, Wang C, Si L, Zou C, Li Q, Gao X. CADM2, as a new target of miR-10b, promotes tumor metastasis through FAK/AKT pathway in hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:46. [PMID: 29506532 PMCID: PMC5836378 DOI: 10.1186/s13046-018-0699-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 02/07/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cell adhesion molecules (CADMs) comprise of a protein family whose functions include maintenance of cell polarity and tumor suppression. Hypo-expression of CADM2 gene expression has been observed in several cancers including hepatocellular carcinoma (HCC). However, the role and mechanisms of CADM2 in HCC remain unclear. METHODS The expression of CADM2 and miRNA-10b (miR-10b) in HCC tissues and cell lines were detected using real-time PCR and Western blotting. Immunofluorescence was used to detect Epithelial-mesenchymal transition (EMT) progression in HCC cell lines. Dual-luciferase reporter assay was used to determine miR-10b binding to CADM2 3'UTR. Wound healing assay and Transwell assay were performed to examine the migration and invasion of HCC cells. RESULTS We report the effect of CADM2 as a tumor suppressor in HCC. Firstly, we confirmed that CADM2 expression was significantly down regulated in HCC tissues compared to normal tissues according to TCGA data analysis and fresh HCC sample detection. Secondly, overexpression of CADM2 could inhibit EMT process, migratory and invasion ability of HCC cells. Furthermore, the results indicated that CADM2 is a direct target of miR-10b in HCC cells and miR-10b/CADM2 modulates EMT process and migration ability via focal adhesion kinase (FAK) /AKT signaling pathway in HCC. CONCLUSIONS Our study demonstrates that miR-10b-CADM2-FAK/AKT axis plays an important role in HCC metastasis, which might be a novel potential therapeutic option for HCC treatment.
Collapse
Affiliation(s)
- Dongliang Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yongjian Zhang
- Department of Hepatobiliary and Pancreas, Heilongjiang Cancer Hospital, Harbin, China
| | - He Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Chao Zhan
- Department of Hepatobiliary and Pancreas, Heilongjiang Cancer Hospital, Harbin, China
| | - Xin Li
- Department of Respiratory Medical Oncology, Heilongjiang Cancer Hospital, Harbin, China
| | - Tu Ba
- Department of Neck and Breast Surgery, Mudanjiang Tumor Hospital, Mudanjiang, China
| | - Zini Qiu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Fang E
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Guixiang Lv
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Chendan Zou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Chuxuan Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Lining Si
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China.,Department of Critical-care Medicine, the Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| | - Chaoxia Zou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China. .,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medicine Sciences, Harbin, Heilongjiang, 150081, China.
| | - Qiang Li
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China. .,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medicine Sciences, Harbin, Heilongjiang, 150081, China. .,Key Laboratory of Cardiovascular Medicine Research of Harbin Medical University, Ministry of Education, Harbin, China.
| |
Collapse
|
52
|
Meel MH, Schaper SA, Kaspers GJL, Hulleman E. Signaling pathways and mesenchymal transition in pediatric high-grade glioma. Cell Mol Life Sci 2018; 75:871-887. [PMID: 29164272 PMCID: PMC5809527 DOI: 10.1007/s00018-017-2714-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/24/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022]
Abstract
Pediatric high-grade gliomas (pHGG), including diffuse intrinsic pontine gliomas (DIPG), are the most lethal types of cancer in children. In recent years, it has become evident that these tumors are driven by epigenetic events, mainly mutations involving genes encoding Histone 3, setting them apart from their adult counterparts. These tumors are exceptionally resistant to chemotherapy and respond only temporarily to radiotherapy. Moreover, their delicate location and diffuse growth pattern make complete surgical resection impossible. In many other forms of cancer, chemo- and radioresistance, in combination with a diffuse, invasive phenotype, are associated with a transcriptional program termed the epithelial-to-mesenchymal transition (EMT). Activation of this program allows cancer cells to survive individually, invade surrounding tissues and metastasize. It also enables them to survive exposure to cytotoxic therapy, including chemotherapeutic drugs and radiation. We here suggest that EMT plays an important, yet poorly understood role in the biology and therapy resistance of pHGG and DIPG. This review summarizes the current knowledge on the major signal transduction pathways and transcription factors involved in the epithelial-to-mesenchymal transition in cancer in general and in pediatric HGG and DIPG in particular. Despite the fact that the mesenchymal transition has not yet been specifically studied in pHGG and DIPG, activation of pathways and high levels of transcription factors involved in EMT have been described. We conclude that the mesenchymal transition is likely to be an important element of the biology of pHGG and DIPG and warrants further investigation for the development of novel therapeutics.
Collapse
Affiliation(s)
- Michaël H Meel
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Sophie A Schaper
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Gertjan J L Kaspers
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands
| | - Esther Hulleman
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| |
Collapse
|
53
|
Li MY, Liu JQ, Chen DP, Li ZY, Qi B, Yin WJ, He L. p68 prompts the epithelial-mesenchymal transition in cervical cancer cells by transcriptionally activating the TGF-β1 signaling pathway. Oncol Lett 2018; 15:2111-2116. [PMID: 29434913 PMCID: PMC5777103 DOI: 10.3892/ol.2017.7552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/28/2017] [Indexed: 11/16/2022] Open
Abstract
Overexpression of p68 has been reported in various types of cancer. However, little study has been conducted on the expression and role of p68 in cervical cancer. Therefore, the present study focuses on the role of p68 in cervical cancer cells, which may elucidate its potential mechanism of cervical cancer progression and shed light on the precision medical treatment of cervical cancer. Firstly, the expression of p68 was analyzed using reverse transcription-quantitative polymerase chain reaction and western blot analysis. The changes to cell morphology were observed using an inverted microscope (XDS-500D; Shanghai Caikon Optical Instrument Co., Ltd., Shanghai, China). Cell migration was determined using an in vitro scratch assay. The present study demonstrated that the mRNA and protein levels of p68 were significantly enhanced in cervical cancer CaSki, HeLa [human papillomavirus (HPV)-18-positive], SiHa (HPV-16-positive) and C-33A (HPV-negative) cell lines compared with the human keratinocyte HaCaT cell line. Overexpression of p68 induced an elongated and spindle-shaped morphology in CaSki cells. Upregulation of p68 increased the expression of α-smooth muscle actin, vimentin and fibronectin however, epithelial marker E-cadherin was significantly decreased. Furthermore, the in vitro scratch assay demonstrated that overexpression of p68 markedly enhanced CaSki cell migration capacity at 24 and 48 h. Knockdown of p68 partially reversed transforming growth factor-β1 (TGF-β1)-induced changes in epithelial-mesenchymal transition (EMT) markers and cell morphological changes. In summary, the present study demonstrated that p68 transcriptionally activated the expression of TGF-β1, thereby prompting EMT in cervical cancer cells.
Collapse
Affiliation(s)
- Ming-Yi Li
- The 5th Ward of The Radiotherapy Department, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Jin-Quan Liu
- The 5th Ward of The Radiotherapy Department, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Dong-Ping Chen
- The 5th Ward of The Radiotherapy Department, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Zhou-Yu Li
- The 5th Ward of The Radiotherapy Department, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Bin Qi
- The 5th Ward of The Radiotherapy Department, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Wen-Jing Yin
- The 5th Ward of The Radiotherapy Department, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Lu He
- The 5th Ward of The Radiotherapy Department, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| |
Collapse
|
54
|
Yang K, Li Y, Lian G, Lin H, Shang C, Zeng L, Chen S, Li J, Huang C, Huang K, Chen Y. KRAS promotes tumor metastasis and chemoresistance by repressing RKIP via the MAPK-ERK pathway in pancreatic cancer. Int J Cancer 2018; 142:2323-2334. [PMID: 29315556 DOI: 10.1002/ijc.31248] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 12/02/2017] [Accepted: 12/19/2017] [Indexed: 12/14/2022]
Abstract
Oncogenic KRAS plays a crucial role in pancreatic ductal adenocarcinoma (PDAC) development and progression. However, the mechanism has not been clearly elucidated. RKIP is a tumor repressor, and loss of RKIP has been shown in PDAC. Here, we found that KRAS expression was inversely correlated with RKIP expression in PDAC fresh tissue regardless of the KRAS mutant status. The negative correlation between KRAS and RKIP was further confirmed in our PDAC tissue microarray. KRAS overexpression and RKIP downregulation were associated with poor clinical outcomes. Knockdown or overexpression of KRAS in PDAC cell lines robustly increased or decreased, respectively, RKIP protein and mRNA levels. Furthermore, the MAPK-ERK pathway was involved in the regulation of RKIP. KRAS-regulated RKIP expression, which in turn affected the expression of pivotal epithelial-mesenchymal transition (EMT) and apoptosis factors. The biological function of the KRAS-RKIP axis was demonstrated in human pancreatic cancer cells in vitro and in vivo. KRAS knockdown increased RKIP expression and inhibited metastasis and chemoresistance. Moreover, the feature of metastasis and chemoresistance was rescued in the KRAS-knockdown cells through the inhibition of RKIP by RNA interference. In conclusion, our studies demonstrate how KRAS inhibits the tumor suppressor RKIP, thus offering novel justification for targeting RKIP as a strategy to overcome KRAS-induced tumor metastasis and chemoresistance in PDAC.
Collapse
Affiliation(s)
- Kege Yang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Yaqing Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guoda Lian
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haoming Lin
- Department of Biliary and Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Changzhen Shang
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linjuan Zeng
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Shaojie Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiajia Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chumei Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kaihong Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yinting Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
55
|
Polo-like kinase 4 mediates epithelial-mesenchymal transition in neuroblastoma via PI3K/Akt signaling pathway. Cell Death Dis 2018; 9:54. [PMID: 29352113 PMCID: PMC5833556 DOI: 10.1038/s41419-017-0088-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 10/03/2017] [Accepted: 10/24/2017] [Indexed: 12/12/2022]
Abstract
Neuroblastoma (NB) is the most common malignant tumor in infancy and most common extracranial solid tumor in childhood. With the improvement of diagnosis and treatment, the survival rate of patients with low-risk and intermediate-risk NB can reach up to 90%. In contrast, for high-risk NBs, the long-term survival rate is still <40% because of heterogeneity of this tumor. The pathogenesis of NB is still not explicit, therefore it is of great significance to explore the mechanism of NB tumorigenesis and discover new therapeutic targets for NB. Polo-like kinase 4 (PLK4), one of the polo-like kinase family members, is an important regulator of centriole replication. The aberrant expression of PLK4 was found in several cancers and a recent study has unraveled a novel function of PLK4 as a mediator of invasion and metastasis in Hela and U2OS cells. However, the function of PLK4 in NB development and progression remains to be elucidated. The study showed the expression level of PLK4 in NB tissues was remarkably upregulated and high expression of PLK4 was negatively correlated with clinical features and survival, which suggested that PLK4 could be a potential tumor-promoting factor of NB. Functional studies indicated downregulation of PLK4 suppressed migration and invasion and promoted apoptosis in NB cells. Further experiments showed that downregulation of PLK4 in NB cells inhibited EMT through the PI3K/Akt signaling pathway. Animal experiments demonstrated that the downregulation of PLK4 in SK-N-BE(2) cells dramatically suppressed tumorigenesis and metastasis. PLK4 may be a promising therapeutic target for NB.
Collapse
|
56
|
The transcription factor RUNX2 regulates receptor tyrosine kinase expression in melanoma. Oncotarget 2018; 7:29689-707. [PMID: 27102439 PMCID: PMC5045426 DOI: 10.18632/oncotarget.8822] [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: 11/16/2015] [Accepted: 03/28/2016] [Indexed: 12/12/2022] Open
Abstract
Receptor tyrosine kinases-based autocrine loops largely contribute to activate the MAPK and PI3K/AKT pathways in melanoma. However, the molecular mechanisms involved in generating these autocrine loops are still largely unknown. In the present study, we examine the role of the transcription factor RUNX2 in the regulation of receptor tyrosine kinase (RTK) expression in melanoma. We have demonstrated that RUNX2-deficient melanoma cells display a significant decrease in three receptor tyrosine kinases, EGFR, IGF-1R and PDGFRβ. In addition, we found co-expression of RUNX2 and another RTK, AXL, in both melanoma cells and melanoma patient samples. We observed a decrease in phosphoAKT2 (S474) and phosphoAKT (T308) levels when RUNX2 knock down resulted in significant RTK down regulation. Finally, we showed a dramatic up regulation of RUNX2 expression with concomitant up-regulation of EGFR, IGF-1R and AXL in melanoma cells resistant to the BRAF V600E inhibitor PLX4720. Taken together, our results strongly suggest that RUNX2 might be a key player in RTK-based autocrine loops and a mediator of resistance to BRAF V600E inhibitors involving RTK up regulation in melanoma.
Collapse
|
57
|
Khalafalla FG, Khan MW. Inflammation and Epithelial-Mesenchymal Transition in Pancreatic Ductal Adenocarcinoma: Fighting Against Multiple Opponents. CANCER GROWTH AND METASTASIS 2017; 10:1179064417709287. [PMID: 28579826 PMCID: PMC5436837 DOI: 10.1177/1179064417709287] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and one of the most lethal human cancers. Inflammation is a critical component in PDAC initiation and progression. Inflammation also contributes to the aggressiveness of PDAC indirectly via induction of epithelial-mesenchymal transition (EMT), altogether leading to enhanced resistance to chemotherapy and poor survival rates. This review gives an overview of the key pro-inflammatory signaling pathways involved in PDAC pathogenesis and discusses the role of inflammation in induction of EMT and development of chemoresistance in patients with PDAC.
Collapse
|
58
|
Singhal SS, Singhal S, Singhal P, Singhal J, Horne D, Awasthi S. Didymin: an orally active citrus flavonoid for targeting neuroblastoma. Oncotarget 2017; 8:29428-29441. [PMID: 28187004 PMCID: PMC5438742 DOI: 10.18632/oncotarget.15204] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 01/27/2017] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma, a rapidly growing yet treatment responsive cancer, is the third most common cancer of children and the most common solid tumor in infants. Unfortunately, neuroblastoma that has lost p53 function often has a highly treatment-resistant phenotype leading to tragic outcomes. In the context of neuroblastoma, the functions of p53 and MYCN (which is amplified in ~25% of neuroblastomas) are integrally linked because they are mutually transcriptionally regulated, and because they together regulate the catalytic activity of RNA polymerases. Didymin is a citrus-derived natural compound that kills p53 wild-type as well as drug-resistant p53-mutant neuroblastoma cells in culture. In addition, orally administered didymin causes regression of neuroblastoma xenografts in mouse models, without toxicity to non-malignant cells, neural tissues, or neural stem cells. RKIP is a Raf-inhibitory protein that regulates MYCN activation, is transcriptionally upregulated by didymin, and appears to play a key role in the anti-neuroblastoma actions of didymin. In this review, we discuss how didymin overcomes drug-resistance in p53-mutant neuroblastoma through RKIP-mediated inhibition of MYCN and its effects on GRK2, PKCs, Let-7 micro-RNA, and clathrin-dependent endocytosis by Raf-dependent and -independent mechanisms. In addition, we will discuss studies supporting potential clinical impact and translation of didymin as a low cost, safe, and effective oral agent that could change the current treatment paradigm for refractory neuroblastoma.
Collapse
Affiliation(s)
- Sharad S. Singhal
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, USA
| | - Sulabh Singhal
- University of California at San Diego, La Jolla, San Diego, CA, USA
| | | | - Jyotsana Singhal
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, USA
| | - Sanjay Awasthi
- Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
59
|
Epithelial-mesenchymal transition in morphogenesis, cancer progression and angiogenesis. Exp Cell Res 2017; 353:1-5. [PMID: 28257786 DOI: 10.1016/j.yexcr.2017.02.041] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 12/18/2022]
Abstract
All organs consist of an epithelium and an associated mesenchyme, so these epithelial-mesenchymal intercations are among the most important phenomena in nature. The aim of this article is the summarize the common mechanisms involved in the establishment of epithelial mesenchymal transition in three biological processes, namely organogenesis, tumor progression and metastasis, and angiogenesis, apparently independent each from other. A common feature of these processes is the fact that specialized epithelial cells lose their features, including cell adhesion and polarity, reorganize their cytoskeleton, and acquire a mesenchymal morphology and the ability to migrate.
Collapse
|
60
|
Cintra Lopes Carapeto F, Neves Comodo A, Germano A, Pereira Guimarães D, Barcelos D, Fernandes M, Landman G. Marker Protein Expression Combined With Expression Heterogeneity is a Powerful Indicator of Malignancy in Acral Lentiginous Melanomas. Am J Dermatopathol 2017; 39:114-120. [PMID: 28134728 DOI: 10.1097/dad.0000000000000635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PATIENTS AND METHODS Samples of acral lentiginous melanomas (ALMs) were obtained from the Department of Pathology at Escola Paulista de Medicina-Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil. Demographic, clinical, and follow-up data were obtained from the charts of Hospital São Paulo. From 2 tissue microarrays containing 60 nevi and quadruplicate samples of ≥1.0-mm of 49 ALM, sections were stained to evaluate SCF, KIT, BRAF, CYCLIND1, MYC, and PTEN immunohistochemical protein expression. RESULTS Nevi and ALM from 2006 to 2010 were reviewed and collected. All specimens were in the vertical growth phase, and histopathological parameters indicated that tumors were at an advanced stage at diagnosis. Average tumor thickness was 6.95 mm, 63% were ulcerated, average mitotic index was 5 mitotic cells per mm, and 43% were at Clark's level V. Compared with nevi, the χ test showed that ALM significantly correlated with SCF protein expression (P = 0.001) and expression heterogeneity (P < 0.000). Similar findings were observed for KIT (P = 0.005, P = 0.003, respectively), MYC (P < 0.000, P < 0.000), and PTEN (P = 0.005, P < 0.000). Malignancy did not correlate with BRAF and CYCLIN D1 expression (P = 0.053 and P = 0.259, respectively), but it did significantly correlate with their heterogeneous expression (P < 0.000, P = 0.024, respectively). Combined protein expression had an odds ratio of greater malignancy when BRAF and MYC were positive and/or heterogeneously expressed (OR of 78 and 95, respectively). DISCUSSION AND CONCLUSION We show that marker protein expression, when combined with heterogeneous expression as shown by immunohistochemistry, is a powerful indicator of malignancy in ALMs, especially, when protein pairs are combined.
Collapse
|
61
|
Guarneri C, Bevelacqua V, Polesel J, Falzone L, Cannavò PS, Spandidos DA, Malaponte G, Libra M. NF‑κB inhibition is associated with OPN/MMP‑9 downregulation in cutaneous melanoma. Oncol Rep 2017; 37:737-746. [PMID: 28075446 PMCID: PMC5355753 DOI: 10.3892/or.2017.5362] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/20/2016] [Indexed: 01/21/2023] Open
Abstract
The development of cutaneous melanoma is influenced by genetic factors, including BRAF mutations and environmental factors, such as ultraviolet exposure. Its progression has been also associated with the involvement of several tumour microenvironmental molecules. Among these, nuclear factor‑κB (NF‑κB) has been indicated as a key player of osteopontin (OPN) and matrix metalloproteinase‑9 (MMP‑9) activation. However, whether NF‑κB plays a role in the development and progression of melanoma in association with the OPN/MMP‑9 axis according to the BRAFV600E mutation status has not been investigated in detail to date. Thus, in the present study, in order to shed light on this matter, 148 patients with melanoma and 53 healthy donors were recruited for the analysis of OPN, MMP‑9 and NF‑κB. Significantly higher circulating levels of OPN and MMP‑9 were observed in the patients with melanoma when compared to the healthy donors. Similar data were obtained for NF‑κB p65 activity. The OPN levels did not differ significantly between melanomas with or without BRAFV600E mutation. However, as regards NF‑κB and MMP‑9, significant differences were observed between the melanomas with or without BRAFV600E mutation. To determine whether NF‑κB inhibition is associated with a decrease in the levels of OPN and MMP‑9, peripheral blood mononuclear cells from 29 patients with melanoma were treated with the NF‑κB inhibitor, dehydroxymethylepoxyquinomycin (DHMEQ), with or without OPN. As expected, the inhibition of NF‑κB induced a marked decrease in both the OPN and MMP‑9 levels. Furthermore, the decrease in MMP‑9 levels was higher among melanomas harbouring the BRAFV600E mutation. Overall, our data suggest that the activation of MMP‑9 is associated with the BRAFV600E mutation status. Furthermore, such an activation is mediated by NF‑κB, suggesting its role as therapeutic target in patients with melanoma.
Collapse
Affiliation(s)
- Claudio Guarneri
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, I-98125 Messina, Italy
| | - Valentina Bevelacqua
- Department of Biomedical and Biotechnological Sciences, Section of General and Clinical Pathology and Oncology, University of Catania, I-95124 Catania, Italy
| | - Jerry Polesel
- Unit of Cancer Epidemiology, CRO Aviano National Cancer Institute, IRCCS, I-33081 Aviano, Italy
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences, Section of General and Clinical Pathology and Oncology, University of Catania, I-95124 Catania, Italy
| | - Patrizia S. Cannavò
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, I-98125 Messina, Italy
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion 71003, Greece
| | - Grazia Malaponte
- Department of Biomedical and Biotechnological Sciences, Section of General and Clinical Pathology and Oncology, University of Catania, I-95124 Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Section of General and Clinical Pathology and Oncology, University of Catania, I-95124 Catania, Italy
| |
Collapse
|
62
|
Potential therapeutic targets of epithelial-mesenchymal transition in melanoma. Cancer Lett 2017; 391:125-140. [PMID: 28131904 DOI: 10.1016/j.canlet.2017.01.029] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/02/2017] [Accepted: 01/18/2017] [Indexed: 12/16/2022]
Abstract
Melanoma is a cutaneous neoplastic growth of melanocytes with great potential to invade and metastasize, especially when not treated early and effectively. Epithelial-mesenchymal transition (EMT) is the process by which melanocytes lose their epithelial characteristics and acquire mesenchymal phenotypes. Mesenchymal protein expression increases the motility, invasiveness, and metastatic potential of melanoma. Many pathways play a role in promotion of mesenchymal protein expression including RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, Wnt/β-catenin, and several others. Downstream effectors of these pathways induce expression of EMT transcription factors including Snail, Slug, Twist, and Zeb that promote repression of epithelial and induction of mesenchymal character. Emerging research has demonstrated that a variety of small molecule inhibitors as well as phytochemicals can influence the progression of EMT and may even reverse the process, inducing re-expression of epithelial markers. Phytochemicals are of particular interest as supplementary treatment options because of their relatively low toxicities and anti-EMT properties. Modulation of EMT signaling pathways using synthetic small molecules and phytochemicals is a potential therapeutic strategy for reducing the aggressive progression of metastatic melanoma. In this review, we discuss the emerging pathways and transcription factor targets that regulate EMT and evaluate potential synthetic small molecules and naturally occurring compounds that may reduce metastatic melanoma progression.
Collapse
|
63
|
Daud A, Gill J, Kamra S, Chen L, Ahuja A. Indirect treatment comparison of dabrafenib plus trametinib versus vemurafenib plus cobimetinib in previously untreated metastatic melanoma patients. J Hematol Oncol 2017; 10:3. [PMID: 28052762 PMCID: PMC5209913 DOI: 10.1186/s13045-016-0369-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 12/06/2016] [Indexed: 11/20/2022] Open
Abstract
Background Metastatic melanoma is an aggressive form of skin cancer with a high mortality rate and the fastest growing global incidence rate of all malignancies. The introduction of BRAF/MEK inhibitor combinations has yielded significant increases in PFS and OS for melanoma. However, at present, no direct comparisons between different BRAF/MEK combinations have been conducted. In light of this, an indirect treatment comparison was performed between two BRAF/MEK inhibitor combination therapies for metastatic melanoma, dabrafenib plus trametinib and vemurafenib plus cobimetinib, in order to understand the relative efficacy and toxicity profiles of these therapies. Methods A systematic literature search identified two randomized trials as suitable for indirect comparison: the coBRIM trial of vemurafenib plus cobimetinib versus vemurafenib and the COMBI-v trial of dabrafenib plus trametinib versus vemurafenib. The comparison followed the method of Bucher et al. and analyzed both efficacy (overall survival [OS], progression-free survival [PFS], and overall response rate [ORR]) and safety outcomes (adverse events [AEs]). Results The indirect comparison revealed similar efficacy outcomes between both therapies, with no statistically significant difference between therapies for OS (hazard ratio [HR] 0.94, 95% confidence interval [CI] 0.68 − 1.30), PFS (HR 1.05, 95% CI 0.79 − 1.40), or ORR (risk ratio [RR] 0.90, 95% CI 0.74 − 1.10). Dabrafenib plus trametinib differed significantly from vemurafenib plus cobimetinib with regard to the incidence of treatment-related AE (RR 0.92, 95% CI 0.87 − 0.97), any AE grade ≥3 (RR 0.71, 95% CI 0.60 − 0.85) or dose interruption/modification (RR 0.77, 95% CI 0.60 − 0.99). Several categories of AEs occurred significantly more frequently with vemurafenib plus cobimetinib, while some occurred significantly more frequently with dabrafenib plus trametinib. For severe AEs (grade 3 or above), four occurred significantly more frequently with vemurafenib plus cobimetinib and no severe AE occurred significantly more frequently with dabrafenib plus trametinib. Conclusions This indirect treatment comparison suggested that dabrafenib plus trametinib had comparable efficacy to vemurafenib plus cobimetinib but was associated with reduced adverse events.
Collapse
Affiliation(s)
- Adil Daud
- Medicine and Dermatology, University of California, 1600 Divisadero Street Rm A 743, San Francisco, CA, 94143, USA.
| | | | | | - Lei Chen
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Amit Ahuja
- PAREXEL International, Chandigarh, India
| |
Collapse
|
64
|
Fisetin, a dietary flavonoid, augments the anti-invasive and anti-metastatic potential of sorafenib in melanoma. Oncotarget 2016; 7:1227-41. [PMID: 26517521 PMCID: PMC4811456 DOI: 10.18632/oncotarget.6237] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 10/11/2015] [Indexed: 01/07/2023] Open
Abstract
Melanoma is the most aggressive and deadly form of cutaneous neoplasm due to its propensity to metastasize. Oncogenic BRAF drives sustained activation of the BRAF/MEK/ERK (MAPK) pathway and cooperates with PI3K/AKT/mTOR (PI3K) signaling to induce epithelial to mesenchymal transition (EMT), leading to cell invasion and metastasis. Therefore, targeting these pathways is a promising preventive/therapeutic strategy. We have shown that fisetin, a flavonoid, reduces human melanoma cell invasion by inhibiting EMT. In addition, fisetin inhibited melanoma cell proliferation and tumor growth by downregulating the PI3K pathway. In this investigation, we aimed to determine whether fisetin can potentiate the anti-invasive and anti-metastatic effects of sorafenib in BRAF-mutated melanoma. We found that combination treatment (fisetin + sorafenib) more effectively reduced the migration and invasion of BRAF-mutated melanoma cells both in vitro and in raft cultures compared to individual agents. Combination treatment also effectively inhibited EMT as observed by a decrease in N-cadherin, vimentin and fibronectin and an increase in E-cadherin both in vitro and in xenograft tumors. Furthermore, combination therapy effectively inhibited Snail1, Twist1, Slug and ZEB1 protein expression compared to monotherapy. The expression of MMP-2 and MMP-9 in xenograft tumors was further reduced in combination treatment compared to individual agents. Bioluminescent imaging of athymic mice, intravenously injected with stably transfected CMV-luciferase-ires-puromycin. T2A.EGFP-tagged A375 melanoma cells, demonstrated fewer lung metastases following combination treatment versus monotherapy. Our findings demonstrate that fisetin potentiates the anti-invasive and anti-metastatic effects of sorafenib. Our data suggest that fisetin may be a worthy adjuvant chemotherapy for the management of melanoma.
Collapse
|
65
|
Wang E, Wang D, Li B, Ma H, Wang C, Guan L, Zhang H, Yi L, Li S. Capn4 promotes epithelial-mesenchymal transition in human melanoma cells through activation of the Wnt/β-catenin pathway. Oncol Rep 2016; 37:379-387. [PMID: 27878263 DOI: 10.3892/or.2016.5247] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/01/2016] [Indexed: 11/06/2022] Open
Abstract
Melanoma, as one of the most highly metastatic types of cancer, is resistant to current treatment methods, including popular targeted molecular therapy. Consequently, it is essential to develop a deeper understanding of the mechanisms involved in melanoma progression so that alternative treatments may be identified. To date, accumulating evidence supports the use of calpains, including calpain small subunit 1 (also known as Capn4 or CAPNS1), which affect cancer progression through many pathways, such as epithelial‑mesenchymal transition (EMT), the Wnt/β-catenin (β-catenin) and the nuclear factor κB (NF-κB) signaling pathways. The EMT pathway is well known as one of the most important events in tumor metastasis. The present study observed cross-talk among the EMT, β-catenin and NF-κB pathways. To identify the underlying mechanisms of Capn4 activity in melanoma cells, we determined Capn4 expression by gene chip and immunohistochemistral analyses in melanoma tissues and cells in vitro. The extent of apoptosis as determined by TUNEL assay, DAPI staining, and cleaved-caspase-3 assay was increased in human melanoma cells in which Capn4 expression had been knocked down when compared with untreated cells. Transwell assays and xenograft tumorigenicity studies were also performed to assess the effects of Capn4 on migration and invasion in vitro and tumor growth in vivo, respectively. The levels of β-catenin, vimentin, E-cadherin and N-cadherin were altered in human melanoma cells as determined by western blot analysis assay. Our study demonstrated that Capn4 is an underlying target for melanoma treatment.
Collapse
Affiliation(s)
- Enwen Wang
- Department of Radiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Donglin Wang
- Department of Medical Oncology, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Bing Li
- Department of Ear-Nose-Throat, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Huiwen Ma
- Department of Medical Oncology, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Chunmei Wang
- Department of Medical Oncology, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Lili Guan
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Haiwei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Lin Yi
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Hospital and Institute and Cancer Center, Chongqing 400030, P.R. China
| | - Shaolin Li
- Department of Radiology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
66
|
Liu Y, Chu Z, Li Q, Peng B, Xu S, Lian CG, Geng S. Downregulation of Bmi-1 suppresses epithelial‑mesenchymal transition in melanoma. Oncol Rep 2016; 37:139-146. [PMID: 27878257 DOI: 10.3892/or.2016.5244] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/08/2016] [Indexed: 01/28/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) contributes to the invasion and metastasis of numerous malignant cancers, including melanoma. A significant higher expression of B-lymphoma Moloney murine leukemia virus insertion region-1 (Bmi-1) has been reported in cell lines from metastatic melanoma compared to cell lines from primary melanoma. There are studies that show that knockdown of Bmi-1 could induce E-cadherin expression in melanoma cells. However, the role of Bmi-1 in mediating EMT-like changes in melanoma has not yet been fully studied. In the present study, knockdown of Bmi-1 by shRNA transduction decreased the invasion properties of the cultured human melanoma cells A375 by a Matrigel invasion assay, along with alterations in EMT-related markers E-cadherin, α-catenin, vimentin and N-cadherin. The aforementioned altered expression of EMT markers was verified in BALB/c-nude mouse xenografts. Furthermore, to explore the underlying regulatory mechanism of EMT, we detected the significant downregulation of p-Akt/p‑NF-κB/MMP-2 and the upregulation of PTEN in Bmi-1-silenced A375 cells. The present study demonstrated that knockdown of Bmi-1 significantly inhibited the aggressive behavior of melanoma by reversing EMT-like changes via the PTEN/p-Akt/p‑NF-κB/MMP-2 pathway.
Collapse
Affiliation(s)
- Yanting Liu
- Department of Dermatology, Northwest Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zhaowei Chu
- Department of Dermatology, Northwest Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Qingyan Li
- Department of Dermatology, Northwest Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Bin Peng
- Department of Dermatology, Northwest Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Suyun Xu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christine G Lian
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Songmei Geng
- Department of Dermatology, Northwest Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
67
|
Patel DH, Sharma N. Inhibitory effect of quercetin on epithelial to mesenchymal transition in SK-MEL-28 human melanoma cells defined by in vitro analysis on 3D collagen gels. Onco Targets Ther 2016; 9:6445-6459. [PMID: 27799792 PMCID: PMC5077264 DOI: 10.2147/ott.s109253] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Considering the emerging concept of complementary and alternative medicine under the paucity of effective treatment for melanoma, we aimed to understand the effect of quercetin (Qu) on collagen I-induced epithelial-mesenchymal transition (EMT) in melanoma cells. To investigate the effect of Qu in melanoma cells, we used multiple methods, including real-time reverse transcription polymerase chain reaction, migration assay, and wound healing assay. We found that EMT was altered by Qu in melanoma cells. Qu-treated cells exhibited decreased migration and invasion activities. Mechanistically, a high expression of epithelial markers and a decrease in the expression of mesenchymal markers were found to be associated with reversal of EMT in melanoma cells. Time-dependent apoptosis was observed in Qu-treated melanoma cells, which was further confirmed by the upregulation in the protein levels of Caspase 3, a proapoptotic marker. Thus, our findings suggest Qu as a promising dietary compound under the new complementary and alternative medicine category of therapeutic drugs in the chemoprevention of melanoma.
Collapse
Affiliation(s)
- Dhairya H Patel
- Symbiosis School of Biomedical Sciences, Symbiosis International University, Gram - Lavale, Taluka - Mulshi, Pune, India
| | - Neeti Sharma
- Symbiosis School of Biomedical Sciences, Symbiosis International University, Gram - Lavale, Taluka - Mulshi, Pune, India
| |
Collapse
|
68
|
Pieniazek M, Donizy P, Halon A, Leskiewicz M, Matkowski R. Prognostic significance of immunohistochemical epithelial–mesenchymal transition markers in skin melanoma patients. Biomark Med 2016; 10:975-85. [DOI: 10.2217/bmm-2016-0133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To investigate secreted protein acidic and rich in cystein (SPARC) and neural cadherin (NCAD), which are associated with epithelial–mesenchymal transition in primary skin melanoma and nodal metastases and their prognostic impact in melanoma patients. Methods: Expression of proteins was assessed by immunochemistry in archival paraffin samples from 103 primary melanoma tumors and 16 nodal metastases. Results: Increased expression of SPARC and NCAD in primary skin melanoma was associated with decreased overall survival, adverse clinicopathological features and particularly with microsatellitosis (SPARC) and ulceration (NCAD). In univariate Cox regression analysis, both biomarkers were significantly associated with the risk of death; the multivariate Cox regression analysis identified no significance. Conclusion: The most important result of our study was that we confirmed the strict correlation between SPARC and NCAD expression and clinicopathological parameters related with melanoma progression, which is a specific clinical equivalent of the molecular mechanisms of epithelial–mesenchymal transition process and confirms its key role in the disease outcome.
Collapse
Affiliation(s)
- Malgorzata Pieniazek
- Department of Clinical Oncology, Tadeusz Koszarowski Regional Oncology Center, Opole, Katowicka 66a, Poland
| | - Piotr Donizy
- Department of Pathomorphology & Oncological Cytology, Wroclaw Medical University, Borowska 213, 50–556 Wroclaw, Poland
| | - Agnieszka Halon
- Department of Pathomorphology & Oncological Cytology, Wroclaw Medical University, Borowska 213, 50–556 Wroclaw, Poland
| | - Marek Leskiewicz
- Department of Statistics, Wroclaw University of Economics, Komandorska 118–120, 53–345 Wroclaw, Poland
| | - Rafal Matkowski
- Department of Oncology & Division of Surgical Oncology, Wroclaw Medical University, pl. Hirszfelda 12, 53–413 Wroclaw, Poland
- Lower Silesian Oncology Centre, pl. Hirszfelda 12, 53–413 Wroclaw, Poland
| |
Collapse
|
69
|
Deltex-3-like (DTX3L) stimulates metastasis of melanoma through FAK/PI3K/AKT but not MEK/ERK pathway. Oncotarget 2016; 6:14290-9. [PMID: 26033450 PMCID: PMC4546467 DOI: 10.18632/oncotarget.3742] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/08/2015] [Indexed: 12/19/2022] Open
Abstract
Deltex-3-like (DTX3L), an E3 ligase, is a member of the Deltex (DTX) family and is also called B-lymphoma and BAL-associated protein (BBAP). Previously, we established RFP/RET-transgenic mice, in which systemic hyperpigmented skin, benign melanocytic tumor(s) and melanoma(s) develop stepwise. Here we showed that levels of Dtx3l/DTX3L in spontaneous melanoma in RFP/RET-transgenic mice and human melanoma cell lines were significantly higher than those in benign melanocytic cells and primarily cultured normal human epithelial melanocytes, respectively. Immunohistochemical analysis of human tissues showed that more than 80% of the melanomas highly expressed DTX3L. Activity of FAK/PI3K/AKT signaling, but not that of MEK/ERK signaling, was decreased in Dtx3l/DTX3L-depleted murine and human melanoma cells. In summary, we demonstrated not only increased DTX3L level in melanoma cells but also DTX3L-mediated regulation of invasion and metastasis in melanoma through FAK/PI3K/AKT but not MEK/ERK signaling. Our analysis in human BRAFV600E inhibitor-resistant melanoma cells showed about 80% decreased invasion in the DTX3L-depleted cells compared to that in the DTX3L-intact cells. Thus, DTX3L is clinically a potential therapeutic target as well as a potential biomarker for melanoma.
Collapse
|
70
|
Jia D, Jolly MK, Boareto M, Parsana P, Mooney SM, Pienta KJ, Levine H, Ben-Jacob E. OVOL guides the epithelial-hybrid-mesenchymal transition. Oncotarget 2016; 6:15436-48. [PMID: 25944618 PMCID: PMC4558162 DOI: 10.18632/oncotarget.3623] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/10/2015] [Indexed: 01/25/2023] Open
Abstract
Metastasis involves multiple cycles of Epithelial-to-Mesenchymal Transition (EMT) and its reverse-MET. Cells can also undergo partial transitions to attain a hybrid epithelial/mesenchymal (E/M) phenotype that has maximum cellular plasticity and allows migration of Circulating Tumor Cells (CTCs) as a cluster. Hence, deciphering the molecular players helping to maintain the hybrid E/M phenotype may inform anti-metastasis strategies. Here, we devised a mechanism-based mathematical model to couple the transcription factor OVOL with the core EMT regulatory network miR-200/ZEB that acts as a three-way switch between the E, E/M and M phenotypes. We show that OVOL can modulate cellular plasticity in multiple ways - restricting EMT, driving MET, expanding the existence of the hybrid E/M phenotype and turning both EMT and MET into two-step processes. Our theoretical framework explains the differences between the observed effects of OVOL in breast and prostate cancer, and provides a platform for investigating additional signals during metastasis.
Collapse
Affiliation(s)
- Dongya Jia
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA.,Graduate Program in Systems, Synthetic and Physical Biology, Rice University, Houston, TX 77005, USA
| | - Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA.,Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Marcelo Boareto
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA.,Institute of Physics, University of Sao Paulo, Sao Paulo 05508, Brazil
| | - Princy Parsana
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Steven M Mooney
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA.,Department of Bioengineering, Rice University, Houston, TX 77005, USA.,Department of Physics and Astronomy, Rice University, Houston, TX 77005, USA
| | - Eshel Ben-Jacob
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA.,Department of Biosciences, Rice University, Houston, TX 77005, USA.,School of Physics and Astronomy and The Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
71
|
Xiao D, Barry S, Kmetz D, Egger M, Pan J, Rai SN, Qu J, McMasters KM, Hao H. Melanoma cell-derived exosomes promote epithelial-mesenchymal transition in primary melanocytes through paracrine/autocrine signaling in the tumor microenvironment. Cancer Lett 2016; 376:318-27. [PMID: 27063098 DOI: 10.1016/j.canlet.2016.03.050] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 12/30/2022]
Abstract
The tumor microenvironment is abundant with exosomes that are secreted by the cancer cells themselves. Exosomes are nanosized, organelle-like membranous structures that are increasingly being recognized as major contributors in the progression of malignant neoplasms. A critical element in melanoma progression is its propensity to metastasize, but little is known about how melanoma cell-derived exosomes modulate the microenvironment to optimize conditions for tumor progression and metastasis. Here, we provide evidence that melanoma cell-derived exosomes promote phenotype switching in primary melanocytes through paracrine/autocrine signaling. We found that the mitogen-activated protein kinase (MAPK) signaling pathway was activated during the exosome-mediated epithelial-to-mesenchymal transition (EMT)-resembling process, which promotes metastasis. Let-7i, an miRNA modulator of EMT, was also involved in this process. We further defined two other miRNA modulators of EMT (miR-191 and let-7a) in serum exosomes for differentiating stage I melanoma patients from non-melanoma subjects. These results provide the first strong molecular evidence that melanoma cell-derived exosomes promote the EMT-resembling process in the tumor microenvironment. Thus, novel strategies targeting EMT and modulating the tumor microenvironment may emerge as important approaches for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Deyi Xiao
- The Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Kosair Charities Clinical & Translational Research Building, 505 S. Hancock St, Room 407, Louisville, KY 40292, USA
| | - Samantha Barry
- The Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Kosair Charities Clinical & Translational Research Building, 505 S. Hancock St, Room 407, Louisville, KY 40292, USA
| | - Daniel Kmetz
- The Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Kosair Charities Clinical & Translational Research Building, 505 S. Hancock St, Room 407, Louisville, KY 40292, USA
| | - Michael Egger
- The Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Kosair Charities Clinical & Translational Research Building, 505 S. Hancock St, Room 407, Louisville, KY 40292, USA
| | - Jianmin Pan
- Biostatistics Shared Facility, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Shesh N Rai
- Biostatistics Shared Facility, James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Jifu Qu
- The Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Kosair Charities Clinical & Translational Research Building, 505 S. Hancock St, Room 407, Louisville, KY 40292, USA
| | - Kelly M McMasters
- The Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Kosair Charities Clinical & Translational Research Building, 505 S. Hancock St, Room 407, Louisville, KY 40292, USA.
| | - Hongying Hao
- The Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Kosair Charities Clinical & Translational Research Building, 505 S. Hancock St, Room 407, Louisville, KY 40292, USA.
| |
Collapse
|
72
|
Hallums DP, Gomez R, Doyle AP, Viet CT, Schmidt BL, Jeske NA. RAF Kinase Inhibitory Protein Expression and Phosphorylation Profiles in Oral Cancers. CLINICS IN SURGERY 2016; 1:1100. [PMID: 28529999 PMCID: PMC5436720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Raf Kinase Inhibitory Protein (RKIP) expression has been profiled for a number of unique tissue cancers. However, certain tissues have not been explored, and oral and oropharyngeal cancers stand out as high priority targets, given their relatively high incidence, high morbidity rate, and in many cases, preventable nature. The purpose of this study was to examine changes in RKIP expression and phosphorylation in tissues resected from oral cancer patients, and compare to results generated from immortalized cell lines raised from primary oral cancer tissues, including oral squamous cell carcinoma line 4 (SCC4) and human squamous cell carcinoma line 3 (HSC3). Out of 4 human samples collected from male and female patients across various ages with variable risk factors, we observed an across the board reduction in RKIP expression. Two human samples demonstrated a significant increase in phosphorylated RKIP when normalized to total RKIP, however all 4 were increased when normalized to total cellular protein. The immortalized oral cancer cell culture HSC3 revealed significant increases in phosphorylated RKIP with no change in total RKIP expression, while line SCC4 demonstrated an increase in both total and phosphorylated RKIP. Results presented here indicate that oral cancers behave similarly to other cancers in terms of changes in RKIP expression and phosphorylation, although immortalized cell line expression profiles significantly differ from human tissue biopsies.
Collapse
Affiliation(s)
- DP Hallums
- Departments of Oral and Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, USA
| | - R Gomez
- Departments of Oral and Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, USA
| | - AP Doyle
- Departments of Pharmacology, University of Texas Health Science Center at San Antonio, USA
| | - CT Viet
- Department of Oral Maxillofacial Surgery, New York University, USA,Department of Oral Maxillofacial Surgery, Bluestone Center for Clinical Research, New York University, USA
| | - BL Schmidt
- Department of Oral Maxillofacial Surgery, New York University, USA,Department of Oral Maxillofacial Surgery, Bluestone Center for Clinical Research, New York University, USA
| | - NA Jeske
- Departments of Oral and Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, USA,Departments of Pharmacology, University of Texas Health Science Center at San Antonio, USA,Departments of Physiology, University of Texas Health Science Center at San Antonio, USA,Correspondence: Nathaniel A. Jeske, Department of Oral & Maxillofacial Surgery, University of Texas Health Science Center of San Antonio, Center for Biomedical Neuroscience, 7703 Floyd Curl Dr., San Antonio, TX 78229-3900, USA, Tel: (210) 567-3466; Fax: (210) 567-2995;
| |
Collapse
|
73
|
Baquero P, Jiménez-Mora E, Santos A, Lasa M, Chiloeches A. TGFβ induces epithelial-mesenchymal transition of thyroid cancer cells by both the BRAF/MEK/ERK and Src/FAK pathways. Mol Carcinog 2015; 55:1639-1654. [PMID: 26392228 DOI: 10.1002/mc.22415] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/08/2015] [Accepted: 09/14/2015] [Indexed: 11/10/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is a crucial process in tumour progression, by which epithelial cells acquire a mesenchymal phenotype, increasing its motility and the ability to invade distant sites. Here, we describe the molecular mechanisms by which V600E BRAF, TGFβ and the Src/FAK complex cooperatively regulate EMT induction and cell motility of anaplastic thyroid cancer cells. Analysis of EMT marker levels reveals a positive correlation between TGFβ and Snail expression, with a concomitant downregulation of E-cadherin, accompanied by an increase of cell migration and invasion. Furthermore, we show that V600E BRAF depletion by siRNA or inhibition of its activity by treatment with its inhibitor PLX4720 reverses the TGFβ-mediated effects on Snail, E-cadherin, migration and invasion. Moreover, V600E BRAF induces TGFβ secretion through a MEK/ERK-dependent mechanism. In addition, TGFβ activates the Src/FAK complex, which in turn regulates the expression of Snail and E-cadherin as well as cell migration. The inhibition of Src with the inhibitor SU6656 or abrogation of FAK expression with a specific siRNA reverses the TGFβ-induced effects. Interestingly, we demonstrate that activation of the Src/FAK complex by TGFβ is independent of V600E BRAF signalling, since inhibition of this oncogene does not affect its phosphorylation. Our data strongly suggest that TGFβ induces EMT and aggressiveness of thyroid cancer cells by parallel mechanisms involving both the V600E BRAF/MEK/ERK and Src/FAK pathways independently. Thus, we describe novel functions for Src/FAK in mediating the EMT program and aggressiveness regulated by TGFβ, establishing the inhibition of these proteins as a possible effective approach in preventing tumour progression of V600E BRAF-expressing thyroid tumours. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pablo Baquero
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Campus Universitario, Alcalá de Henares, Madrid, Spain
| | - Eva Jiménez-Mora
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Campus Universitario, Alcalá de Henares, Madrid, Spain
| | - Adrián Santos
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Campus Universitario, Alcalá de Henares, Madrid, Spain
| | - Marina Lasa
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain
| | - Antonio Chiloeches
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Campus Universitario, Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
74
|
Antitumor Effects and Related Mechanisms of Penicitrinine A, a Novel Alkaloid with a Unique Spiro Skeleton from the Marine Fungus Penicillium citrinum. Mar Drugs 2015; 13:4733-53. [PMID: 26264002 PMCID: PMC4557002 DOI: 10.3390/md13084733] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/01/2015] [Accepted: 07/01/2015] [Indexed: 12/13/2022] Open
Abstract
Penicitrinine A, a novel alkaloid with a unique spiro skeleton, was isolated from a marine-derived fungus Penicillium citrinum. In this study, the isolation, structure and biosynthetic pathway elucidation of the new compound were described. This new compound showed anti-proliferative activity on multiple tumor types. Among them, the human malignant melanoma cell A-375 was confirmed to be the most sensitive. Morphologic evaluation, apoptosis rate analysis, Western blot and real-time quantitative PCR (RT-qPCR) results showed penicitrinine A could significantly induce A-375 cell apoptosis by decreasing the expression of Bcl-2 and increasing the expression of Bax. Moreover, we investigated the anti-metastatic effects of penicitrinine A in A-375 cells by wound healing assay, trans-well assay, Western blot and RT-qPCR. The results showed penicitrinine A significantly suppressed metastatic activity of A-375 cells by regulating the expression of MMP-9 and its specific inhibitor TIMP-1. These findings suggested that penicitrinine A might serve as a potential antitumor agent, which could inhibit the proliferation and metastasis of tumor cells.
Collapse
|
75
|
Chen SY, Shiau AL, Li YT, Lin CC, Jou IM, Liu MF, Wu CL, Wang CR. Transcription factor snail regulates tumor necrosis factor α-mediated synovial fibroblast activation in the rheumatoid joint. Arthritis Rheumatol 2015; 67:39-50. [PMID: 25303734 DOI: 10.1002/art.38899] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 09/25/2014] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The transcription factor Snail is involved in various biologic functions. We hypothesized that this molecule regulates tumor necrosis factor α (TNFα)-mediated synovial fibroblast activation in the rheumatoid joint. The aim of this study was to examine the role of Snail in the expression of cadherin-11 (Cad-11) and myofibroblast markers, interleukin-6 (IL-6) production, and the invasive ability of cells. METHODS Synovium samples were obtained from patients with rheumatoid arthritis (RA) and from rats with collagen-induced arthritis (CIA). Synovial fibroblasts were treated with TNFα or a Wnt signaling inducer, and the joints of rats with CIA were injected with a TNFα antagonist. Modulation of Snail expression in the synovial fibroblasts and joints was performed by lentiviral vector-mediated transfer of complementary DNA or short hairpin RNA. RESULTS The expression of Snail and Cad-11 was higher in synovium and synovial fibroblasts from patients with RA compared with patients with osteoarthritis and was increased in rats with CIA. TNFα stimulation or activation of Wnt signaling up-regulated the expression of Snail, Cad-11, and α-smooth muscle actin (α-SMA) in synovial fibroblasts, and anti-TNFα therapy down-regulated the expression of Snail, Cad-11, and α-SMA in the joints of rats with CIA. Although synovial fibroblast transfectants in which Snail was overexpressed showed increased expression of Cad-11 and α-SMA and enhanced TNFα-mediated invasive capacity and IL-6 production, synovial fibroblast transfectants from rats with CIA in which Snail was silenced showed decreased expression and had the opposite effect on these functions. Normal joints in which Snail was overexpressed had hyperplastic synovium, with increased expression of Cad-11, α-SMA, and IL-6. Silencing Snail expression ameliorated arthritis, with reduced Cad-11 expression and reduced levels of extracellular matrix deposition in the joints of rats with CIA, whereas overexpression of Snail exacerbated arthritis, with increased Cad-11 expression and increased levels of extracellular matrix deposition. CONCLUSION Our results demonstrate that Snail regulates TNFα-mediated activation of synovial fibroblasts in the rheumatoid joint. These findings may contribute to the pharmacologic development of therapeutics targeting synovial fibroblasts in patients with RA.
Collapse
Affiliation(s)
- Shih-Yao Chen
- National Cheng Kung University Medical College, Tainan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Bonavida B. RKIP-mediated chemo-immunosensitization of resistant cancer cells via disruption of the NF-κB/Snail/YY1/RKIP resistance-driver loop. Crit Rev Oncog 2015; 19:431-45. [PMID: 25597353 DOI: 10.1615/critrevoncog.2014011929] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cancer remains one of the most dreadful diseases. Whereas most treatment regimens for various cancers have resulted in improved clinical responses and sometimes cures, unfortunately, subsets of cancer patients are either pretreatment resistant or develop resistance following therapy. These subsets of patients develop cross-resistance to unrelated therapeutics and usually succumb to death. Thus, delineating the underlying molecular mechanisms of resistance of various cancers and identifying molecular targets for intervention are the current main focus of research investigations. One approach to investigate cancer resistance has been to identify pathways that regulate resistance and develop means to disrupt these pathways in order to override resistance and sensitize the resistant cells to cell death. Hence, we have identified one pathway that is dysregulated in cancer, namely, the NF-κB/Snail/YY1/RKIP loop, that has been shown to regulate, in large part, tumor cell resistance to apoptosis by chemotherapeutic and immunotherapeutic cytotoxic drugs. The dysregulated resistant loop is manifested by the overexpression of NF-κB, Snail, and YY1 activities and the underexpression of RKIP. The induction of RKIP expression results in the downregulation of NF-κB, Snail, and YY1 and the sensitization of resistant cells to drug-induced apoptosis. These findings identified RKIP, in addition to its antiproliferative and metastatic suppressor functions, as an anti-resistance factor. This brief review describes the role of RKIP in the regulation of drug sensitivity via disruption of the NF-κB/Snail/ YY1/RKIP loop that regulates resistance in cancer cells.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, USA
| |
Collapse
|
77
|
Li FZ, Dhillon AS, Anderson RL, McArthur G, Ferrao PT. Phenotype switching in melanoma: implications for progression and therapy. Front Oncol 2015; 5:31. [PMID: 25763355 PMCID: PMC4327420 DOI: 10.3389/fonc.2015.00031] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/30/2015] [Indexed: 02/04/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a key process associated with the progression of epithelial cancers to metastatic disease. In melanoma, a similar process of phenotype switching has been reported and EMT-related genes have been implicated in promotion to a metastatic state. This review examines recent research on the role of signaling pathways and transcription factors regulating EMT-like processes in melanoma and their association with response to therapy in patients, especially response to BRAF inhibition, which is initially effective but limited by development of resistance and subsequent progression. We highlight studies implicating specific roles of various receptor tyrosine kinases (RTKs) in advancing melanoma progression by conferring a proliferative advantage and through promoting invasive phenotypes and metastasis. We also review the current knowledge of the mechanisms underlying resistance to BRAF inhibition and the potential role of melanoma phenotype switching in this process. In particular, we discuss how these important new insights may significantly enhance our ability to predict patterns of melanoma progression during treatment, and may facilitate rational development of combination therapies in the future.
Collapse
Affiliation(s)
- Frederic Zhentao Li
- Oncogenic Signaling and Growth Control Program, Research Division, Peter MacCallum Cancer Center , East Melbourne, VIC , Australia ; Sir Peter MacCallum Department of Oncology, The University of Melbourne , East Melbourne, VIC , Australia
| | - Amardeep Singh Dhillon
- Oncogenic Signaling and Growth Control Program, Research Division, Peter MacCallum Cancer Center , East Melbourne, VIC , Australia ; Sir Peter MacCallum Department of Oncology, The University of Melbourne , East Melbourne, VIC , Australia ; Department of Pathology, The University of Melbourne , East Melbourne, VIC , Australia
| | - Robin L Anderson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne , East Melbourne, VIC , Australia ; Metastasis Research Laboratory, Research Division, Peter MacCallum Cancer Center , East Melbourne, VIC , Australia
| | - Grant McArthur
- Oncogenic Signaling and Growth Control Program, Research Division, Peter MacCallum Cancer Center , East Melbourne, VIC , Australia ; Sir Peter MacCallum Department of Oncology, The University of Melbourne , East Melbourne, VIC , Australia ; Department of Pathology, The University of Melbourne , East Melbourne, VIC , Australia ; Department of Medicine, St Vincent's Hospital, The University of Melbourne , East Melbourne, VIC , Australia
| | - Petranel T Ferrao
- Oncogenic Signaling and Growth Control Program, Research Division, Peter MacCallum Cancer Center , East Melbourne, VIC , Australia ; Sir Peter MacCallum Department of Oncology, The University of Melbourne , East Melbourne, VIC , Australia ; Department of Pathology, The University of Melbourne , East Melbourne, VIC , Australia
| |
Collapse
|
78
|
Liu H, Li P, Li B, Sun P, Zhang J, Wang B, Jia B. RKIP promotes cisplatin-induced gastric cancer cell death through NF-κB/Snail pathway. Tumour Biol 2014; 36:1445-53. [PMID: 25547433 DOI: 10.1007/s13277-014-2496-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 08/13/2014] [Indexed: 01/29/2023] Open
Abstract
The objectives of this study were to explore the expression profiles of Raf kinase inhibitor protein (RKIP) in human gastric cancer cell line (SGC-7901) and cisplatin-resistant cell line (SGC-7901/DDP) and investigate the role of RKIP in the sensitivity of human gastric cancer cells to cisplatin and its signaling pathways, with an attempt to identify new approaches and strategies for the management of gastric cancer. The human gastric cancer cell line (SGC-7901) and cisplatin-resistant cell line (SGC-7901/DDP) were separately cultured in vitro. The expression profiles of RKIP in these two cell lines were detected by Western blotting. Forty-eight hours after the transfection of RKIP siRNA in SGC-7901 cells, the change of RKIP expression in the cells was detected using Western blotting, and the change of cell viability after the interference of RKIP expression was determined using 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) method. The effect of the ectopic expression of RKIP on the cisplatin-induced viability of gastric cancer cell was detected using MTT method. The effect of the ectopic expression of RKIP on the cisplatin-induced apoptosis of gastric cancer cell was detected using flow cytometry after having been double stained with Annexin V/PI. The effect of the ectopic expression of RKIP on the NF-κB and Snail expressions in cisplatin-induced gastric cancer cells was detected using Western blotting. As shown by the Western blotting, the expression of RKIP in SGC-7901/DDP cells significantly decreased when compared with that in SGC-7901 cells (P < 0.05). Compared with the control group, the expression of RKIP in SGC-7901 cells significantly decreased 48 h after the transfection of RKIP siRNA (P < 0.01). After the SGC-7901 cells were transfected with RKIP siRNA, the cell viability was significantly increased (P < 0.05); after the SGC-7901/DDP cells were transfected with RKIP recombinant plasmid, the cell viability was significantly decreased (P < 0.05). After the RKIP expression was suppressed in the cisplatin-treated SGC-7901 cells, the cell viability significantly increased (P < 0.05), and the amount of apoptotic cells significantly decreased (P < 0.05). In contrast, after the RKIP overexpression in the cisplatin-treated SGC-7901/DDP cells, the cell viability significantly decreased (P < 0.05), and the amount of apoptotic cells significantly increased (P < 0.05). The suppression of RKIP expression in SGC-7901 cells could significantly promote the increase of NF-κB expression (P < 0.05); in contrast, the increased expression of RKIP in SGC-7901/DDP cells significantly inhibited the expression of Snail (P < 0.05). The expression of RKIP is downregulated in cisplatin-resistant cell line (SGC-7901/DDP). The overexpression of RKIP can enhance the sensitivity of human gastric cancer cells to cisplatin, which may be achieved via the NF-κB/Snail signaling pathway.
Collapse
Affiliation(s)
- Hongyi Liu
- Department of Surgical Oncology, General Hospital of Chinese People's Liberation Army, No. 28, Fuxing Rd, Beijing, 100853, China
| | | | | | | | | | | | | |
Collapse
|
79
|
Lei YY, Wang WJ, Mei JH, Wang CL. Mitogen-Activated Protein Kinase Signal Transduction in Solid Tumors. Asian Pac J Cancer Prev 2014; 15:8539-48. [DOI: 10.7314/apjcp.2014.15.20.8539] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
80
|
Liu H, Li P, Li B, Sun P, Zhang J, Wang B, Jia B. RKIP inhibits gastric cancer cell survival and invasion by regulating the expression of HMGA2 and OPN. Tumour Biol 2014; 35:11949-58. [PMID: 25172097 DOI: 10.1007/s13277-014-2486-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/08/2014] [Indexed: 12/28/2022] Open
Abstract
The objective of this study was to explore the mechanism via which Raf kinase inhibitor protein (RKIP) suppresses the invasion of gastric cancer cells and promote apoptosis, with an attempt to provide evidences for the application of RKIP in treating gastric cancer. The recombinant plasmid pcDNA3.1-RKIP or RKIP-shRNA was transfected into the gastric cancer cell line SGC-7901 using liposome. Then, the messenger RNA (mRNA) and protein expressions of RKIP, HMGA2, and OPN were detected using qPCR and Western blotting. The effects of HMGA2 on the proliferation, apoptosis, and invasion of SGC-7901 cells were detected using flow cytometry and Transwell assay. To further explore the regulatory effect of PKIP on the biological activities of HMGA2, we over-expressed or knock down RKIP and HMGA2 simultaneously and detected its effects on the proliferation, apoptosis, and invasion of SGC-7901 cells. As shown by qPCR and Western blotting, after over-expression of RKIP in SGC-7901 cells, the mRNA and protein expressions of RKIP significantly increased (P < 0.01), whereas the mRNA and protein expressions of HMGA2 and OPN significantly decreased (P < 0.01). In contrast, the transfection of RKIP-shRNA in the SGC-7901 cells resulted in opposite results. After over-expression of HMGA2 in SGC-7901 cells, the protein expression of HMGA2 significantly increased (P < 0.01); however, it significantly decreased after the transfection of HMGA2-shRNA (P < 0.01). As shown by Transwell assay and flow cytometry, After the over-expression of HMGA2 in SGC-7901 cells, the (G2 + S) phase fraction significantly increased (P < 0.01); also, the percentage of the apoptotic cells significantly declined (P < 0.05) and the number of invasive cells significantly increased (P < 0.05). However, the interference of the expression of HMGA2 resulted in opposite results. The simultaneous over-expression of RKIP and HMGA2 in SGC-7901 cells or the simultaneous interference of RKIP and HMGA2 showed no significant difference with the control group in terms of (G2 + S) phase fraction, percentage of apoptotic cells, and number of invasive cells (P > 0.05). In conclusion, RKIP can inhibit the survival and invasion of gastric cancer cells and promote apoptosis, possibly by regulating the expression of HMGA2 or OPN.
Collapse
Affiliation(s)
- Hongyi Liu
- Department of Surgical Oncology, General Hospital of Chinese People's Liberation Army, No. 28, Fuxing Rd, Beijing, 100853, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
81
|
Liu H, Li P, Li B, Sun P, Zhang J, Wang B, Jia B. RKIP suppresses gastric cancer cell proliferation and invasion and enhances apoptosis regulated by microRNA-224. Tumour Biol 2014; 35:10095-103. [PMID: 25017365 DOI: 10.1007/s13277-014-2303-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 07/01/2014] [Indexed: 12/26/2022] Open
Abstract
The purposes of this study were to determine the expression profile of Raf kinase inhibitor protein (RKIP) in human gastric cancer cells and its effect on the biological characteristics of SGC-7901 cell lines, to examine the modulatory effect of microRNA-224 (miR-224) on RKIP. The research will provide novel strategies for gastric cancer treatment in the future. Quantitative real-time reverse transcription PCR (qRT-PCR) was employed to determine the expression profile of RKIP in gastric cancer cell lines (SGC-7901, MGC80-3, and MKN45). A eukaryotic expression vector, pcDNA3.1-RKIP, was constructed and transfected into SGC-7901 cells. Changes in RKIP protein expression were examined by Western blot assays, and the effect of RKIP overexpression on SCG-7901 cell viability was determined by 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-diphenytetrazoliumromide (MTT) assays. The effect of RKIP overexpression on SGC-7901 cell proliferation and apoptosis was analyzed by flow cytometry and that on the migration of SGC-7901 cells was investigated by Transwell migration assays. RKIP was identified to be a regulatory target gene of miR-224 using a luciferase reporter gene system, and the effect of miR-224 on intracellular RKIP protein expression was examined by Western blot assays. The regulatory effect of miR-224 on the biological characteristics of RKIP was investigated by MTT, flow cytometry, and Transwell invasion chamber assays. The expression of RKIP in gastric cancer cells was decreased significantly in comparison to that of normal gastric mucosal epithelial cells (GES-1) (p < 0.01), as demonstrated by qRT-PCR assays. Compared with the control group, the up-regulation of RKIP intracellular expression was observed in SGC-7901 cells after transfection of pcDNA3.1-RKIP for 48 h (p < 0.01). There were significant decreases in cell viability and the S-phase fraction (p < 0.05), concomitant with a significant increase in apoptosis (p < 0.01), as well as a significant reduction in cells migrating through Transwell chambers (p < 0.05), as shown by MTT, flow cytometry, and Transwell invasion chamber assays. A significant decrease in luciferase activities in cells transfected with a miR-224 mimic was observed compared with that of the control group (p < 0.05), as suggested by the luciferase reporter gene system. As shown by Western blot assays, there was a significant decrease in RKIP expression in SGC-7901 cells transfected with the miR-224 mimic for 48 h compared with the control group (p < 0.05). As shown by MTT, flow cytometry, and Transwell invasion chamber assays, the changes in biological characteristics induced by RKIP overexpression could be suppressed in SGC-7901 cells after transfection of the miR-224 mimic. In conclusion, the down-regulation of RKIP expression was observed in human gastric cell lines, and miR-224 could negatively regulate the expression and biological characteristics of RKIP, contributing to suppress the proliferation and invasion of gastric cells.
Collapse
Affiliation(s)
- Hongyi Liu
- Department of Surgical Oncology, General Hospital of Chinese People's Liberation Army, No. 28, Fuxing Rd, 100853, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
82
|
Chu W, Song X, Yang X, Ma L, Zhu J, He M, Wang Z, Wu Y. Neuropilin-1 promotes epithelial-to-mesenchymal transition by stimulating nuclear factor-kappa B and is associated with poor prognosis in human oral squamous cell carcinoma. PLoS One 2014; 9:e101931. [PMID: 24999732 PMCID: PMC4084996 DOI: 10.1371/journal.pone.0101931] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 06/12/2014] [Indexed: 12/19/2022] Open
Abstract
Background The epithelial-to-mesenchymal transition (EMT) is a key process in carcinogenesis, invasion, and metastasis of oral squamous cell carcinoma (OSCC). In our previous studies, we found that neuropilin-1 (NRP1) is overexpressed in tongue squamous cell carcinoma and that this overexpression is associated with cell migration and invasion. Nuclear factor-kappa B (NF-κB) plays an essential role both in the induction and the maintenance of EMT and tumor metastasis. Therefore, we hypothesized that NRP1 induces EMT, and that NRP1-induced migration and invasion may be an important mechanism for promoting invasion and metastasis of OSCC through NF-κB activation. Methods/Results The variations in gene and protein expression and the changes in the biological behavior of OSCC cell lines transfected with a vector encoding NRP1, or the corresponding vector control, were evaluated. NRP1 overexpression promoted EMT and was associated with enhanced invasive and metastatic properties. Furthermore, the induction of EMT promoted the acquisition of some cancer stem cell (CSC)-like characteristics in OSCC cells. We addressed whether selective inhibition of NF-κB suppresses the NRP1-mediated EMT by treating cells with pyrrolidinedithiocarbamate ammonium (PDTC), an inhibitor of NF-κB. Immunohistochemical analysis of NRP1 in OSCC tissue samples further supported a key mediator role for NRP1 in tumor progression, lymph node metastasis, and indicated that NRP1 is a predictor for poor prognosis in OSCC patients. Conclusion Our results indicate that NRP1 may regulate the EMT process in OSCC cell lines through NF-κB activation, and that higher NRP1 expression levels are associated with lymph node metastasis and poor prognosis in OSCC patients. Further investigation of the role of NRP1 in tumorigenesis may help identify novel targets for the prevention and therapy of oral cancers.
Collapse
Affiliation(s)
- Weiming Chu
- Institute of Stomatology, Nanjing Medical University, Nanjing, PR China
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Jiangsu Province, Nanjing, PR China
| | - Xiaomeng Song
- Institute of Stomatology, Nanjing Medical University, Nanjing, PR China
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Jiangsu Province, Nanjing, PR China
| | - Xueming Yang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Jiangsu Province, Nanjing, PR China
| | - Lu Ma
- Institute of Stomatology, Nanjing Medical University, Nanjing, PR China
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Jiangsu Province, Nanjing, PR China
| | - Jiang Zhu
- Institute of Stomatology, Nanjing Medical University, Nanjing, PR China
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Jiangsu Province, Nanjing, PR China
| | - Mengying He
- Institute of Stomatology, Nanjing Medical University, Nanjing, PR China
- Department of Basic Science of Stomatology, College of Stomatology, Nanjing Medical University, Nanjing, PR China
| | - Zilu Wang
- Institute of Stomatology, Nanjing Medical University, Nanjing, PR China
- * E-mail: (ZW); (YW)
| | - Yunong Wu
- Institute of Stomatology, Nanjing Medical University, Nanjing, PR China
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Jiangsu Province, Nanjing, PR China
- * E-mail: (ZW); (YW)
| |
Collapse
|
83
|
Epithelial mesenchymal transition in early invasive breast cancer: an immunohistochemical and reverse phase protein array study. Breast Cancer Res Treat 2014; 145:339-48. [PMID: 24771047 DOI: 10.1007/s10549-014-2927-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 03/17/2014] [Indexed: 12/13/2022]
Abstract
Epithelial mesenchymal transition (EMT), as defined by loss of epithelial characteristics and gain of a mesenchymal phenotype, has been reported in vivo although the occurrence of events remains unclear. This study aims at exploration of EMT portraits of breast cancer (BC) with relevance to different molecular pathways, especially potential EMT triggers and BC molecular subtypes. Immunohistochemical (IHC) expression of markers/triggers of EMT was studied on a well-defined cohort of invasive non-lobular BC (n = 1,035), prepared as tissue microarrays. IHC panel of biomarkers included cadherins (cad; E-cad and N-cad), TGFβ1, PIK3CA, pAkt, and others. Reverse phase protein array (RPPA) was performed for quantitative analysis of proteins extracted from formalin fixed paraffin embedded tissues of a subset of cases from this cohort. Four combinatorial phenotypic groups representing cadherin switch were defined, including E-cad(+)/N-cad(-), E-cad(-)/N-cad(-), E-cad(+)/N-cad(+), and E-cad(-)/N-cad(+). Statistically significant association was noticed between these phenotypes and histological tumour grade, tumour type and size and NPI staging classes. The E-cad/N-cad switch occurred more frequently in the triple negative molecular class, both basal and non-basal, and in the HER2(+) subtype than in luminal BC. Significant outcome differences were observed between cadherin switch combinatorial groups regarding BCSS and DMFS (p < 0.001). Results of RPPA confirm those observed using IHC regarding differential expressions of EMT markers/triggers. EMT/cadherin switch programs in BC appear to occur in synergy with TGFβ1 and PIK3/Akt pathways activation. These data explain, at translational proteomic level, the molecular heterogeneity and in turn the varied clinical behaviour of BC molecular subtypes. RPPA is a promising high-throughput technique in monitoring subtle quantitative changes in protein expression in archival material.
Collapse
|
84
|
Pal HC, Sharma S, Strickland LR, Katiyar SK, Ballestas ME, Athar M, Elmets CA, Afaq F. Fisetin inhibits human melanoma cell invasion through promotion of mesenchymal to epithelial transition and by targeting MAPK and NFκB signaling pathways. PLoS One 2014; 9:e86338. [PMID: 24466036 PMCID: PMC3900533 DOI: 10.1371/journal.pone.0086338] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/06/2013] [Indexed: 11/19/2022] Open
Abstract
Malignant melanoma is responsible for approximately 75% of skin cancer-related deaths. BRAF plays an important role in regulating the mitogen-activated protein kinase (MAPK) signaling cascade in melanoma with activating mutations in the serine/threonine kinase BRAF occurring in 60-70% of malignant melanomas. The BRAF-MEK-ERK (MAPK) pathway is a key regulator of melanoma cell invasion. In addition, activation of NFκB via the MAPK pathway is regulated through MEK-induced activation of IKK. These pathways are potential targets for prevention and treatment of melanoma. In this study, we investigated the effect of fisetin, a phytochemical present in fruits and vegetables, on melanoma cell invasion and epithelial-mesenchymal transition, and delineated the underlying molecular mechanism. Treatment of multiple human malignant melanoma cell lines with fisetin (5-20 µM) resulted in inhibition of cell invasion. BRAF mutated melanoma cells were more sensitive to fisetin treatment, and this was associated with a decrease in the phosphorylation of MEK1/2 and ERK1/2. In addition, fisetin inhibited the activation of IKK leading to a reduction in the activation of the NFκB signaling pathway. Treatment of cells with an inhibitor of MEK1/2 (PD98059) or of NFκB (caffeic acid phenethyl ester) also reduced melanoma cell invasion. Furthermore, treatment of fisetin promoted mesenchymal to epithelial transition in melanoma cells, which was associated with a decrease in mesenchymal markers (N-cadherin, vimentin, snail and fibronectin) and an increase in epithelial markers (E-cadherin and desmoglein). Employing three dimensional skin equivalents consisting of A375 cells admixed with normal human keratinocytes embedded onto a collagen-constricted fibroblast matrix, we found that treatment of fisetin reduced the invasive potential of melanoma cells into the dermis and increased the expression of E-cadherin with a concomitant decrease in vimentin. These results indicate that fisetin inhibits melanoma cell invasion through promotion of mesenchymal to epithelial transition and by targeting MAPK and NFκB signaling pathways.
Collapse
Affiliation(s)
- Harish Chandra Pal
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Samriti Sharma
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Leah Ray Strickland
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Santosh K. Katiyar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Mary E. Ballestas
- Department of Pediatrics Infectious Disease, Children's of Alabama, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Craig A. Elmets
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Farrukh Afaq
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
85
|
Wang Q, Wu X, Wu T, Li GM, Shi Y. Clinical significance of RKIP mRNA expression in non-small cell lung cancer. Tumour Biol 2014; 35:4377-80. [PMID: 24420151 DOI: 10.1007/s13277-013-1575-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 12/17/2013] [Indexed: 12/31/2022] Open
Abstract
Raf-1 kinase inhibitor protein (RKIP) expression was associated with the onset, development, invasion, and metastasis of numerous tumor types including prostate cancer, melanoma, colorectal cancer, liver cancer, and breast cancer. However, RKIP mRNA expression and the clinical significance in non-small cell lung cancers (NSCLC) remain unresolved. Real-time PCR was performed to detect the expression of RKIP mRNA in 126 pairs of lung tumor tissues (TT) and surrounding normal tissues (sNT). Correlations between RKIP mRNA expression and clinicopathological features were evaluated by statistical analysis. In the 126 patients examined, RKIP mRNA expression was significantly lower in lung TT than the sNT (p < 0.05). Our results indicated that downregulation of RKIP mRNA expression was associated with a poorer N-stage (p = 0.019) and poorer pathological TNM stage (p = 0.015). However, no significant association was observed between the expression status of RKIP mRNA and clinicopathologic factors, such as gender, age, histological type, and the size of the tumor (p > 0.05). The level of RKIP mRNA expression was found to be significantly downregulated in NSCLC, and the lower mRNA levels correlated with poorer differentiation, advanced pathologic TNM stage in patients with NSCLC.
Collapse
Affiliation(s)
- Qin Wang
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Clinical School of Nanjing, Second Military Medical University, Nanjing, 210002, China
| | | | | | | | | |
Collapse
|
86
|
Yang D, Qi Z. [Expression and significance of Raf kinase inhibitory protein in lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 15:597-601. [PMID: 23075684 PMCID: PMC5999832 DOI: 10.3779/j.issn.1009-3419.2012.10.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
背景与目的 Raf激酶抑制蛋白(Raf kinase inhibitor protein, RKIP)属于磷脂酰乙醇胺结合蛋白家族的成员,RKIP参与ERK/MAPK、G蛋白偶联受体和NF-κΒ等信号传导过程,且RKIP的表达减弱或丢失与多种肿瘤的发生发展及侵润转移相关。本研究旨在探讨RKIP在非小细胞肺癌(non-small cell lung cancer, NSCLC)组织中的表达及其与NSCLC临床病理特征的相关性。 方法 应用RT-PCR、Western blot及免疫组化方法检测83例NSCLC及其癌旁组织标本中RKIP的表达,并结合临床病理学资料进行统计学分析,所有病例均经病理诊断确诊,均无其它部位原发肿瘤,术前无化疗、放疗和免疫治疗史。 结果 NSCLC中RKIP mRNA及蛋白的表达明显低于癌旁组织,差异有统计学意义(P < 0.05)。RKIP与肿瘤分化程度、TNM分期、有无淋巴结转移及生存期有关(P < 0.05),但与患者的性别、吸烟、年龄及肿瘤的大小无关(P > 0.05)。 结论 RKIP的低表达与NSCLC的发生及侵袭转移有关,可作为NSCLC预测及预后评估的指标。
Collapse
Affiliation(s)
- Dayun Yang
- Department of Internal Medicine, the Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | | |
Collapse
|
87
|
Bao Y, Ruan LJ, Mo JF. Low trichorhinophalangeal syndrome 1 gene transcript levels in basal-like breast cancer associate with mesenchymal-to-epithelial transition. ACTA ACUST UNITED AC 2013; 28:129-34. [PMID: 24074613 DOI: 10.1016/s1001-9294(13)60037-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To investigate trichorhinophalangeal syndrome 1 gene (TRPS-1) expression patterns in different subtypes of breast cancer and its correlations with other genes and survival using microarray data sets. METHODS The transcripts of TRPS-1 and its role in survival in breast cancer were analyzed using published microarray data sets#x02014;Netherlands Cancer Institute (NKI) cohort and Wang cohort. RESULTS TRPS-1 expression was lower in basal-like breast cancer. The mRNA levels of TRPS-1 negatively correlated with Slug (Pearson correlation coefficient=-0.1366, P=0.0189 in NKI data set and Pearson correlation coefficient=-0.1571, P=0.0078 in Wang data set), FOXC1 (Pearson correlation coefficient=-0.1211, P=0.0376 in NKI data set and Pearson correlation coefficient=-0.1709, P=0.0037 in Wang data set), and CXCL1 (Pearson correlation coefficient=-0.1197, P=0.0399 in NKI data set and Pearson correlation coefficient=-0.3436, P<0.0001 in Wang data set), but positively correlated with BRCA1 (Pearson correlation coefficient=0.1728, P=0.0029 in NKI data set and Pearson correlation coefficient=0.1805, P=0.0022 in Wang data set). Low TRPS-1 expression associated with poor overall survival (hazard ratio 1.79, 95% CI of ratio 0.9894 to 3.238, P=0.054) and relapse-free survival (hazard ratio 1.913, 95% CI of ratio 1.159 to 3.156, P<0.05). The low TRPS-1 mRNA levels predicted poor outcome in breast cancer patients by the 70-gene signature. CONCLUSION The strong expression of TRPS-1 may serve as a good prognostic marker in breast cancer.
Collapse
Affiliation(s)
- Yi Bao
- Key Laboratory, Jiaxing Second Hospital, Jiaxing University, Jiaxing, Zhejiang 314000, China
| | | | | |
Collapse
|
88
|
Regad T. Molecular and cellular pathogenesis of melanoma initiation and progression. Cell Mol Life Sci 2013; 70:4055-65. [PMID: 23532409 PMCID: PMC11113476 DOI: 10.1007/s00018-013-1324-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 03/07/2013] [Accepted: 03/11/2013] [Indexed: 12/12/2022]
Abstract
Melanoma is a malignant tumor of melanocytes that can spread to other organs of the body, resulting in severe and/or lethal malignancies. Melanocytes are pigment-producing cells found in the deep layer of the epidermis and are originated from melanocytes stem cells through a cellular process called melanogenesis. Several genes and epigenetic and micro-environmental factors are involved in this process via the regulation and maintenance of the balance between melanocytes stem cells proliferation and their differentiation into melanocytes. Dysregulation of this balance through gain or loss of function of key genes implicated in the control and regulation of cell cycle progression and/or differentiation results in melanoma initiation and progression. This review aims to provide a comprehensive overview about the origin of melanocytes, the oncogenic events involved in melanocytes stem cells transformation, and the mechanisms implicated in the perpetuation of melanoma malignant phenotype.
Collapse
Affiliation(s)
- Tarik Regad
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK,
| |
Collapse
|
89
|
Ronca R, Di Salle E, Giacomini A, Leali D, Alessi P, Coltrini D, Ravelli C, Matarazzo S, Ribatti D, Vermi W, Presta M. Long Pentraxin-3 Inhibits Epithelial–Mesenchymal Transition in Melanoma Cells. Mol Cancer Ther 2013; 12:2760-71. [DOI: 10.1158/1535-7163.mct-13-0487] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
90
|
Cardile V, Malaponte G, Loreto C, Libra M, Caggia S, Trovato FM, Musumeci G. Raf kinase inhibitor protein (RKIP) and phospho-RKIP expression in melanomas. Acta Histochem 2013; 115:795-802. [PMID: 23601922 DOI: 10.1016/j.acthis.2013.03.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 03/10/2013] [Indexed: 11/21/2022]
Abstract
Melanoma, a cancer notorious for its high potential to metastasize, arises from melanocytes, cells dedicated to melanin production and located in the basal layer of the epidermis. Raf-1 kinase inhibitor protein (RKIP) is an inhibitory molecule that down-regulates the effects of the Ras/Raf/MEK/ERK signaling pathway. The aim of this study was to examine the expression of RKIP and pRKIP in melanomas at different stages. We evaluated the RKIP and pRKIP protein by immunohistochemistry in control skin, pigmented nevi and melanomas, and through Western blotting in human normal melanocytes and in four different melanoma-derived cell lines (WM35, A375, M14, and A2058). Our results demonstrated a correlation between the expression of RKIP and pRKIP, and metastatic ability in melanoma cells. This raises the possibility to analyze both RKIP and pRKIP in all melanomas. Down-regulation of both RKIP and pRKIP expression could represent a useful marker of metastatic melanoma. On the contrary for non-metastatic melanoma, especially in Clark I and II, low RKIP and high pRKIP expression could be indicative. In conclusion, the observed negative correlation of the RKIP and pRKIP expression in metastatic melanomas indicates that expression of these proteins may become a prognostic marker for the progression of human cutaneous melanoma. We propose that the investigation of both RKIP and pRKIP may provide a useful tool indicative for metastatic or non-metastatic melanoma in different Clark's level melanomas. Further studies are required to verify the molecular background of the observed RKIP and pRKIP variations.
Collapse
Affiliation(s)
- Venera Cardile
- Department of Bio-medical Sciences, Section of Physiology, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
91
|
Hirano T, Satow R, Kato A, Tamura M, Murayama Y, Saya H, Kojima H, Nagano T, Okabe T, Fukami K. Identification of novel small compounds that restore E-cadherin expression and inhibit tumor cell motility and invasiveness. Biochem Pharmacol 2013; 86:1419-29. [PMID: 24035834 DOI: 10.1016/j.bcp.2013.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/05/2013] [Accepted: 09/05/2013] [Indexed: 01/27/2023]
Abstract
Tumor dissemination and invasive behavior are associated with a majority of cancer-related mortality cases. Loss of E-cadherin, which is caused by several tumor-promoting factors, is associated with metastasis and poor prognosis in many neoplasms. In this study, we aimed to identify small molecule compounds that restore the expression of E-cadherin, because these molecules are most likely to suppress tumor malignancy by restoring E-cadherin function and/or by inhibiting signals that suppress E-cadherin expression. Here, we developed a fluorescence screen system based on E-cadherin expression. A pilot drug library screen revealed that methotrexate (MTX) strongly induces E-cadherin expression in a colorectal cancer cell line, SW620. From the screen for 9600 compounds, we identified 9 hit compounds, which restored the expression of E-cadherin in SW620 and/or a melanoma cell line, SK-MEL-28. We confirmed that MTX and the other identified compounds transcriptionally promote E-cadherin expression. Among these, 2 compounds suppressed migration/invasion capacity in colorectal cancer cells and 3 in melanoma cells. A compound reduced SW620 migration and invasion with subtle effects on cell viability in SW620, SK-MEL-28, and a non-tumor cell line, HaCaT, with decrease in AKT and ERK1/2 protein levels. One of the other compounds reduced SK-MEL-28 cell migration and invasion and affected the viability only of SW620 and SK-MEL-28 cells but not HaCaT cells. These results suggest that these compounds would be attractive lead molecules as anti-metastasis agents.
Collapse
Affiliation(s)
- Tamaki Hirano
- Laboratory of Genome and Biosignal, Tokyo University of Pharmacy and Life Sciences, Hachioji-shi, Tokyo 192-0392, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Garnier D, Magnus N, Meehan B, Kislinger T, Rak J. Qualitative changes in the proteome of extracellular vesicles accompanying cancer cell transition to mesenchymal state. Exp Cell Res 2013; 319:2747-57. [PMID: 23954818 DOI: 10.1016/j.yexcr.2013.08.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 07/22/2013] [Accepted: 08/02/2013] [Indexed: 12/14/2022]
Abstract
Transitions of the cancer cell phenotype between epithelial and mesenchymal states (EMT) are likely to alter the patterns of intercellular communication. In this regard we have previously documented that EMT-like changes trigger quantitative rearrangements in exosomal vesicle emission in A431 cancer cells driven by oncogenic epidermal growth factor receptor (EGFR). Here we report that extracellular vesicles (EVs) produced by these cancer cells in their epithelial and mesenchymal states exhibit profound qualitative differences in their proteome. Thus, induction of the EMT-like state through blockade of E-cadherin and EGFR stimulation provoked a mesenchymal shift in cellular morphology and enrichment in the CD44-high/CD24-low immunophenotype, often linked to cellular stemness. This change also resulted in reprogramming of the EV-related proteome (distinct from that of corresponding cells), which contained 30 unique protein signals, and revealed enrichment in pathways related to cellular growth, cell-to-cell signaling, and cell movement. Some of the most prominent EV-related proteins were validated, including integrin α2 and tetraspanin CD9. We propose that changes in cellular differentiation status translate into unique qualitative rearrangements in the cargo of EVs, a process that may have implications for intercellular communication and could serve as source of new biomarkers to detect EMT-like processes in cancer.
Collapse
Affiliation(s)
- Delphine Garnier
- Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
93
|
The metastasis-promoting roles of tumor-associated immune cells. J Mol Med (Berl) 2013; 91:411-29. [PMID: 23515621 DOI: 10.1007/s00109-013-1021-5] [Citation(s) in RCA: 277] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 03/06/2013] [Accepted: 03/07/2013] [Indexed: 12/12/2022]
Abstract
Tumor metastasis is driven not only by the accumulation of intrinsic alterations in malignant cells, but also by the interactions of cancer cells with various stromal cell components of the tumor microenvironment. In particular, inflammation and infiltration of the tumor tissue by host immune cells, such as tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, have been shown to support tumor growth in addition to invasion and metastasis. Each step of tumor development, from initiation through metastatic spread, is promoted by communication between tumor and immune cells via the secretion of cytokines, growth factors, and proteases that remodel the tumor microenvironment. Invasion and metastasis require neovascularization, breakdown of the basement membrane, and remodeling of the extracellular matrix for tumor cell invasion and extravasation into the blood and lymphatic vessels. The subsequent dissemination of tumor cells to distant organ sites necessitates a treacherous journey through the vasculature, which is fostered by close association with platelets and macrophages. Additionally, the establishment of the pre-metastatic niche and specific metastasis organ tropism is fostered by neutrophils and bone marrow-derived hematopoietic immune progenitor cells and other inflammatory cytokines derived from tumor and immune cells, which alter the local environment of the tissue to promote adhesion of circulating tumor cells. This review focuses on the interactions between tumor cells and immune cells recruited to the tumor microenvironment and examines the factors allowing these cells to promote each stage of metastasis.
Collapse
|
94
|
Swoboda RK, Herlyn M. There is a world beyond protein mutations: the role of non-coding RNAs in melanomagenesis. Exp Dermatol 2013; 22:303-6. [PMID: 23489578 DOI: 10.1111/exd.12117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2013] [Indexed: 12/17/2022]
Abstract
Until recently, the general perception has been that mutations in protein-coding genes are responsible for tumorigenesis. With the discovery of (V600E)BRAF in about 50% of cutaneous melanomas, there was an increased effort to find additional mutations. However, mutations characterized in melanoma to date cannot account for the development of all melanomas. With the discovery of microRNAs as important players in melanomagenesis, protein mutations are no longer considered the sole drivers of tumors. Recent research findings have expanded the view for tumor initiation and progression to additional non-coding RNAs. The data suggest that tumorigenesis is likely an interplay between mutated proteins and deregulation of non-coding RNAs in the cell with an additional role of the tumor environment. With the exception of microRNAs, our knowledge of the role of non-coding RNAs in melanoma is in its infancy. Using few examples, we will summarize some of the roles of non-coding RNAs in tumorigenesis. Thus, there is a whole world beyond protein-coding sequences and microRNAs, which can cause melanoma.
Collapse
|
95
|
Baquero P, Sánchez-Hernández I, Jiménez-Mora E, Orgaz JL, Jiménez B, Chiloeches A. (V600E)BRAF promotes invasiveness of thyroid cancer cells by decreasing E-cadherin expression through a Snail-dependent mechanism. Cancer Lett 2013; 335:232-41. [PMID: 23435375 DOI: 10.1016/j.canlet.2013.02.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 02/12/2013] [Indexed: 01/07/2023]
Abstract
BRAF is a main oncogene in human thyroid cancer. Here, we show that BRAF depletion by siRNA or inhibition of its activity by treatment with BRAF inhibitor PLX4720 decreases migration and invasion in thyroid cancer cells expressing oncogenic (V600E)BRAF through a MEK/ERK-dependent mechanism, since treatment with the MEK inhibitor U0126 exerts the same effect. Moreover, over-expression of (V600E)BRAF increases migration and invasion of wild-type BRAF thyroid cells. Using the same strategies, we demonstrate that these effects are mediated by upregulation of the transcriptional repressor Snail with a concomitant decrease of its target E-cadherin, both hallmarks of EMT. These results reveal a novel (V600E)BRAF-induced mechanism in thyroid tumours progression and provides a rationale for using the PLX4720 inhibitor to target (V600E)BRAF signalling to effectively control progression of thyroid cancer.
Collapse
Affiliation(s)
- Pablo Baquero
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
96
|
Dynoodt P, Speeckaert R, De Wever O, Chevolet I, Brochez L, Lambert J, Van Gele M. miR-145 overexpression suppresses the migration and invasion of metastatic melanoma cells. Int J Oncol 2013; 42:1443-51. [PMID: 23404256 DOI: 10.3892/ijo.2013.1823] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 12/07/2012] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are post-transcriptional modulators of gene expression which play important roles in tumorigenesis and cancer metastasis. Since they are often highly deregulated in various types of cancer, miRNAs may be effective treatment targets. miRNA profiling studies of melanoma have led to the identification of several tumor suppressor miRNAs. One of these include miR-145, although functional data proving its specific function are limited. Therefore, in this study, we examined the expression levels of miR-145 in three melanoma cell lines (BLM, FM3P and WM793). Additional gain-of-function experiments revealed that miR-145 exerts an anti-proliferative effect in the primary, non-invasive melanoma cell line, WM793, whereas cell migration and the invasive potential of metastatic melanoma cells was suppressed following transfection with miR-145 mimics. In order to investigate the mechanisms by which miR-145 exerts its invasion suppressor function, we examined the expression level of target genes [fascin homolog 1 (FSCN1), myosin‑Va (MYO5A and SOX9] and that of an indirect target (RAB27A) following the overexpression of miR-145. The results showed that SOX9, MYO5A and RAB27A were not involved in the biological effects caused by miR-145 mimics. Surprisingly, we discovered that miR-145 in melanoma, in contrast to many other tumor types, does not necessarily act via the target, FSCN1, since the downregulation of FSCN1 did not inhibit cell proliferation or migration but, on the contrary, increased cell invasion in two out of the three melanoma cell lines examined. Our in vitro data is in accordance with previously reported in vivo data describing the low expression of FSCN1 in malignant melanomas when compared to dysplastic nevi, suggesting that the expression of FSCN1 decreases as the formation and progression stage of melanoma advances. In conclusion, our data provide evidence that miR-145 is an invasion suppressor in metastatic melanoma cells. Despite the fact that it remains unclear which genes or pathways are regulated by miR-145 in melanoma, miR-145 may serve as a useful therapeutic agent in melanoma when re-expressed in situ.
Collapse
Affiliation(s)
- Peter Dynoodt
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium
| | | | | | | | | | | | | |
Collapse
|
97
|
Boyd SC, Mijatov B, Pupo GM, Tran SL, Gowrishankar K, Shaw HM, Goding CR, Scolyer RA, Mann GJ, Kefford RF, Rizos H, Becker TM. Oncogenic B-RAF(V600E) signaling induces the T-Box3 transcriptional repressor to repress E-cadherin and enhance melanoma cell invasion. J Invest Dermatol 2012. [PMID: 23190890 PMCID: PMC3788590 DOI: 10.1038/jid.2012.421] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Approximately 50% of melanomas require oncogenic B-RAFV600E signaling for proliferation, survival and metastasis, and the use of highly selective B-RAF inhibitors has yielded remarkable, albeit short term, clinical responses. Re-activation of signaling downstream of B-RAF is frequently associated with acquired resistance to B-RAF inhibitors, and the identification of B-RAF targets may therefore provide new strategies for managing melanoma. In this report, we applied whole genome expression analyses to reveal that oncogenic B-RAFV600E regulates genes associated with epithelial-mesenchymal transition in normal cutaneous human melanocytes. Most prominent was the B-RAF-mediated transcriptional repression of E-cadherin, a keratinocyte-melanoma adhesion molecule whose loss is intimately associated with melanoma invasion and metastasis. Here we identify a link between oncogenic B-RAF, the transcriptional repressor Tbx3 and E-cadherin. We show that B-RAFV600E induces the expression of Tbx3, which potently represses E-cadherin expression in melanocytes and melanoma cells. Tbx3 expression is normally restricted to developmental embryonic tissues, promoting cell motility but is also aberrantly increased in various cancers and has been linked to tumor cell invasion and metastasis. We propose that this B-RAF/Tbx3/E-cadherin pathway plays a critical role in promoting the metastasis of B-RAF mutant melanomas.
Collapse
Affiliation(s)
- Suzanah C Boyd
- University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Das SK, Bhutia SK, Sokhi UK, Azab B, Su ZZ, Boukerche H, Anwar T, Moen EL, Chatterjee D, Pellecchia M, Sarkar D, Fisher PB. Raf kinase inhibitor RKIP inhibits MDA-9/syntenin-mediated metastasis in melanoma. Cancer Res 2012; 72:6217-26. [PMID: 23066033 DOI: 10.1158/0008-5472.can-12-0402] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Melanoma differentiation associated gene-9 (MDA-9), also known as syntenin, functions as a positive regulator of melanoma progression and metastasis. In contrast, the Raf kinase inhibitor, RKIP, a negative modulator of RAF-stimulated MEKK activation, is strongly downregulated in metastatic melanoma cells. In this study, we explored a hypothesized inverse relationship between MDA-9 and RKIP in melanoma. Tumor array and cell line analyses confirmed an inverse relationship between expression of MDA-9 and RKIP during melanoma progression. We found that MDA-9 transcriptionally downregulated RKIP in support of a suggested cross-talk between these two proteins. Furthermore, MDA-9 and RKIP physically interacted in a manner that correlated with a suppression of FAK and c-Src phosphorylation, crucial steps necessary for MDA-9 to promote FAK/c-Src complex formation and initiate signaling cascades that drive the MDA-9-mediated metastatic phenotype. Finally, ectopic RKIP expression in melanoma cells overrode MDA-9-mediated signaling, inhibiting cell invasion, anchorage-independent growth, and in vivo dissemination of tumor cells. Taken together, these findings establish RKIP as an inhibitor of MDA-9-dependent melanoma metastasis, with potential implications for targeting this process therapeutically.
Collapse
Affiliation(s)
- Swadesh K Das
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Ruan JS, Liu YP, Zhang L, Yan LG, Fan FT, Shen CS, Wang AY, Zheng SZ, Wang SM, Lu Y. Luteolin reduces the invasive potential of malignant melanoma cells by targeting β3 integrin and the epithelial-mesenchymal transition. Acta Pharmacol Sin 2012; 33:1325-31. [PMID: 22983392 DOI: 10.1038/aps.2012.93] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIM To investigate whether luteolin, a highly prevalent flavonoid, reverses the effects of epithelial-mesenchymal transition (EMT) in vitro and in vivo and to determine the mechanisms underlying this reversal. METHODS Murine malignant melanoma B16F10 cells were exposed to 1% O(2) for 24 h. Cellular mobility and adhesion were assessed using Boyden chamber transwell assay and cell adhesion assay, respectively. EMT-related proteins, such as E-cadherin and N-cadherin, were examined using Western blotting. Female C57BL/6 mice (6 to 8 weeks old) were injected with B16F10 cells (1×10(6) cells in 0.2 mL per mouse) via the lateral tail vein. The mice were treated with luteolin (10 or 20 mg/kg, ip) daily for 23 d. On the 23rd day after tumor injection, the mice were sacrificed, and the lungs were collected, and metastatic foci in the lung surfaces were photographed. Tissue sections were analyzed with immunohistochemistry and HE staining. RESULTS Hypoxia changed the morphology of B16F10 cells in vitro from the cobblestone-like to mesenchymal-like strips, which was accompanied by increased cellular adhesion and invasion. Luteolin (5-50 μmol/L) suppressed the hypoxia-induced changes in the cells in a dose-dependent manner. Hypoxia significantly decreased the expression of E-cadherin while increased the expression of N-cadherin in the cells (indicating the occurrence of EMT-like transformation), which was reversed by luteolin (5 μmol/L). In B16F10 cells, luteolin up-regulated E-cadherin at least partly via inhibiting the β3 integrin/FAK signal pathway. In experimental metastasis model mice, treatment with luteolin (10 or 20 mg/kg) reduced metastatic colonization in the lungs by 50%. Furthermore, the treatment increased the expression of E-cadherin while reduced the expression of vimentin and β3 integrin in the tumor tissues. CONCLUSION Luteolin inhibits the hypoxia-induced EMT in malignant melanoma cells both in vitro and in vivo via the regulation of β3 integrin, suggesting that luteolin may be applied as a potential anticancer chemopreventative and chemotherapeutic agent.
Collapse
|
100
|
Xu X, Zhou Y, Xie C, Wei SM, Gan H, He S, Wang F, Xu L, Lu J, Dai W, He L, Chen P, Wang X, Guo C. Genome-wide screening reveals an EMT molecular network mediated by Sonic hedgehog-Gli1 signaling in pancreatic cancer cells. PLoS One 2012; 7:e43119. [PMID: 22900095 PMCID: PMC3416762 DOI: 10.1371/journal.pone.0043119] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 07/17/2012] [Indexed: 12/21/2022] Open
Abstract
AIMS The role of sonic hedgehog (SHH) in epithelial mesenchymal transition (EMT) of pancreatic cancer (PC) is known, however, its mechanism is unclear. Because SHH promotes tumor development predominantly through Gli1, we sought to understand its mechanism by identifying Gli1 targets in pancreatic cancer cells. METHODS First, we investigated invasion, migration, and EMT in PC cells transfected with lentiviral Gli1 interference vectors or SHH over-expression vectors in vitro and in vivo. Next, we determined the target gene profiles of Gli1 in PC cells using cDNA microarray assays. Finally, the primary regulatory networks downstream of SHH-Gli1 signaling in PC cells were studied through functional analyses of these targets. RESULTS Our results indicate there is decreased E-cadherin expression upon increased expression of SHH/Gli1. Migration of PC cells increased significantly in a dose-dependent manner within 24 hours of Gli1 expression (P<0.05). The ratio of liver metastasis and intrasplenic miniature metastasis increased markedly upon activation of SHH-Gli1 signals in nude mice. Using cDNA microarray, we identified 278 upregulated and 59 downregulated genes upon Gli1 expression in AsPC-1 cells. The data indicate that SHH-Gli1 signals promote EMT by mediating a complex signaling network including TGFβ, Ras, Wnt, growth factors, PI3K/AKT, integrins, transmembrane 4 superfamily (TM4SF), and S100A4. CONCLUSION Our results suggest that targeting the molecular connections established between SHH-Gli1 signaling and EMT could provide effective therapies for PC.
Collapse
Affiliation(s)
- Xuanfu Xu
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Yingqun Zhou
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Chuangao Xie
- Department of Gastroenterology, The Second Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Shu-mei Wei
- Department of Gastroenterology, The Second Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Huizhong Gan
- Department of Gastroenterology, The First People's Hospital of Hefei, Anhui Medical University, Heifei, Anhui Province, China
| | - Shengli He
- Department of Integrative Oncology, Minhang Branch of Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fan Wang
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Ling Xu
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Jie Lu
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Weiqi Dai
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Lei He
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Ping Chen
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Xingpeng Wang
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| | - Chuanyong Guo
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, China
| |
Collapse
|