1
|
Vieira Cardoso II, Nunes Rosa M, Antunes Moreno D, Barbosa Tufi LM, Pereira Ramos L, Bourdeth Pereira LA, Silva L, Soares Galvão JM, Tosi IC, Van Helvoort Lengert A, Cavalcanti Da Cruz M, Teixeira SA, Reis RM, Lopes LF, Tomazini Pinto M. Cisplatin‑resistant germ cell tumor models: An exploration of the epithelial‑mesenchymal transition regulator SLUG. Mol Med Rep 2024; 30:228. [PMID: 39392037 PMCID: PMC11484538 DOI: 10.3892/mmr.2024.13352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/27/2024] [Indexed: 10/12/2024] Open
Abstract
Germ cell tumors (GCTs) constitute diverse neoplasms arising in the gonads or extragonadal locations. Testicular GCTs (TGCTs) are the predominant solid tumors in adolescents and young men. Despite cisplatin serving as the primary therapeutic intervention for TGCTs, 10‑20% of patients with advanced disease demonstrate resistance to cisplatin‑based chemotherapy, and epithelial‑mesenchymal transition (EMT) is a potential contributor to this resistance. EMT is regulated by various factors, including the snail family transcriptional repressor 2 (SLUG) transcriptional factor, and, to the best of our knowledge, remains unexplored within TGCTs. Therefore, the present study investigated the EMT transcription factor SLUG in TGCTs. In silico analyses were performed to investigate the expression of EMT markers in TGCTs. In addition, a cisplatin‑resistant model for TGCTs was developed using the NTERA‑2 cell line, and a mouse model was also established. Subsequently, EMT was assessed both in vitro and in vivo within the cisplatin‑resistant models using quantitative PCR and western blot analyses. The results of the in silico analysis showed that the different histologies exhibited distinct expression profiles for EMT markers. Seminomas exhibited a lower expression of EMT markers, whereas embryonal carcinomas and mixed GCT demonstrated high expression. Notably, patients with lower SLUG expression had longer median progression‑free survival (46.4 months vs. 28.0 months, P=0.022). In the in vitro analysis, EMT‑associated genes [fibronectin; vimentin (VIM); actin, α2, smooth muscle; collagen type I α1; transforming growth factor‑β1; and SLUG] were upregulated in the cisplatin‑resistant NTERA‑2 (NTERA‑2R) cell line after 72 h of cisplatin treatment. Consistent with this finding, the NTERA‑2R mouse model demonstrated a significant upregulation in the expression levels of VIM and SLUG. In conclusion, the present findings suggested that SLUG may serve a crucial role in connecting EMT with the development of cisplatin resistance, and targeting SLUG may be a putative therapeutic strategy to mitigate cisplatin resistance.
Collapse
Affiliation(s)
| | - Marcela Nunes Rosa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | - Daniel Antunes Moreno
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | | | - Lorrayne Pereira Ramos
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | | | - Lenilson Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | | | - Isabela Cristiane Tosi
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | | | | | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
- Life and Health Sciences Research Institute Medical School, University of Minho, 710057 Braga, Portugal
| | - Luiz Fernando Lopes
- Barretos Children's Cancer Hospital, Hospital de Amor, Barretos, São Paulo 14784400, Brazil
| | - Mariana Tomazini Pinto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
- Barretos Children's Cancer Hospital, Hospital de Amor, Barretos, São Paulo 14784400, Brazil
| |
Collapse
|
2
|
Liu L, Deng P, Liu S, Hong JH, Xiao R, Guan P, Wang Y, Wang P, Gao J, Chen J, Sun Y, Chen J, Mai HQ, Tan J. Enhancer remodeling activates NOTCH3 signaling to confer chemoresistance in advanced nasopharyngeal carcinoma. Cell Death Dis 2023; 14:513. [PMID: 37563118 PMCID: PMC10415329 DOI: 10.1038/s41419-023-06028-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
Acquired resistance to chemotherapy is one of the major causes of mortality in advanced nasopharyngeal carcinoma (NPC). However, effective strategies are limited and the underlying molecular mechanisms remain elusive. In this study, through transcriptomic profiling analysis of 23 tumor tissues, we found that NOTCH3 was aberrantly highly expressed in chemoresistance NPC patients, with NOTCH3 overexpression being positively associated with poor clinical outcome. Mechanistically, using an established NPC cellular model, we demonstrated that enhancer remodeling driven aberrant hyperactivation of NOTCH3 in chemoresistance NPC. We further showed that NOTCH3 upregulates SLUG to induce chemo-resistance of NPC cells and higher expression of SLUG have poorer prognosis. Genetic or pharmacological perturbation of NOTCH3 conferred chemosensitivity of NPC in vitro and overexpression of NOTCH3 enhanced chemoresistance of NPC in vivo. Together, these data indicated that genome-wide enhancer reprogramming activates NOTCH3 to confer chemoresistance of NPC, suggesting that targeting NOTCH3 may provide a potential therapeutic strategy to effectively treat advanced chemoresistant NPC.
Collapse
Affiliation(s)
- Lizhen Liu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Peng Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sailan Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Han Hong
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Rong Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Peiyong Guan
- Genome Institute of Singapore, A*STAR, Singapore, Republic of Singapore
| | - Yali Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Peili Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiuping Gao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jinghong Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yichen Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianfeng Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hai-Qiang Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Tan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
3
|
Interplay between Partial EMT and Cisplatin Resistance as the Drivers for Recurrence in HNSCC. Biomedicines 2022; 10:biomedicines10102482. [PMID: 36289744 PMCID: PMC9598677 DOI: 10.3390/biomedicines10102482] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/20/2022] Open
Abstract
This study aims to investigate the role of partial epithelial to mesenchymal transition (pEMT)-related proteins in modulating Cisplatin resistance in head and neck squamous cell carcinoma (HNSCC). SCC-25 cells were pre-treated with TGF-beta1 followed by transient Krüppel-like Factor 4 (KLF4)-overexpression and Cisplatin treatment. Cell growth, cell morphological changes and cell migration were assessed using Juli BR live cell video-microscopy. In addition, Ki-67 and Slug immunostaining and follow-up image cytometric analysis of primary and recurrent HNSCC tumors were performed to evaluate the proliferation index (PI) and the EMT-like phenotype. We observed that proliferating and Slug-positive tumor cells expand after therapy in HNSCC. Subsequently, protein analysis revealed the stabilization of Slug, upregulation of Vimentin and phospho-p38 (p-p38) in Cisplatin-resistant SCC-25 cells. Moreover, KLF4-overexpression contributed to Cisplatin sensitivity by reduction of Slug at the protein level. This work strongly suggests that an pEMT-like pathway is activated in recurrent and Cisplatin-resistant HNSCC. Finally, stable KLF4-overexpression might sensitize HNSCC tumor cells for Cisplatin treatment.
Collapse
|
4
|
Griso AB, Acero-Riaguas L, Castelo B, Cebrián-Carretero JL, Sastre-Perona A. Mechanisms of Cisplatin Resistance in HPV Negative Head and Neck Squamous Cell Carcinomas. Cells 2022; 11:561. [PMID: 35159370 PMCID: PMC8834318 DOI: 10.3390/cells11030561] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 11/16/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are the eighth most common cancers worldwide. While promising new therapies are emerging, cisplatin-based chemotherapy remains the gold standard for advanced HNSCCs, although most of the patients relapse due to the development of resistance. This review aims to condense the different mechanisms involved in the development of cisplatin resistance in HNSCCs and highlight future perspectives intended to overcome its related complications. Classical resistance mechanisms include drug import and export, DNA repair and oxidative stress control. Emerging research identified the prevalence of these mechanisms in populations of cancer stem cells (CSC), which are the cells mainly contributing to cisplatin resistance. The use of old and new CSC markers has enabled the identification of the characteristics within HNSCC CSCs predisposing them to treatment resistance, such as cell quiescence, increased self-renewal capacity, low reactive oxygen species levels or the acquisition of epithelial to mesenchymal transcriptional programs. In the present review, we will discuss how cell intrinsic and extrinsic cues alter the phenotype of CSCs and how they influence resistance to cisplatin treatment. In addition, we will assess how the stromal composition and the tumor microenvironment affect drug resistance and the acquisition of CSCs' characteristics through a complex interplay between extracellular matrix content as well as immune and non-immune cell characteristics. Finally, we will describe how alterations in epigenetic modifiers or other signaling pathways can alter tumor behavior and cell plasticity to induce chemotherapy resistance. The data generated in recent years open up a wide range of promising strategies to optimize cisplatin therapy, with the potential to personalize HNSCC patient treatment strategies.
Collapse
Affiliation(s)
- Ana Belén Griso
- Laboratory of Experimental Therapies and Biomarkers in Cancer, IdiPAZ, 28046 Madrid, Spain; (A.B.G.); (L.A.-R.)
| | - Lucía Acero-Riaguas
- Laboratory of Experimental Therapies and Biomarkers in Cancer, IdiPAZ, 28046 Madrid, Spain; (A.B.G.); (L.A.-R.)
| | - Beatriz Castelo
- Medical Oncology Department, University Hospital La Paz, 28046 Madrid, Spain;
| | | | - Ana Sastre-Perona
- Laboratory of Experimental Therapies and Biomarkers in Cancer, IdiPAZ, 28046 Madrid, Spain; (A.B.G.); (L.A.-R.)
| |
Collapse
|
5
|
Shen J, Meng Y, Wang K, Gao M, Du J, Wang J, Li Z, Zuo D, Wu Y. EML4-ALK G1202R mutation induces EMT and confers resistance to ceritinib in NSCLC cells via activation of STAT3/Slug signaling. Cell Signal 2022; 92:110264. [DOI: 10.1016/j.cellsig.2022.110264] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/30/2022]
|
6
|
Pechlivanis M, Campbell BK, Hovens CM, Corcoran NM. Biomarkers of Response to Neoadjuvant Androgen Deprivation in Localised Prostate Cancer. Cancers (Basel) 2021; 14:cancers14010166. [PMID: 35008330 PMCID: PMC8750084 DOI: 10.3390/cancers14010166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Prostate cancer is the second leading cause of cancer deaths in men. Attempts to improve patient outcomes include trials of neoadjuvant androgen deprivation therapy for patients with high-risk disease. Neoadjuvant treatment refers to androgen deprivation therapy that is administered prior to surgery (or radiation therapy). Patients typically respond well to this treatment regimen, showing a decrease in tumour size, but a significant proportion of patients eventually relapse and progress to metastatic disease. The mechanisms driving this resistance to neoadjuvant treatment are currently unknown. This review explores theories of resistance broadly, and their possible applications in the prostate cancer setting. Additionally, this review draws comparisons between breakthrough resistance and neoadjuvant resistance, and lastly investigates the current biomarkers for treatment sensitivity. Abstract Prostate cancer (PCa) is a hormone driven cancer, characterised by defects in androgen receptor signalling which drive the disease process. As such, androgen targeted therapies have been the mainstay for PCa treatment for over 70 years. High-risk PCa presents unique therapeutic challenges, namely in minimising the primary tumour, and eliminating any undetected micro metastases. Trials of neoadjuvant androgen deprivation therapy aim to address these challenges. Patients typically respond well to neoadjuvant treatment, showing regression of the primary tumour and negative surgical margins at the time of resection, however the majority of patients relapse and progress to metastatic disease. The mechanisms affording this resistance are largely unknown. This commentary attempts to explore theories of resistance more broadly, namely, clonal evolution, cancer stem cells, cell persistence, and drug tolerance. Moreover, it aims to explore the application of these theories in the PCa setting. This commentary also highlights the distinction between castration resistant PCa, and neoadjuvant resistant disease, and identifies the markers and characteristics of neoadjuvant resistant disease presented by current literature.
Collapse
Affiliation(s)
- Maree Pechlivanis
- Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia; (B.K.C.); (C.M.H.); (N.M.C.)
- Correspondence: ; Tel.: +61-3-9342-7294; Fax: +61-3-9342-8928
| | - Bethany K. Campbell
- Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia; (B.K.C.); (C.M.H.); (N.M.C.)
| | - Christopher M. Hovens
- Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia; (B.K.C.); (C.M.H.); (N.M.C.)
| | - Niall M. Corcoran
- Department of Surgery, University of Melbourne, Parkville, VIC 3050, Australia; (B.K.C.); (C.M.H.); (N.M.C.)
- Department of Urology, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
- Department of Urology, Western Health, Footscray, VIC 3011, Australia
- Victorian Comprehensive Cancer Centre, Melbourne, VIC 3000, Australia
| |
Collapse
|
7
|
Cmero M, Kurganovs NJ, Stuchbery R, McCoy P, Grima C, Ngyuen A, Chow K, Mangiola S, Macintyre G, Howard N, Kerger M, Dundee P, Ruljancich P, Clarke D, Grummet J, Peters JS, Costello AJ, Norden S, Ryan A, Parente P, Hovens CM, Corcoran NM. Loss of SNAI2 in Prostate Cancer Correlates With Clinical Response to Androgen Deprivation Therapy. JCO Precis Oncol 2021; 5:PO.20.00337. [PMID: 34322653 PMCID: PMC8238292 DOI: 10.1200/po.20.00337] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 03/29/2021] [Accepted: 04/22/2021] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Androgen receptor (AR) signaling is important in prostate cancer progression, and therapies that target this pathway have been the mainstay of treatment for advanced disease for over 70 years. Tumors eventually progress despite castration through a number of well-characterized mechanisms; however, little is known about what determines the magnitude of response to short-term pathway inhibition. METHODS We evaluated a novel combination of AR-targeting therapies (degarelix, abiraterone, and bicalutamide) and noted that the objective patient response to therapy was highly variable. To investigate what was driving treatment resistance in poorly responding patients, as a secondary outcome we comprehensively characterized pre- and post-treatment samples using both whole-genome and RNA sequencing. RESULTS We find that resistance following short-term treatment differs molecularly from typical progressive castration-resistant disease, associated with transcriptional reprogramming, to a transitional epithelial-to-mesenchymal transition (EMT) phenotype rather than an upregulation of AR signaling. Unexpectedly, tolerance to therapy appears to be the default state, with treatment response correlating with the prevalence of tumor cells deficient for SNAI2, a key regulator of EMT reprogramming. CONCLUSION We show that EMT characterizes acutely resistant prostate tumors and that deletion of SNAI2, a key transcriptional regulator of EMT, correlates with clinical response.
Collapse
Affiliation(s)
- Marek Cmero
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia.,Division of Bioinformatics, Walter and Eliza Hall Institute, Parkville, Victoria, Australia.,Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Natalie J Kurganovs
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | - Ryan Stuchbery
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | - Patrick McCoy
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | - Corrina Grima
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | - Anne Ngyuen
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | - Ken Chow
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia.,Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Stefano Mangiola
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia.,Division of Bioinformatics, Walter and Eliza Hall Institute, Parkville, Victoria, Australia
| | - Geoff Macintyre
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas Howard
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Michael Kerger
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Philip Dundee
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Urology, Peninsula Health, Frankston, Victoria, Australia
| | - Paul Ruljancich
- Department of Urology, Box Hill Hospital, Box Hill, Victoria, Australia.,Epworth Eastern Hospital, Box Hill, Victoria, Australia
| | - David Clarke
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Jeremy Grummet
- Department of Urology, Alfred Hospital, Prahan, Victoria, Australia.,Monash University, Clayton, Victoria, Australia
| | - Justin S Peters
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Anthony J Costello
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia.,Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Sam Norden
- TissuPath, Mount Waverly, Victoria, Australia
| | - Andrew Ryan
- TissuPath, Mount Waverly, Victoria, Australia
| | - Phillip Parente
- Monash University, Clayton, Victoria, Australia.,Department of Medical Oncology, Box Hill Hospital, Box Hill, Victoria, Australia
| | - Christopher M Hovens
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia.,Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Niall M Corcoran
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia.,Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Urology, Peninsula Health, Frankston, Victoria, Australia.,Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Gross KM, Zhou W, Breindel JL, Ouyang J, Jin DX, Sokol ES, Gupta PB, Huber K, Zou L, Kuperwasser C. Loss of Slug Compromises DNA Damage Repair and Accelerates Stem Cell Aging in Mammary Epithelium. Cell Rep 2020; 28:394-407.e6. [PMID: 31291576 DOI: 10.1016/j.celrep.2019.06.043] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 04/16/2019] [Accepted: 06/11/2019] [Indexed: 12/27/2022] Open
Abstract
DNA damage activates checkpoints that limit the replicative potential of stem cells, including differentiation. These checkpoints protect against cancer development but also promote tissue aging. Because mice lacking Slug/Snai2 exhibit limited stem cell activity, including luminobasal differentiation, and are protected from mammary cancer, we reasoned that Slug might regulate DNA damage checkpoints in mammary epithelial cells. Here, we show that Slug facilitates efficient execution of RPA32-mediated DNA damage response (DDR) signaling. Slug deficiency leads to delayed phosphorylation of ataxia telangiectasia mutated and Rad3-related protein (ATR) and its effectors RPA32 and CHK1. This leads to impaired RAD51 recruitment to DNA damage sites and persistence of unresolved DNA damage. In vivo, Slug/Snai2 loss leads to increased DNA damage and premature aging of mammary epithelium. Collectively, our work demonstrates that the mammary stem cell regulator Slug controls DDR checkpoints by dually inhibiting differentiation and facilitating DDR repair, and its loss causes unresolved DNA damage and accelerated aging.
Collapse
Affiliation(s)
- Kayla M Gross
- Department of Developmental, Molecular, & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA; Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Wenhui Zhou
- Department of Developmental, Molecular, & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA; Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Jerrica L Breindel
- Department of Biomedical Sciences, Quinnipiac University, Hamden, CT 06518, USA
| | - Jian Ouyang
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Dexter X Jin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Ethan S Sokol
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Piyush B Gupta
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Kathryn Huber
- Department of Radiation Oncology, Tufts Medical Center, Boston, MA 02111, USA
| | - Lee Zou
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Charlotte Kuperwasser
- Department of Developmental, Molecular, & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA; Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
9
|
Vrenken KS, Vervoort BMT, van Ingen Schenau DS, Derks YHW, van Emst L, Grytsenko PG, Middelbeek JAJ, van Leeuwen FN. The transcriptional repressor SNAI2 impairs neuroblastoma differentiation and inhibits response to retinoic acid therapy. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165644. [PMID: 31862304 DOI: 10.1016/j.bbadis.2019.165644] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 12/03/2019] [Accepted: 12/14/2019] [Indexed: 02/07/2023]
Abstract
Neuroblastoma is the most common extracranial solid tumor in children and originates from poorly differentiated neural crest progenitors. High-risk neuroblastoma patients frequently present with metastatic disease at diagnosis. Despite intensive treatment, patients often develop refractory disease characterized by poorly differentiated, therapy resistant cells. Although adjuvant therapy using retinoic acid (RA)-induced differentiation may increase event-free survival, in the majority of cases response to RA-therapy is inadequate. Consequently, current research aims to identify novel therapeutic targets that enhance the sensitivity to RA and induce neuroblastoma cell differentiation. The similarities between neural crest development and neuroblastoma progression provide an appealing starting point. During neural crest development the EMT-transcription factor SNAI2 plays an important role in neural crest specification as well as neural crest cell migration and survival. Here, we report that CRISPR/Cas9 mediated deletion as well as shRNA mediated knockdown of the EMT-transcription factor SNAI2 promotes cellular differentiation in a variety of neuroblastoma models. By comparing mRNA expression data from independent patient cohorts, we show that a SNAI2 activity-based gene expression signature significantly correlates with event-free survival. Loss of SNAI2 function reduces self-renewal, 3D invasion as well as metastatic spread in vivo, while strongly sensitizing neuroblastoma cells to RA-induced growth inhibition. Together, our data demonstrate that SNAI2 maintains progenitor-like features in neuroblastoma cells while interfering with RA-induced growth inhibition. We propose that targeting gene regulatory circuits, such as those controlling SNAI2 function, may allow reversion of RA-therapy resistant neuroblastoma cells to a more differentiated and therapy responsive phenotype.
Collapse
Affiliation(s)
- Kirsten S Vrenken
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Britt M T Vervoort
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dorette S van Ingen Schenau
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yvonne H W Derks
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Liesbeth van Emst
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Pavlo G Grytsenko
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen A J Middelbeek
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Frank N van Leeuwen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
10
|
Zhou W, Gross KM, Kuperwasser C. Molecular regulation of Snai2 in development and disease. J Cell Sci 2019; 132:132/23/jcs235127. [PMID: 31792043 DOI: 10.1242/jcs.235127] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The transcription factor Snai2, encoded by the SNAI2 gene, is an evolutionarily conserved C2H2 zinc finger protein that orchestrates biological processes critical to tissue development and tumorigenesis. Initially characterized as a prototypical epithelial-to-mesenchymal transition (EMT) transcription factor, Snai2 has been shown more recently to participate in a wider variety of biological processes, including tumor metastasis, stem and/or progenitor cell biology, cellular differentiation, vascular remodeling and DNA damage repair. The main role of Snai2 in controlling such processes involves facilitating the epigenetic regulation of transcriptional programs, and, as such, its dysregulation manifests in developmental defects, disruption of tissue homeostasis, and other disease conditions. Here, we discuss our current understanding of the molecular mechanisms regulating Snai2 expression, abundance and activity. In addition, we outline how these mechanisms contribute to disease phenotypes or how they may impact rational therapeutic targeting of Snai2 dysregulation in human disease.
Collapse
Affiliation(s)
- Wenhui Zhou
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA.,Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kayla M Gross
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA.,Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Charlotte Kuperwasser
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA .,Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
11
|
Li Z, Chen Y, An T, Liu P, Zhu J, Yang H, Zhang W, Dong T, Jiang J, Zhang Y, Jiang M, Yang X. Nuciferine inhibits the progression of glioblastoma by suppressing the SOX2-AKT/STAT3-Slug signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:139. [PMID: 30922391 PMCID: PMC6440136 DOI: 10.1186/s13046-019-1134-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/13/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Nuciferine (NF), extracted from the leaves of N. nucifera Gaertn, has been shown to exhibit anti-tumor and anti-viral pharmacological properties. It can also penetrate the blood brain barrier (BBB). However, the mechanism by which NF inhibits glioblastoma (GBM) progression is not well understood. We aimed to determine the anti-tumor effect of NF on GBM cell lines and clarify the potential molecular mechanism involved. METHODS U87MG and U251 cell lines were used in vitro to assess the anti-tumor efficacy of NF. Cytotoxicity, viability, and proliferation were evaluated by MTT and colony formation assay. After Annexin V-FITC and PI staining, flow cytometry was performed to evaluate apoptosis and cell cycle changes in NF-treated GBM cells. Wound healing and Transwell assays were used to assess migration and invasion of GBM cells. Western blot analysis, immunofluorescence staining, immunohistochemistry, and bioinformatics were used to gain insights into the molecular mechanisms. Preclinical therapeutic efficacy was mainly estimated by ultrasound and MRI in xenograft nude mouse models. RESULTS NF inhibited the proliferation, mobility, stemness, angiogenesis, and epithelial-to-mesenchymal transition (EMT) of GBM cells. Additionally, NF induced apoptosis and G2 cell cycle arrest. Slug expression was also decreased by NF via the AKT and STAT3 signaling pathways. Interestingly, we discovered that NF affected GBM cells partly by targeting SOX2, which may be upstream of the AKT and STAT3 pathways. Finally, NF led to significant tumor control in GBM xenograft models. CONCLUSIONS NF inhibited the progression of GBM via the SOX2-AKT/STAT3-Slug signaling pathway. SOX2-targeting with NF may offer a novel therapeutic approach for GBM treatment.
Collapse
Affiliation(s)
- Zizhuo Li
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Yaodong Chen
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Tingting An
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Pengfei Liu
- Department of Magnetic Resonance, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Jiyuan Zhu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Haichao Yang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Wei Zhang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Tianxiu Dong
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Jian Jiang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Yu Zhang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Maitao Jiang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xiuhua Yang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
| |
Collapse
|
12
|
FAM3B/PANDER inhibits cell death and increases prostate tumor growth by modulating the expression of Bcl-2 and Bcl-X L cell survival genes. BMC Cancer 2018; 18:90. [PMID: 29357840 PMCID: PMC5778767 DOI: 10.1186/s12885-017-3950-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023] Open
Abstract
Background FAM3B/PANDER is a novel cytokine-like protein that induces apoptosis in insulin-secreting beta-cells. Since in silico data revealed that FAM3B can be expressed in prostate tumors, we evaluated the putative role of this cytokine in prostate tumor progression. Methods FAM3B expression was analyzed by quantitative PCR in tumor tissue clinical samples and prostate tumor cell lines. Culture growth and viability of DU145 cell line were evaluated after treatment with either exogenous FAM3B protein obtained from conditioned media (CM) of 293 T cells overexpressing FAM3B or a recombinant FAM3B protein produced in a bacterial host. DU145 cells overexpressing FAM3B protein were produced by lentiviral-mediated transduction of full-length FAM3B cDNA. Cell viability and apoptosis were analyzed in DU145/FAM3B cells after treatment with several cell death inducers, such as TNF-alpha, staurosporine, etoposide, camptothecin, and serum starvation conditions. Anchorage-independent growth in soft agarose assay was used to evaluate in vitro tumorigenicity. In vivo tumorigenicity and invasiveness were evaluated by tumor xenograft growth in nude mice. Results We observed an increase in FAM3B expression in prostate tumor samples when compared to normal tissues. DU145 cell viability and survival increased after exogenous treatment with recombinant FAM3B protein or FAM3B-secreted protein. Overexpression of FAM3B in DU145 cells promoted inhibition of DNA fragmentation and phosphatidylserine externalization in a time and dose-dependent fashion, upon apoptosis triggered by TNF-alpha. These events were accompanied by increased gene expression of anti-apoptotic Bcl-2 and Bcl-XL, decreased expression of pro-apoptotic Bax and diminished caspase-3, −8 and −9 proteolytic activities. Furthermore, inhibition of Bcl-2 anti-apoptotic family proteins with small molecules antagonists decreases protective effects of FAM3B in DU145 cells. When compared to the respective controls, cells overexpressing FAM3B displayed a decreased anchorage- independent growth in vitro and increased tumor growth in xenografted nude mice. The immunohistochemistry analysis of tumor xenografts revealed a similar anti-apoptotic phenotype displayed by FAM3B-overexpressing tumor cells. Conclusions Taken together, by activating pro-survival mechanisms FAM3B overexpression contributes to increased resistance to cell death and tumor growth in nude mice, highlighting a putative role for this cytokine in prostate cancer progression. Electronic supplementary material The online version of this article (10.1186/s12885-017-3950-9) contains supplementary material, which is available to authorized users.
Collapse
|
13
|
Gallik KL, Treffy RW, Nacke LM, Ahsan K, Rocha M, Green-Saxena A, Saxena A. Neural crest and cancer: Divergent travelers on similar paths. Mech Dev 2017; 148:89-99. [PMID: 28888421 PMCID: PMC5811199 DOI: 10.1016/j.mod.2017.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 08/20/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
Neural crest cells are multipotent progenitors that dynamically interpret diverse microenvironments to migrate significant distances as a loosely associated collective and contribute to many tissues in the developing vertebrate embryo. Uncovering details of neural crest migration has helped to inform a general understanding of collective cell migration, including that which occurs during cancer metastasis. Here, we discuss several commonalities and differences of neural crest and cancer cell migration and behavior. First, we focus on some of the molecular pathways required for the initial specification and potency of neural crest cells and the roles of many of these pathways in cancer progression. We also describe epithelial-to-mesenchymal transition, which plays a critical role in initiating both neural crest migration and cancer metastasis. Finally, we evaluate studies that demonstrate myriad forms of cell-cell and cell-environment communication during neural crest and cancer collective migration to highlight the remarkable similarities in their molecular and cell biological regulation.
Collapse
Affiliation(s)
- Kristin L Gallik
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Randall W Treffy
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Lynne M Nacke
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kamil Ahsan
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA
| | - Manuel Rocha
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA
| | - Abigail Green-Saxena
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Ankur Saxena
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
14
|
SLUG transcription factor: a pro-survival and prognostic factor in gastrointestinal stromal tumour. Br J Cancer 2017; 116:1195-1202. [PMID: 28334729 PMCID: PMC5418455 DOI: 10.1038/bjc.2017.82] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The SLUG transcription factor has been linked with the KIT signalling pathway that is important for gastrointestinal stromal tumour (GIST) tumourigenesis. Its clinical significance in GIST is unknown. METHODS Influence of SLUG expression on cell proliferation and viability were investigated in GIST48 and GIST882 cell lines. The association between tumour SLUG expression in immunohistochemistry and recurrence-free survival (RFS) was studied in two clinical GIST series, one with 187 patients treated with surgery alone, and another one with 313 patients treated with surgery and adjuvant imatinib. RESULTS SLUG downregulation inhibited cell proliferation, induced cell death in both cell lines, and sensitised GIST882 cells to lower imatinib concentrations. SLUG was expressed in 125 (25.0%) of the 500 clinical GISTs evaluated, and expression was associated with several factors linked with unfavourable prognosis. SLUG expression was associated with unfavourable RFS both when patients were treated with surgery alone (HR=3.40, 95% CI=1.67-6.89, P=0.001) and when treated with surgery plus adjuvant imatinib (HR=1.83, 95% CI=1.29-2.60, P=0.001). CONCLUSIONS GIST patients with high tumour SLUG expression have unfavourable RFS. SLUG may mediate pro-survival signalling in GISTs.
Collapse
|
15
|
Zhao XY, Li L, Wang XB, Fu RJ, Lv YP, Jin W, Meng C, Chen GQ, Huang L, Zhao KW. Inhibition of Snail Family Transcriptional Repressor 2 (SNAI2) Enhances Multidrug Resistance of Hepatocellular Carcinoma Cells. PLoS One 2016; 11:e0164752. [PMID: 27760172 PMCID: PMC5070735 DOI: 10.1371/journal.pone.0164752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 10/02/2016] [Indexed: 01/22/2023] Open
Abstract
China accounts for almost half of the total number of liver cancer cases and deaths worldwide, and hepatocellular carcinoma (HCC) is the most primary liver cancer. Snail family transcriptional repressor 2 (SNAI2) is known as an epithelial to mesenchymal transition-inducing transcription factor that drives neoplastic epithelial cells into mesenchymal phenotype. However, the roles of endogenous SNAI2 remain controversial in different types of malignant tumors. Herein, we surprisingly identify that anchorage-independent growth, including the formation of tumor sphere and soft agar colony, is significantly increased when SNAI2 expression is inhibited by shRNAs in HCC cells. Suppression of SNAI2 suffices to up-regulate several cancer stem genes. Although unrelated to the metastatic ability, SNAI2 inhibition does increase the efflux of Hoechst 33342 and enhance multidrug resistance in vitro and in vivo. In agreement with this data, we demonstrate for the first time that decreasing SNAI2 level can transcriptionally upregulate several ATP binding cassette (ABC) transporter genes such as ABCB1. Moreover, ABC transporters' inhibitor verapamil can rescue the multidrug resistance induced by SNAI2 inhibition. Our results implicate that SNAI2 behaves as a tumor suppressor by inhibiting multidrug resistance via suppressing ABC transporter genes in HCC cells.
Collapse
Affiliation(s)
- Xin-Yu Zhao
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Lei Li
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xiao-Bo Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Rong-Jie Fu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences and SJTU-SM, Shanghai, China
| | - Ya-Ping Lv
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Wei Jin
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Chao Meng
- Ren-Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo-Qiang Chen
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Lei Huang
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Ke-Wen Zhao
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| |
Collapse
|
16
|
Middelbeek J, Visser D, Henneman L, Kamermans A, Kuipers AJ, Hoogerbrugge PM, Jalink K, van Leeuwen FN. TRPM7 maintains progenitor-like features of neuroblastoma cells: implications for metastasis formation. Oncotarget 2016; 6:8760-76. [PMID: 25797249 PMCID: PMC4496182 DOI: 10.18632/oncotarget.3315] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/08/2015] [Indexed: 12/18/2022] Open
Abstract
Neuroblastoma is an embryonal tumor derived from poorly differentiated neural crest cells. Current research is aimed at identifying the molecular mechanisms that maintain the progenitor state of neuroblastoma cells and to develop novel therapeutic strategies that induce neuroblastoma cell differentiation. Mechanisms controlling neural crest development are typically dysregulated during neuroblastoma progression, and provide an appealing starting point for drug target discovery. Transcriptional programs involved in neural crest development act as a context dependent gene regulatory network. In addition to BMP, Wnt and Notch signaling, activation of developmental gene expression programs depends on the physical characteristics of the tissue microenvironment. TRPM7, a mechanically regulated TRP channel with kinase activity, was previously found essential for embryogenesis and the maintenance of undifferentiated neural crest progenitors. Hence, we hypothesized that TRPM7 may preserve progenitor-like, metastatic features of neuroblastoma cells. Using multiple neuroblastoma cell models, we demonstrate that TRPM7 expression closely associates with the migratory and metastatic properties of neuroblastoma cells in vitro and in vivo. Moreover, microarray-based expression profiling on control and TRPM7 shRNA transduced neuroblastoma cells indicates that TRPM7 controls a developmental transcriptional program involving the transcription factor SNAI2. Overall, our data indicate that TRPM7 contributes to neuroblastoma progression by maintaining progenitor-like features.
Collapse
Affiliation(s)
- Jeroen Middelbeek
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Daan Visser
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Linda Henneman
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Arthur J Kuipers
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Peter M Hoogerbrugge
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Princes Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Kees Jalink
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Ferrari-Amorotti G, Chiodoni C, Shen F, Cattelani S, Soliera AR, Manzotti G, Grisendi G, Dominici M, Rivasi F, Colombo MP, Fatatis A, Calabretta B. Suppression of invasion and metastasis of triple-negative breast cancer lines by pharmacological or genetic inhibition of slug activity. Neoplasia 2015; 16:1047-58. [PMID: 25499218 PMCID: PMC4557365 DOI: 10.1016/j.neo.2014.10.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 10/10/2014] [Accepted: 10/13/2014] [Indexed: 01/13/2023] Open
Abstract
Most triple-negative breast cancers (TNBCs) exhibit gene expression patterns associated with epithelial-to-mesenchymal transition (EMT), a feature that correlates with a propensity for metastatic spread. Overexpression of the EMT regulator Slug is detected in basal and mesenchymal-type TNBCs and is associated with reduced E-cadherin expression and aggressive disease. The effects of Slug depend, in part, on the interaction of its N-terminal SNAG repressor domain with the chromatin-modifying protein lysine demethylase 1 (LSD1); thus, we investigated whether tranylcypromine [also known as trans-2-phenylcyclopropylamine hydrochloride (PCPA) or Parnate], an inhibitor of LSD1 that blocks its interaction with Slug, suppresses the migration, invasion, and metastatic spread of TNBC cell lines. We show here that PCPA treatment induces the expression of E-cadherin and other epithelial markers and markedly suppresses migration and invasion of TNBC cell lines MDA-MB-231 and BT-549. These effects were phenocopied by Slug or LSD1 silencing. In two models of orthotopic breast cancer, PCPA treatment reduced local tumor growth and the number of lung metastases. In mice injected directly in the blood circulation with MDA-MB-231 cells, PCPA treatment or Slug silencing markedly inhibited bone metastases but had no effect on lung infiltration. Thus, blocking Slug activity may suppress the metastatic spread of TNBC and, perhaps, specifically inhibit homing/colonization to the bone.
Collapse
Affiliation(s)
- Giovanna Ferrari-Amorotti
- Dipartimento di Medicina Diagnostica, Clinica a di Sanità Pubblica, University of Modena and Reggio Emilia, Modena, Italy.
| | | | - Fei Shen
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, PA, USA
| | - Sara Cattelani
- Dipartimento di Medicina Diagnostica, Clinica a di Sanità Pubblica, University of Modena and Reggio Emilia, Modena, Italy
| | - Angela Rachele Soliera
- Dipartimento di Medicina Diagnostica, Clinica a di Sanità Pubblica, University of Modena and Reggio Emilia, Modena, Italy
| | - Gloria Manzotti
- Dipartimento di Medicina Diagnostica, Clinica a di Sanità Pubblica, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto, Modena, Italy
| | - Massimo Dominici
- Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto, Modena, Italy
| | - Francesco Rivasi
- Dipartimento di Anatomia Patologica e Medicina Legale, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Alessandro Fatatis
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, PA, USA; Department of Pathology, Drexel University, Philadelphia, PA, USA; Program in Biology of Prostate Cancer, Kimmel Cancer Center, Philadelphia, PA, USA
| | - Bruno Calabretta
- Dipartimento di Medicina Diagnostica, Clinica a di Sanità Pubblica, University of Modena and Reggio Emilia, Modena, Italy; Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Elevated TGF-β1 and -β2 expression accelerates the epithelial to mesenchymal transition in triple-negative breast cancer cells. Cytokine 2015; 75:151-8. [PMID: 26088755 DOI: 10.1016/j.cyto.2015.05.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 04/07/2015] [Accepted: 05/23/2015] [Indexed: 12/31/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is a key process in tumor invasion and migration. Transforming growth factor-βs (TGF-βs) are multifunctional growth factors and powerful modulators of the EMT. Here, we investigated the relationship between TGF-β expression and invasion by treated triple-negative breast cancer (TNBC) cells. Our results show that invasion capacity of TNBC cells was markedly higher than that of non-TNBC cells. In addition, EMT-related gene signatures, including vimentin (vim), fibronectin (FN), snail, and slug were highly expressed in TNBC cells. Interestingly, our results show that TGF-β1 and β2 mRNA expression levels were higher in TNBC cells than those in non-TNBC cells. Thus, we examined the effect of the TGF-β receptor I/II inhibitor LY2109761 on EMT-related gene expression and cell motility. Our data show that vim, FN, and slug mRNA expression levels dose-dependently decreased in response to LY2109761. TNBC cell motility also decreased in response to LY2109761. Finally, we investigated the effect of LY2109761 on TGF-β1 or TGF-β2-induced E-cadherin (E-cad), vim, and FN mRNA and protein expression. The reduction in E-cad and induction of vim and FN expression by TGF-β1 or TGF-β2 were completely reversed by LY2109761 treatment in HCC1806 TNBC cells. Taken together, we demonstrated that elevated TGF-β expression triggers invasion and migration by TNBCs through the EMT process. Inhibiting the TGF-β signaling pathway is considered a promising therapeutic strategy for treating TNBC.
Collapse
|
19
|
Chen PM, Cheng YW, Wu TC, Chen CY, Lee H. MnSOD overexpression confers cisplatin resistance in lung adenocarcinoma via the NF-κB/Snail/Bcl-2 pathway. Free Radic Biol Med 2015; 79:127-37. [PMID: 25499851 DOI: 10.1016/j.freeradbiomed.2014.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 11/03/2014] [Accepted: 12/02/2014] [Indexed: 12/27/2022]
Abstract
Manganese superoxide dismutase (MnSOD) has been shown to be associated with doxorubicin resistance in gastric cancer cells, but the underlying mechanism of MnSOD in drug resistance remains unclear. A recent study indicated that NF-κB activation by MnSOD promoted tumor malignancy in lung adenocarcinoma. Therefore, we hypothesized that MnSOD-mediated NF-κB activation might confer cisplatin resistance in lung adenocarcinoma via the NF-κB/Bcl-2/Snail pathway. Here, the inhibition concentration of cisplatin with 50% cell viability (IC50) was positively correlated with MnSOD expression and its activity in a panel of lung adenocarcinoma cells. The IC50 value was markedly increased and decreased by MnSOD overexpression and knockdown, respectively, in lung cancer cells. Mechanistically, an increase in Bcl-2 by MnSOD-mediated NF-κB activation confers greater cisplatin resistance than cIAP2, Bcl-xL, Mcl-1, and Snail. MnSOD-mediated cisplatin resistance can be overcome by a Bcl-2 antagonist (ABT-199) or IKKβ inhibitor (curcumin) in cells and xenograft tumors. MnSOD expression was positively correlated with nuclear p65 protein and Bcl-2 mRNA expression in tumors from patients with lung adenocarcinomas. A retrospective study indicated that it was more common for MnSOD-positive, nuclear p65-positive, or high Bcl-2 mRNA tumors to have an unfavorable response to cisplatin-based chemotherapy than their counterparts. Therefore, we suggest that ABT-199 or curcumin may be potentially useful to improve tumor regression and chemotherapeutic response in patients with MnSOD/Bcl-2-positive tumors.
Collapse
Affiliation(s)
- Po-Ming Chen
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 115, Taiwan
| | - Ya-Wen Cheng
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 115, Taiwan
| | - Tzu-Chin Wu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Yi Chen
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 115, Taiwan.
| |
Collapse
|
20
|
Lai WY, Wang WY, Chang YC, Chang CJ, Yang PC, Peck K. Synergistic inhibition of lung cancer cell invasion, tumor growth and angiogenesis using aptamer-siRNA chimeras. Biomaterials 2014; 35:2905-14. [PMID: 24397988 DOI: 10.1016/j.biomaterials.2013.12.054] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/19/2013] [Indexed: 02/07/2023]
Abstract
Early metastasis is one of the major causes of mortality among patient with lung cancer. The process of tumor metastasis involves a cascade of events, including epithelial-mesenchymal transition, tumor cell migration and invasion, and angiogenesis. To specifically suppress tumor invasion and angiogenesis, two nucleolin aptamer-siRNA chimeras (aptNCL-SLUGsiR and aptNCL-NRP1siR) were used to block key signaling pathways involved in lung cancer metastasis that are pivotal to metastatic tumor cells but not to normal cells under ordinary physiologic conditions. Through nucleolin-mediated endocytosis, the aptNCL-siRNA chimeras specifically and significantly knocked down the expressions of SLUG and NRP1 in nucleolin-expressing cancer cells. Furthermore, simultaneous suppression of SLUG and NRP1 expressions by the chimeras synergistically retarded cancer cell motility and invasive ability. The synergistic effect was also observed in a xenograft mouse model, wherein the combined treatment using two chimeras suppressed tumor growth, the invasiveness, circulating tumor cell amount, and angiogenesis in tumor tissue without affecting liver and kidney functions. This study demonstrates that combined treatment of aptNCL-SLUGsiR and aptNCL-NRP1siR can synergistically suppress lung cancer cell invasion, tumor growth and angiogenesis by cancer-specific targeting combined with gene-specific silencing.
Collapse
Affiliation(s)
- Wei-Yun Lai
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 115, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Wei-Ya Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yi-Chung Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Cheng-Ju Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Pan-Chyr Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; National Taiwan University, Taipei 100, Taiwan.
| | - Konan Peck
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
21
|
Schwankhaus N, Gathmann C, Wicklein D, Riecken K, Schumacher U, Valentiner U. Cell adhesion molecules in metastatic neuroblastoma models. Clin Exp Metastasis 2014; 31:483-96. [PMID: 24549749 DOI: 10.1007/s10585-014-9643-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 02/03/2014] [Indexed: 12/11/2022]
Abstract
Several cell adhesion molecules (CAMs) including selectins, integrins, cadherins and immunoglobulin-like CAMs are involved in leukocyte adhesion especially at sites of inflammation. In cancer cells, these CAMs have been associated with the growth and metastatic behavior in several malignant entities. In this study adhesion of LAN 1 and SK-N-SH neuroblastoma cells to selectins, hyaluronan and endothelial cells were determined under flow conditions. Furthermore cells were injected subcutaneously into wildtype and selectin deficient scid mice and their growth and metastatic behavior were analyzed. Under shear stress SK-N-SH cells firmly adhered to E-selectin-Fc-fusion protein, hyaluronan and endothelial cells, while LAN 1 cells showed less or hardly any adhesive events by comparison. In the SK-N-SH xenograft model metastasis formation was slightly dependent on the expression of selectins, while LAN 1 cells developed metastases completely independent of selectin expression. The different adhesive and metastatic properties of LAN 1 and SK-N-SH cells are reflected by a different expression profile of several CAMs. The results indicate that endothelial selectins are not essential for metastasis formation of human LAN 1 and SK-N-SH cells. However, other CAMs namely CD44, N-cadherin, NCAM and integrins were upregulated or downregulated, respectively, in SK-N-SH and LAN 1 cells and are potential adhesion molecules involved in the metastatic cascade of these cells.
Collapse
Affiliation(s)
- Nina Schwankhaus
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Christina Gathmann
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Daniel Wicklein
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Clinic for Stem Cell Transplantation, Center for Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Udo Schumacher
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ursula Valentiner
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
22
|
Kong Y, Ma LQ, Bai PS, Da R, Sun H, Qi XG, Ma JQ, Zhao RM, Chen NZ, Nan KJ. Helicobacter pylori promotes invasion and metastasis of gastric cancer cells through activation of AP-1 and up-regulation of CACUL1. Int J Biochem Cell Biol 2013; 45:2666-78. [PMID: 24004834 DOI: 10.1016/j.biocel.2013.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 08/22/2013] [Accepted: 08/25/2013] [Indexed: 02/07/2023]
Abstract
Infection with Helicobacter pylori is important in the development and progression of gastric cancer. However, the mechanisms that regulate this activation in gastric tumors remain elusive. CACUL1 has been cloned and identified as a novel gene that is expressed in many types of cancer and is involved in cell cycle regulation and tumor growth. The current study aimed to examine the expression of CACUL1 in gastric cancer samples and analyze its correlation with H. pylori infection. We found that CACUL1 was highly expressed in gastric cancer tissues and negatively correlated with gastric cancer differentiation and TNM stage. In addition, CACUL1 expression was high in H. pylori-infected tissues compared with H. pylori non-infected tissue. We found that H. pylori could up-regulate CACUL1 expression through activating protein 1. The up-regulation of CACUL1 expression could promote matrix metalloproteinase 9 and Slug expression to increase invasion and metastasis of tumor cells. These results suggested that H. pylori-triggered CACUL1 production occurred in an activating protein 1-dependent manner and regulated matrix metalloproteinase 9 and Slug expression to affect the invasion and metastasis of tumor cells. Therefore, CACUL1 is a potential therapeutic target for the treatment of aggressive gastric cancer.
Collapse
Affiliation(s)
- Ying Kong
- Department of Oncology, First Hospital of Xi'an Jiaotong University, No. 277 YanTa West Road, Xi'an, Shaanxi 710061, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Davis FM, Parsonage MT, Cabot PJ, Parat MO, Thompson EW, Roberts-Thomson SJ, Monteith GR. Assessment of gene expression of intracellular calcium channels, pumps and exchangers with epidermal growth factor-induced epithelial-mesenchymal transition in a breast cancer cell line. Cancer Cell Int 2013; 13:76. [PMID: 23890218 PMCID: PMC3733826 DOI: 10.1186/1475-2867-13-76] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 07/24/2013] [Indexed: 01/05/2023] Open
Abstract
Background Epithelial-mesenchymal transition (EMT) is a process implicated in cancer metastasis that involves the conversion of epithelial cells to a more mesenchymal and invasive cell phenotype. In breast cancer cells EMT is associated with altered store-operated calcium influx and changes in calcium signalling mediated by activation of cell surface purinergic receptors. In this study, we investigated whether MDA-MB-468 breast cancer cells induced to undergo EMT exhibit changes in mRNA levels of calcium channels, pumps and exchangers located on intracellular calcium storing organelles, including the Golgi, mitochondria and endoplasmic reticulum (ER). Methods Epidermal growth factor (EGF) was used to induce EMT in MDA-MB-468 breast cancer cells. Serum-deprived cells were treated with EGF (50 ng/mL) for 12 h and gene expression was assessed using quantitative RT-PCR. Results and conclusions These data reveal no significant alterations in mRNA levels of the Golgi calcium pump secretory pathway calcium ATPases (SPCA1 and SPCA2), or the mitochondrial calcium uniporter (MCU) or Na+/Ca2+ exchanger (NCLX). However, EGF-induced EMT was associated with significant alterations in mRNA levels of specific ER calcium channels and pumps, including (sarco)-endoplasmic reticulum calcium ATPases (SERCAs), and inositol 1,4,5-trisphosphate receptor (IP3R) and ryanodine receptor (RYR) calcium channel isoforms. The most prominent change in gene expression between the epithelial and mesenchymal-like states was RYR2, which was enriched 45-fold in EGF-treated MDA-MB-468 cells. These findings indicate that EGF-induced EMT in breast cancer cells may be associated with major alterations in ER calcium homeostasis.
Collapse
Affiliation(s)
- Felicity M Davis
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | | | | | | | |
Collapse
|
24
|
Teitz T, Inoue M, Valentine MB, Zhu K, Rehg JE, Zhao W, Finkelstein D, Wang YD, Johnson MD, Calabrese C, Rubinstein M, Hakem R, Weiss WA, Lahti JM. Th-MYCN mice with caspase-8 deficiency develop advanced neuroblastoma with bone marrow metastasis. Cancer Res 2013; 73:4086-97. [PMID: 23536557 PMCID: PMC3702642 DOI: 10.1158/0008-5472.can-12-2681] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neuroblastoma, the most common extracranial pediatric solid tumor, is responsible for 15% of all childhood cancer deaths. Patients frequently present at diagnosis with metastatic disease, particularly to the bone marrow. Advances in therapy and understanding of the metastatic process have been limited due, in part, to the lack of animal models harboring bone marrow disease. The widely used transgenic model, the Th-MYCN mouse, exhibits limited metastasis to this site. Here, we establish the first genetic immunocompetent mouse model for metastatic neuroblastoma with enhanced secondary tumors in the bone marrow. This model recapitulates 2 frequent alterations in metastatic neuroblastoma, overexpression of MYCN and loss of caspase-8 expression. Mouse caspase-8 gene was deleted in neural crest lineage cells by crossing a Th-Cre transgenic mouse with a caspase-8 conditional knockout mouse. This mouse was then crossed with the neuroblastoma prone Th-MYCN mouse. Although overexpression of MYCN by itself rarely caused bone marrow metastasis, combining MYCN overexpression and caspase-8 deletion significantly enhanced bone marrow metastasis (37% incidence). Microarray expression studies of the primary tumors mRNAs and microRNAs revealed extracellular matrix structural changes, increased expression of genes involved in epithelial to mesenchymal transition, inflammation, and downregulation of miR-7a and miR-29b. These molecular changes have been shown to be associated with tumor progression and activation of the cytokine TGF-β pathway in various tumor models. Cytokine TGF-β can preferentially promote single cell motility and blood-borne metastasis and therefore activation of this pathway may explain the enhanced bone marrow metastasis observed in this animal model.
Collapse
Affiliation(s)
- Tal Teitz
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital,
Memphis, TN 38105, USA
| | - Madoka Inoue
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital,
Memphis, TN 38105, USA
| | - Marcus B. Valentine
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital,
Memphis, TN 38105, USA
| | - Kejin Zhu
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital,
Memphis, TN 38105, USA
| | - Jerold E. Rehg
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
38105, USA
| | - Wei Zhao
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis,
TN 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital,
Memphis, TN 38105, USA
| | - Yong-Dong Wang
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s
Research Hospital, Memphis, TN 38105, USA
| | - Melissa D. Johnson
- Animal Imaging Center, St. Jude Children’s Research Hospital, Memphis, TN
38105, USA
| | - Christopher Calabrese
- Animal Imaging Center, St. Jude Children’s Research Hospital, Memphis, TN
38105, USA
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología
Molecular, Consejo Nacional de Investigaciones Científicas y Tecnológicas and
Universidad de Buenos Aires C1428ADN, Argentina
| | - Razqallah Hakem
- Department of Medical Biophysics, Ontario Cancer Institute, University of Toronto,
Toronto, Ontario M5G 2M9, Canada
| | - William A. Weiss
- Departments of Neurology, Pediatrics and Neurological Surgery, University of
California, San Francisco, CA 94158, USA
| | - Jill M. Lahti
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital,
Memphis, TN 38105, USA
| |
Collapse
|
25
|
Wang YP, Wang MZ, Luo YR, Shen Y, Wei ZX. Lentivirus-mediated shRNA interference targeting SLUG inhibits lung cancer growth and metastasis. Asian Pac J Cancer Prev 2013; 13:4947-51. [PMID: 23244088 DOI: 10.7314/apjcp.2012.13.10.4947] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Lung cancer is a deadly cancer, whose kills more people worldwide than any other malignancy. SLUG (SNAI2, Snail2) is involved in the epithelial mesenchymal transition in physiological and in pathological contexts and is implicated in the development and progression of lung cancer. METHODS We constructed a lentivirus vector with SLUG shRNA (LV-shSLUG). LV-shSLUG and a control lentivirus were infected into the non-small cell lung cancer cell A549 and real-time PCR, Western blot and IHC were applied to assess expression of the SLUG gene. Cell proliferation and migration were detected using MTT and clony formation methods. RESULTS Real-time PCR, Western Blot and IHC results confirmed down-regulation of SLUG expression by its shRNA by about 80%~90% at both the mRNA and protein levels. Knockdown of SLUG significantly suppressed lung cancer cell proliferation. Furthermore, knockdown of SLUG significantly inhibited lung cancer cell invasion and metastasis. Finally, knockdown of SLUG induced the down-regulation of Bcl-2 and up-regulation of E-cadherin. CONCLUSION These results indicate that SLUG is a newly identified gene associated with lung cancer growth and metastasis. SLUG may serve as a new therapeutic target for the treatment of lung cancer in the future.
Collapse
Affiliation(s)
- Yao-Peng Wang
- Department of Thoracic Surgery, Affiliated Hospital of Medical College Qingdao University, Qingdao, Shandong, China
| | | | | | | | | |
Collapse
|
26
|
Toll-like receptor 3 expression inhibits cell invasion and migration and predicts a favorable prognosis in neuroblastoma. Cancer Lett 2013; 336:338-46. [PMID: 23541683 DOI: 10.1016/j.canlet.2013.03.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 03/17/2013] [Accepted: 03/20/2013] [Indexed: 12/20/2022]
Abstract
To evaluate the clinical significance of TLR3 expression on neuroblastomas, we performed immunohistochemical study on archival tissues and in vitro studies on neuroblastoma cell lines. The results showed that positive TLR3 expression was associated with favorable histology and prognosis. Activation of TLR3 by polyinosinic:polycytidylic acid [poly(I:C)] treatment is effective to suppress cell migration and invasion and to decrease organized assembly of F-actin and filopodia formation, in TLR3-expressing SK-N-AS cells, which could be reversed by TLR3-targeting siRNA treatment. TLR3 agonist poly(I:C) promotes GAP-43 expression also in SK-N-AS cells only. Taken together, TLR3 could serve to predict favorable behavior in neuroblastomas.
Collapse
|
27
|
Wang Y, Yue B, Yu X, Wang Z, Wang M. SLUG is activated by nuclear factor kappa B and confers human alveolar epithelial A549 cells resistance to tumor necrosis factor-alpha-induced apoptosis. World J Surg Oncol 2013; 11:12. [PMID: 23339680 PMCID: PMC3561261 DOI: 10.1186/1477-7819-11-12] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 12/23/2012] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The role of tumor necrosis factor alpha (TNF-α) in cancer is complex with both apoptotic and anti-apoptotic roles proposed. However the mechanism is not clear. In the study, we designed to investigate the effect of TNF-α on the activation and expression of nuclear factor kappa B (NF-κB)/p65/SLUG/PUMA/Bcl-2 levels in human lung cancer A549 cell line, and in conditions of TNF-α-induced apoptosis. METHODS We have engineered three A549 cell lines that were transiently transfected with PUMA siRNA, SLUG siRNA and Bcl-2 siRNA, respectively. We have measured the in vitro effects of siRNA on apoptosis, and sensitivity to 20 ng/ml of TNF-α treatment for 24-48 h. RESULTS We found the NF-κB activity and PUMA mRNA/protein was significantly increased after treatment of TNF-α for 24 h in untreated A549 cells, and led to a significant increase in TNF-α-induced apoptosis, no significant increase of SLUG and Bcl-2 level was shown. However, after treatment of TNF-α for 48 h in untreated A549 cells, SLUG and Bcl-2 level was significant increased, and PUMA level was significant decreased, and TNF-α-induced apoptosis was significantly decreased compared to the apoptosis level after treatment of TNF-α for 24 h. Inhibition of the NF-κB activity could effectively decrease the PUMA level and increase the SLUG and Bcl-2 level. PUMA silencing by siRNA led to a significant decrease in TNF-α-induced apoptosis after treatment of TNF-α for 24 h. Bcl-2 and SLUG silencing by siRNA led to a significant increase in TNF-α-induced apoptosis for 48 h. Furthermore, SLUG silencing increased PUMA level and decreased Bcl-2 level. CONCLUSIONS The findings suggested that TNF-α treatment promoted apoptosis via the NF-κB-dependent PUMA pathway. The anti-apoptotic role of TNF-α was via NF-κB-dependent SLUG and Bcl-2 pathway at a later time.
Collapse
Affiliation(s)
- Yaopeng Wang
- Department of Thoracic Surgery, the Affiliated Hospital of Medical College Qingdao University, 19 Jiangsu Road, Qingdao, Shandong, 266001, China.
| | | | | | | | | |
Collapse
|
28
|
SFRP2 and slug contribute to cellular resistance to apoptosis in hypertrophic scars. PLoS One 2012; 7:e50229. [PMID: 23226515 PMCID: PMC3514356 DOI: 10.1371/journal.pone.0050229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 10/22/2012] [Indexed: 01/19/2023] Open
Abstract
Hypertrophic scars (HS) are skin disorders which occur after wounding and thermal injury. Our previous studies have suggested that secreted frizzled-related protein 2 (SFRP2) is involved in HS formation and that the suppression of SFRP2 promotes apoptosis of hypertrophic scar fibroblasts (HSFBs). However, the mechanisms have not been clarified. Previous studies revealed that Slug expression inhibits cell apoptosis, in vitro and in vivo, and SFRP2 regulates the expression of Slug in cervical cancer cells. In the present study, we quantified differential expression levels of expression of SFRP2 and Slug in HS and normal skin tissues by immunohistochemistry, both of which have important anti-apoptosis roles. Furthermore, a short hairpin RNA approach was adopted to investigate the potential function of SFRP2 and Slug in HSFB apoptosis. Cell apoptosis was detected using fluorescence-activated cell sorting and Caspase-3 activity was assayed by spectrophotometry. This study demonstrates that SFRP2 expression, as well as Slug, is dramatically up-regulated in HS relative to normal skin tissues, and the Slug expression is positively correlated with SFRP2. Slug expression was down-regulated in SFRP2-deficient cells, and the down-regulation of Slug expression increased sensitivity to apoptosis which was induced through a caspase-3-dependent pathway. The infected cells with reduced levels of Slug were tested for the expression of apoptosis-related genes (Bcl-2, Bax and PUMA) which were previously identified as Slug targets. Bcl-2 expression was down-regulated in Slug-deficient cells. In conclusion, SFRP2 appears to interact with Slug to affect the apoptosis of hypertrophic scar fibroblasts.
Collapse
|
29
|
Tominaga E, Yuasa K, Shimazaki S, Hijikata T. MicroRNA-1 targets Slug and endows lung cancer A549 cells with epithelial and anti-tumorigenic properties. Exp Cell Res 2012; 319:77-88. [PMID: 23142026 DOI: 10.1016/j.yexcr.2012.10.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 09/19/2012] [Accepted: 10/13/2012] [Indexed: 02/07/2023]
Abstract
MicroRNA-1 (miR-1) has recently been suggested to function as a tumor suppressor. Its functional relevance was assessed by exploring structural and tumorigenic properties of lung cancer A549 cells stably transduced with retrovirus containing pre-miR-1. A549 cells overexpressing miR-1 exhibited a significant morphological change from a mesenchymal to an epithelial phenotype characterized by cell polarization and intercellular junctions. The cells showed increased expression of E-cadherin, which colocalized with cortical actin filaments and vinculin to form typical adherens junction at the apical regions of intercellular borders. Additionally, they exhibited occludin-positive tight junctions at similar apical regions. Moreover, their migratory and invasive activities were inhibited, and their sensitivity to doxorubicin was increased slightly compared to control mock-infected cells. These structural and tumorigenic properties induced by miR-1 were associated with the reduced expression of Slug, which was a transcriptional repressor of E-cadherin or an inducer of epithelial-to-mesenchymal transition. Consistently, Slug was identified as a miR-1 target by bioinformatics and a luciferase reporter assay with plasmids containing luciferase-Slug 3'UTR. Collectively, the data presented here suggest that re-expression of miR-1 may be an effective therapy that prevents cancer malignancy by converting cells from a mesenchymal phenotype to an epithelial phenotype via the downregulation of Slug.
Collapse
Affiliation(s)
- Eiji Tominaga
- Department of Anatomy and Cell Biology, Faculty of Pharmacy, Musashino University, Nishitokyo-shi, Tokyo, Japan
| | | | | | | |
Collapse
|
30
|
Ferrari-Amorotti G, Fragliasso V, Esteki R, Prudente Z, Soliera AR, Cattelani S, Manzotti G, Grisendi G, Dominici M, Pieraccioli M, Raschellà G, Chiodoni C, Colombo MP, Calabretta B. Inhibiting interactions of lysine demethylase LSD1 with snail/slug blocks cancer cell invasion. Cancer Res 2012; 73:235-45. [PMID: 23054398 DOI: 10.1158/0008-5472.can-12-1739] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The process of epithelial-mesenchymal transition (EMT) which is required for cancer cell invasion is regulated by a family of E-box-binding transcription repressors, which include Snail (SNAIL1) and Slug (SNAI2). Snail appears to repress the expression of the EMT marker E-cadherin by epigenetic mechanisms dependent on the interaction of its N-terminal SNAG domain with chromatin-modifying proteins including lysine-specific demethylase 1 (LSD1/KDM1A). We assessed whether blocking Snail/Slug-LSD1 interaction by treatment with Parnate, an enzymatic inhibitor of LSD1, or TAT-SNAG, a cell-permeable peptide corresponding to the SNAG domain of Slug, suppresses the motility and invasiveness of cancer cells of different origin and genetic background. We show here that either treatment blocked Slug-dependent repression of the E-cadherin promoter and inhibited the motility and invasion of tumor cell lines without any effect on their proliferation. These effects correlated with induction of epithelial and repression of mesenchymal markers and were phenocopied by LSD1 or Slug downregulation. Parnate treatment also inhibited bone marrow homing/engraftment of Slug-expressing K562 cells. Together, these studies support the concept that targeting Snail/Slug-dependent transcription repression complexes may lead to the development of novel drugs selectively inhibiting the invasive potential of cancer cells.
Collapse
|
31
|
Lambertini E, Lolli A, Vezzali F, Penolazzi L, Gambari R, Piva R. Correlation between Slug transcription factor and miR-221 in MDA-MB-231 breast cancer cells. BMC Cancer 2012; 12:445. [PMID: 23031797 PMCID: PMC3534407 DOI: 10.1186/1471-2407-12-445] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 09/28/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Breast cancer and its metastatic progression is mainly directed by epithelial to mesenchymal transition (EMT), a phenomenon supported by specific transcription factors and miRNAs. METHODS In order to investigate a possible correlation between Slug transcription factor and miR-221, we performed Slug gene silencing in MDA-MB-231 breast cancer cells and evaluated the expression of genes involved in supporting the breast cancer phenotype, using qRT-PCR and Western blot analysis. Chromatin immunoprecipitation and wound healing assays were employed to determine a functional link between these two molecules. RESULTS We showed that Slug silencing significantly decreased the level of miR-221 and vimentin, reactivated Estrogen Receptor α and increased E-cadherin and TRPS1 expression. We demonstrated that miR-221 is a Slug target gene, and identified a specific region of miR-221 promoter that is transcriptionally active and binds the transcription factor Slug "in vivo". In addition, we showed that in Slug-silenced cells, wich retained residual miR-221 (about 38%), cell migration was strongly inhibited. Cell migration was inhibited, but to a less degree, following complete knockdown of miR-221 expression by transfection with antagomiR-221. CONCLUSIONS We report for the first time evidence of a correlation between Slug transcription factor and miR-221 in breast cancer cells. These studies suggest that miR-221 expression is, in part, dependent on Slug in breast cancer cells, and that Slug plays a more important role than miR-221 in cell migration and invasion.
Collapse
Affiliation(s)
- Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, 44121, Italy
| | | | | | | | | | | |
Collapse
|
32
|
Sánchez-Tilló E, Liu Y, de Barrios O, Siles L, Fanlo L, Cuatrecasas M, Darling DS, Dean DC, Castells A, Postigo A. EMT-activating transcription factors in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci 2012; 69:3429-56. [PMID: 22945800 PMCID: PMC11115078 DOI: 10.1007/s00018-012-1122-2] [Citation(s) in RCA: 385] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/20/2012] [Accepted: 08/09/2012] [Indexed: 12/13/2022]
Abstract
Cancer is a complex multistep process involving genetic and epigenetic changes that eventually result in the activation of oncogenic pathways and/or inactivation of tumor suppressor signals. During cancer progression, cancer cells acquire a number of hallmarks that promote tumor growth and invasion. A crucial mechanism by which carcinoma cells enhance their invasive capacity is the dissolution of intercellular adhesions and the acquisition of a more motile mesenchymal phenotype as part of an epithelial-to-mesenchymal transition (EMT). Although many transcription factors can trigger it, the full molecular reprogramming occurring during an EMT is mainly orchestrated by three major groups of transcription factors: the ZEB, Snail and Twist families. Upregulated expression of these EMT-activating transcription factors (EMT-ATFs) promotes tumor invasiveness in cell lines and xenograft mice models and has been associated with poor clinical prognosis in human cancers. Evidence accumulated in the last few years indicates that EMT-ATFs also regulate an expanding set of cancer cell capabilities beyond tumor invasion. Thus, EMT-ATFs have been shown to cooperate in oncogenic transformation, regulate cancer cell stemness, override safeguard programs against cancer like apoptosis and senescence, determine resistance to chemotherapy and promote tumor angiogenesis. This article reviews the expanding portfolio of functions played by EMT-ATFs in cancer progression.
Collapse
Affiliation(s)
- Ester Sánchez-Tilló
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
| | - Yongqing Liu
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- Department of Ophthalmology and Birth Defects Center, Louisville Health Science Center, Louisville, KY 40202 USA
| | - Oriol de Barrios
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
| | - Laura Siles
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
| | - Lucia Fanlo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- Master Program in Biomedical Research, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Miriam Cuatrecasas
- Department of Pathology, Hospital Clinic and IDIBAPS’ Tumor Bank, 08036 Barcelona, Spain
| | - Douglas S. Darling
- Department of Oral Health and Rehabilitation, Center for Genetics and Molecular Medicine, University of Louisville, Louisville, KY 40202 USA
| | - Douglas C. Dean
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- Department of Ophthalmology and Birth Defects Center, Louisville Health Science Center, Louisville, KY 40202 USA
| | - Antoni Castells
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
- Institute of Digestive and Metabolic Diseases, Hospital Clinic, 08036 Barcelona, Spain
| | - Antonio Postigo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- ICREA, 08010 Barcelona, Spain
| |
Collapse
|
33
|
EMT as the ultimate survival mechanism of cancer cells. Semin Cancer Biol 2012; 22:194-207. [DOI: 10.1016/j.semcancer.2012.02.013] [Citation(s) in RCA: 354] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Revised: 02/27/2012] [Accepted: 02/28/2012] [Indexed: 12/24/2022]
|
34
|
Chen D, Zheng XF, Yang ZY, Liu DX, Zhang GY, Jiao XL, Zhao H. S100A4 silencing blocks invasive ability of esophageal squamous cell carcinoma cells. World J Gastroenterol 2012; 18:915-22. [PMID: 22408350 PMCID: PMC3297050 DOI: 10.3748/wjg.v18.i9.915] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 11/16/2011] [Accepted: 11/23/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate a potential role of S100A4 in esophagus squamous cell carcinoma metastasis (ESCCs).
METHODS: Expression of S100A4 and E-cadherin were analyzed in frozen sections from ESCCs (metastasis, n = 28; non-metastasis, n = 20) by reverse transcription-polymerase chain reaction, quantitative polymerase chain reaction and immunohistochemistry. To explore the influence of S100A4 on esophageal cancer invasion and metastasis, S100A4 was overexpressed or silenced by S100A4 siRNA in TE-13 or Eca-109 cells in vitro and in vivo.
RESULTS: We found the mRNA and protein levels of S100A4 expression in ESCCs was significantly upregulated, and more importantly, that expression of S100A4 and E cadherin are strongly negatively correlated in patients who had metastasis. It was indicated that overexpression of S100A4 in TE-13 and Eca-109 cells downregulates the expression of E-cadherin, leading to increased cell migration in vitro, whereas knockdown of S100A4 inhibited cell migration and upregulation of E-cadherin expression. Moreover, the loss of cell metastatic potential was rescued by overexpression of E-cadherin completely. In addition, nude mice inoculated with S100A4 siRNA-transfected cells exhibited a significantly decreased invasion ability in vivo.
CONCLUSION: S100A4 may be involved in ESCC progression by regulate E-cadherin expression, vector-based RNA interference targeting S100A4 is a potential therapeutic method for human ESCC.
Collapse
|
35
|
Xia H, Cheung WKC, Ng SS, Jiang X, Jiang S, Sze J, Leung GKK, Lu G, Chan DTM, Bian XW, Kung HF, Poon WS, Lin MC. Loss of brain-enriched miR-124 microRNA enhances stem-like traits and invasiveness of glioma cells. J Biol Chem 2012; 287:9962-9971. [PMID: 22253443 DOI: 10.1074/jbc.m111.332627] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
miR-124 is a brain-enriched microRNA that plays a crucial role in neural development and has been shown to be down-regulated in glioma and medulloblastoma, suggesting its possible involvement in brain tumor progression. Here, we show that miR-124 is down-regulated in a panel of different grades of glioma tissues and in all of the human glioma cell lines we examined. By integrated bioinformatics analysis and experimental confirmation, we identified SNAI2, which is often up-regulated in glioma, as a direct functional target of miR-124. Because SNAI2 has been shown to regulate stem cell functions, we examined the roles of miR-124 and SNAI2 in glioma cell stem-like traits. The results showed that overexpression of miR-124 and knockdown of SNAI2 reduced neurosphere formation, CD133(+) cell subpopulation, and stem cell marker (BMI1, Nanog, and Nestin) expression, and these effects could be rescued by re-expression of SNAI2. Furthermore, enhanced miR-124 expression significantly inhibited glioma cell invasion in vitro. Finally, stable overexpression of miR-124 and knockdown of SNAI2 inhibited the tumorigenicity and invasion of glioma cells in vivo. These findings reveal, for the first time, that the tumor suppressor activity of miR-124 could be partly due to its inhibitory effects on glioma stem-like traits and invasiveness through SNAI2.
Collapse
Affiliation(s)
- Hongping Xia
- Brain Tumour Centre and Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong,; Department of Chemistry, The University of Hong Kong, Hong Kong,; Department of Surgery, Yijishan Hospital of Wannan Medical College, Sun Yat-Sen University, Guangzhou
| | | | - Samuel S Ng
- Department of Chemistry, The University of Hong Kong, Hong Kong
| | - Xiaochun Jiang
- Department of Surgery, Yijishan Hospital of Wannan Medical College, Sun Yat-Sen University, Guangzhou
| | - Songshan Jiang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-Sen University, Guangzhou
| | - Johnny Sze
- Brain Tumour Centre and Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong,; Department of Chemistry, The University of Hong Kong, Hong Kong
| | - Gilberto K K Leung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Gang Lu
- Brain Tumour Centre and Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Danny T M Chan
- Brain Tumour Centre and Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China, and
| | - Hsiang-Fu Kung
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China, and; Stanley Ho Centre for Emerging Infectious Diseases and State Key Laboratory in Oncology in South China and the School of Biomedical Sciences, Faculty of Medicine and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Brain Tumour Centre and Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong,.
| | - Marie C Lin
- Brain Tumour Centre and Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong,; Department of Chemistry, The University of Hong Kong, Hong Kong,; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China, and.
| |
Collapse
|
36
|
Array-based pharmacogenomics of molecular-targeted therapies in oncology. THE PHARMACOGENOMICS JOURNAL 2012; 12:185-96. [PMID: 22249357 DOI: 10.1038/tpj.2011.53] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The advent of microarrays over the past decade has transformed the way genome-wide studies are designed and conducted, leading to an unprecedented speed of acquisition and amount of new knowledge. Microarray data have led to the identification of molecular subclasses of solid tumors characterized by distinct oncogenic pathways, as well as the development of multigene prognostic or predictive models equivalent or superior to those of established clinical parameters. In the field of molecular-targeted therapy for cancer, in particular, the application of array-based methodologies has enabled the identification of molecular targets with 'key' roles in neoplastic transformation or tumor progression and the subsequent development of targeted agents, which are most likely to be active in the specific molecular setting. Herein, we present a summary of the main applications of whole-genome expression microarrays in the field of molecular-targeted therapies for solid tumors and we discuss their potential in the clinical setting. An emphasis is given on deciphering the molecular mechanisms of drug action, identifying novel therapeutic targets and suitable agents to target them with, and discovering molecular markers/signatures that predict response to therapy or optimal drug dose for each patient.
Collapse
|
37
|
Kim NH, Kim HS, Li XY, Lee I, Choi HS, Kang SE, Cha SY, Ryu JK, Yoon D, Fearon ER, Rowe RG, Lee S, Maher CA, Weiss SJ, Yook JI. A p53/miRNA-34 axis regulates Snail1-dependent cancer cell epithelial-mesenchymal transition. ACTA ACUST UNITED AC 2011; 195:417-33. [PMID: 22024162 PMCID: PMC3206336 DOI: 10.1083/jcb.201103097] [Citation(s) in RCA: 344] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Expression of the essential EMT inducer Snail1 is inhibited by miR-34 through a p53-dependent regulatory pathway. Snail1 is a zinc finger transcriptional repressor whose pathological expression has been linked to cancer cell epithelial–mesenchymal transition (EMT) programs and the induction of tissue-invasive activity, but pro-oncogenic events capable of regulating Snail1 activity remain largely uncharacterized. Herein, we demonstrate that p53 loss-of-function or mutation promotes cancer cell EMT by de-repressing Snail1 protein expression and activity. In the absence of wild-type p53 function, Snail1-dependent EMT is activated in colon, breast, and lung carcinoma cells as a consequence of a decrease in miRNA-34 levels, which suppress Snail1 activity by binding to highly conserved 3′ untranslated regions in Snail1 itself as well as those of key Snail1 regulatory molecules, including β-catenin, LEF1, and Axin2. Although p53 activity can impact cell cycle regulation, apoptosis, and DNA repair pathways, the EMT and invasion programs initiated by p53 loss of function or mutation are completely dependent on Snail1 expression. These results identify a new link between p53, miR-34, and Snail1 in the regulation of cancer cell EMT programs.
Collapse
Affiliation(s)
- Nam Hee Kim
- Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul 120-752, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chesler L, Weiss WA. Genetically engineered murine models--contribution to our understanding of the genetics, molecular pathology and therapeutic targeting of neuroblastoma. Semin Cancer Biol 2011; 21:245-55. [PMID: 21958944 PMCID: PMC3504935 DOI: 10.1016/j.semcancer.2011.09.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 09/14/2011] [Indexed: 01/01/2023]
Abstract
Genetically engineered mouse models (GEMM) have made major contributions to a molecular understanding of several adult cancers and these results are increasingly being translated into the pre-clinical setting where GEMM will very likely make a major impact on the development of targeted therapeutics in the near future. The relationship of pediatric cancers to altered developmental programs, and their genetic simplicity relative to adult cancers provides unique opportunities for the application of new advances in GEMM technology. In neuroblastoma the well-characterized TH-MYCN GEMM is increasingly used for a variety of molecular-genetic, developmental and pre-clinical therapeutics applications. We discuss: the present and historical application of GEMM to neuroblastoma research, future opportunities, and relevant targets suitable for new GEMM strategies in neuroblastoma. We review the potential of these models to contribute both to an understanding of the developmental nature of neuroblastoma and to improved therapy for this disease.
Collapse
Affiliation(s)
- Louis Chesler
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research & The Royal Marsden NHS Trust, Sutton, Surrey SM2 5NG, United Kingdom.
| | | |
Collapse
|
39
|
Abstract
Lung cancer is the leading cause of cancer death worldwide. Cancer metastasis and resistance to treatment (including radiotherapy, chemotherapy and targeted therapy) are two major causes for the poor survival of lung cancer patients. Epithelial-mesenchymal transition (EMT) is involved in cancer cell invasion, resistance to apoptosis and stem cell features. The process of EMT is controlled by a group of transcriptional factors, zinc finger proteins and basic helix-loop-helix factors. Signaling pathways activated by intrinsic or extrinsic stimuli converge on these transcriptional factors and regulated the phenotypic changes of cancer cells. These EMT regulators may play an important role in cancer progression. In lung cancer, Slug is the most thoroughly investigated EMT regulator. The expression of Slug is associated with lung cancer invasion and resistance to target therapy. In this review, we focus on the current understanding of the role of Slug in the carcinogenesis and progression of lung cancer.
Collapse
Affiliation(s)
- Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Chung-Shan South Road, Taipei 100, Taiwan
| | | |
Collapse
|
40
|
Jiang M, Stanke J, Lahti JM. The connections between neural crest development and neuroblastoma. Curr Top Dev Biol 2011; 94:77-127. [PMID: 21295685 DOI: 10.1016/b978-0-12-380916-2.00004-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neuroblastoma (NB), the most common extracranial solid tumor in childhood, is an extremely heterogeneous disease both biologically and clinically. Although significant progress has been made in identifying molecular and genetic markers for NB, this disease remains an enigmatic challenge. Since NB is thought to be an embryonal tumor that is derived from precursor cells of the peripheral (sympathetic) nervous system, understanding the development of normal sympathetic nervous system may highlight abnormal events that contribute to NB initiation. Therefore, this review focuses on the development of the peripheral trunk neural crest, the current understanding of how developmental factors may contribute to NB and on recent advances in the identification of important genetic lesions and signaling pathways involved in NB tumorigenesis and metastasis. Finally, we discuss how future advances in identification of molecular alterations in NB may lead to more effective, less toxic therapies, and improve the prognosis for NB patients.
Collapse
Affiliation(s)
- Manrong Jiang
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | |
Collapse
|
41
|
Tang Y, Liang X, Zhu G, Zheng M, Yang J, Chen Y. Expression and importance of zinc-finger transcription factor Slug in adenoid cystic carcinoma of salivary gland. J Oral Pathol Med 2011; 39:775-80. [PMID: 20738754 DOI: 10.1111/j.1600-0714.2010.00913.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND It has been reported that zinc-finger transcription factor Slug plays a critical role in tumor proliferation and differentiation, relapse, invasion, metastasis, and decreased survival. However, there is little information on the expression and role of Slug in salivary adenoid cystic carcinoma (ACC). METHOD Demographic variables and primary tumor site, dates of diagnoses, perineural invasion, local regional recurrence, and distant metastasis of 121 cases of salivary ACC treated at the Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University between 1996 and 2005 were retrieved. The Slug expression of all these cases was examined by immunohistochemical methods. The association between Slug expression and clinicopathological variables was analyzed using Chi-squared test, and the prognostic factors were examined by univariate and multivariate analyses. RESULTS Our data demonstrated that the positive expression of Slug was observed in 71.90% (87/121) of cases. Slug expression was significantly associated with tumor site, TNM stage, histological pattern, perineural invasion, local regional recurrence and distant metastasis of patients with ACC (P<0.05). And Slug expression, local regional recurrence and distant metastasis were independent and significant prognostic factors in all patients. CONCLUSIONS It is proposed that Slug may play an important role in the invasion and metastasis of ACC, and that Slug had the potential for one of the strong invasion and metastasis indicators at presentation of ACC patients.
Collapse
Affiliation(s)
- Yaling Tang
- State Key Laboratory of Oral Diseases, Sichuan University, No. 14 Sec. 3 Renminnan Road, Chengdu, Sichuan, China
| | | | | | | | | | | |
Collapse
|
42
|
Franco DL, Mainez J, Vega S, Sancho P, Murillo MM, de Frutos CA, Del Castillo G, López-Blau C, Fabregat I, Nieto MA. Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes. J Cell Sci 2011; 123:3467-77. [PMID: 20930141 DOI: 10.1242/jcs.068692] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although TGF-β suppresses early stages of tumour development, it later contributes to tumour progression when cells become resistant to its suppressive effects. In addition to circumventing TGF-β-induced growth arrest and apoptosis, malignant tumour cells become capable of undergoing epithelial-to-mesenchymal transition (EMT), favouring invasion and metastasis. Therefore, defining the mechanisms that allow cancer cells to escape from the suppressive effects of TGF-β is fundamental to understand tumour progression and to design specific therapies. Here, we have examined the role of Snail1 as a suppressor of TGF-β-induced apoptosis in murine non-transformed hepatocytes, rat and human hepatocarcinoma cell lines and transgenic mice. We show that Snail1 confers resistance to TGF-β-induced cell death and that it is sufficient to induce EMT in adult hepatocytes, cells otherwise refractory to this transition upon exposure to TGF-β. Furthermore, we show that Snail1 silencing prevents EMT and restores the cell death response induced by TGF-β. As Snail1 is a known target of TGF-β signalling, our data indicate that Snail1 might transduce the tumour-promoting effects of TGF-β, namely the EMT concomitant with the resistance to cell death.
Collapse
Affiliation(s)
- D Lorena Franco
- Instituto de Neurociencias (CSIC-UMH), 03550 San Juan de Alicante, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
RNA interference targeting slug increases cholangiocarcinoma cell sensitivity to cisplatin via upregulating PUMA. Int J Mol Sci 2011; 12:385-400. [PMID: 21339993 PMCID: PMC3039959 DOI: 10.3390/ijms12010385] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 01/06/2011] [Accepted: 01/07/2011] [Indexed: 11/17/2022] Open
Abstract
Slug is an E-cadherin repressor and a suppressor of PUMA (p53 upregulated modulator of apoptosis) and it has recently been demonstrated that Slug plays an important role in controlling apoptosis. In this study, we examined whether Slug's ability to silence expression suppresses the growth of cholangiocarcinoma cells and/or sensitizes cholangiocarcinoma cells to chemotherapeutic agents through induction of apoptosis. We targeted the Slug gene using siRNA (Slug siRNA) via full Slug cDNA plasmid (Slug cDNA) transfection of cholangiocarcinoma cells. Slug siRNA, cisplatin, or Slug siRNA in combination with cisplatin, were used to treat cholangiocarcinoma cells in vitro. Western blot was used to detect the expression of Slug, PUMA, and E-cadherin protein. TUNEL, Annexin V Staining, and cell cycle analysis were used to detect apoptosis. A nude mice subcutaneous xenograft model of QBC939 cells was used to assess the effect of Slug silencing and/or cisplatin on tumor growth. Immunohistochemical staining was used to analyze the expression of Slug and PUMA. TUNEL was used to detect apoptosis in vivo. The results showed that PUMA and E-cadherin expression in cholangiocarcinoma cells is Slug dependent. We demonstrated that Slug silencing and cisplatin both promote apoptosis by upregulation of PUMA, not by upregulation of E-cadherin. Slug silencing significantly sensitized cholangiocarcinoma cells to cisplatin through upregulation of PUMA. Finally, we showed that Slug silencing suppressed the growth of QBC939 xenograft tumors and sensitized the tumor cells to cisplatin through PUMA upregulation and induction of apoptosis. Our findings indicate that Slug is an important modulator of the therapeutic response of cholangiocarcinoma cells and is potentially useful as a sensitizer in cholangiocarcinoma therapy. One of the mechanisms is the regulation of PUMA by Slug.
Collapse
|
44
|
Wang X, Zhang K, Sun L, Liu J, Lu H. Short interfering RNA directed against Slug blocks tumor growth, metastasis formation, and vascular leakage in bladder cancer. Med Oncol 2010; 28 Suppl 1:S413-22. [PMID: 21053100 DOI: 10.1007/s12032-010-9728-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 10/14/2010] [Indexed: 01/23/2023]
Abstract
In a previous study, we have identified Slug as selectively overexpressed in the highly invasive bladder cancer cells. Furthermore, Slug overexpression was correlated with metastasis in bladder cancer tissues. Here, we investigated the role and underlying mechanism of Slug in the process of growth, invasion and metastasis formation, and vascular leakage in a bladder cancer cell line T24. We knocked down in a bladder cancer cell line T24 by Slug siRNA, and the effects on invasion, metastasis, and proliferation were evaluated in vitro. A pseudometastatic model of bladder cancer in severe combined immunodeficient mice was used to assess the effects of Slug silencing on metastasis and angiogenesis development. Slug-specific siRNA in T24 cells led to a significant decrease in invasiveness and metastasis, proliferation rates, and angiogenesis in vitro. Slug-specific siRNA induced strong changes in cell invasiveness mainly through a mechanism of up-regulation of epithelial markers E-cadherin expression. Interestingly, E-cadherin-specific siRNA attenuated Slug siRNA-induced Matrigel invasion. Moreover, Slug-specific siRNA induced strong changes in microvessel counts in angiogenesis mainly through decreased activity of MMP-2, but not through cadherin expression and decreased activity of MMP-9. Restoration of MMP-2 expression in the Slug-silenced T24 cells resulted in an increased cell angiogenesis. Moreover, Slug-specific siRNA significantly reduced tumor growth by approximately 60% and inhibited metastasis and angiogenesis in a xenograft mouse model. This was accompanied by an increased expression of E-cadherin and a decreased expression of MMP-2 in tumor tissues. These data suggest that Slug regulates tumor growth, invasion and metastasis, and angiogenesis through E-cadherrin and MMPS passway. Blocking Slug signaling in bladder cancer may represent a novel strategy in the future to reduce metastatic disease burden in bladder cancer patients.
Collapse
Affiliation(s)
- Xinsheng Wang
- Department of Urology Surgery, The Affiliated Hospital of Medical College, QingDao University, No 16 Jiangsu Road, QingDao, Shan Dong Province 266003, People's Republic of China
| | | | | | | | | |
Collapse
|
45
|
Chang TH, Tsai MF, Su KY, Wu SG, Huang CP, Yu SL, Yu YL, Lan CC, Yang CH, Lin SB, Wu CP, Shih JY, Yang PC. Slug confers resistance to the epidermal growth factor receptor tyrosine kinase inhibitor. Am J Respir Crit Care Med 2010; 183:1071-9. [PMID: 21037017 DOI: 10.1164/rccm.201009-1440oc] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
RATIONALE Non-small cell lung cancers carrying epidermal growth factor receptor (EGFR) mutations respond well to EGFR tyrosine kinase inhibitors (TKIs), but patients ultimately develop drug resistance and relapse. Although epithelial-mesenchymal transition (EMT) can predict resistance to EGFR TKIs, the molecular mechanisms are still unknown. OBJECTIVES To examine the role of EMT regulators in resistance to gefitinib. METHODS The expression level of EMT regulators in gefitinib-sensitive cells (PC9) and gefitinib-resistant cells (PC9/gef) was determined using quantitative real-time reverse transcription-polymerase chain reaction and Western blot analysis. Molecular manipulations (silencing or overexpression) were performed to investigate the effects of EMT regulators on gefitinib resistance in vitro, and a xenograft mouse model was used for in vivo confirmation. In addition, cancer cells from 44 patients with malignant pleural effusions of lung adenocarcinoma were collected for analysis of EMT regulator mRNA by quantitative real-time reverse transcription-polymerase chain reaction. MEASUREMENTS AND MAIN RESULTS Slug expression, but not that of snail, twist, or zeb-1, was significantly increased in PC9/gef compared with PC9 cells. Slug knockdown in PC9/gef cells reversed resistance to gefitinib, and overexpression of Slug in PC9 cells protected cells from gefitinib-induced apoptosis. Silencing of Slug in gefitinib-resistant cells restored gefitinib-induced apoptosis primarily through Bim up-regulation and activation of caspase-9. Slug enhanced tumor growth in a xenograft mouse model, even with gefitinib treatment. In clinical samples, Slug expression was significantly higher in cancer cells with resistance to EGFR TKIs than in treatment-naive cancer cells. CONCLUSIONS Slug contributes to the resistance to gefitinib and may be a potential therapeutic target for treating resistance to EGFR TKIs.
Collapse
Affiliation(s)
- Tzu-Hua Chang
- Graduate Institute of Medical Sciences, National Defense Medical Center, #161 Sec. 6 Minquan E. Rd., Neihu Dist., Taipei City, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proc Natl Acad Sci U S A 2010; 107:15449-54. [PMID: 20713713 DOI: 10.1073/pnas.1004900107] [Citation(s) in RCA: 797] [Impact Index Per Article: 56.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) produces cancer cells that are invasive, migratory, and exhibit stem cell characteristics, hallmarks of cells that have the potential to generate metastases. Inducers of the EMT include several transcription factors (TFs), such as Goosecoid, Snail, and Twist, as well as the secreted TGF-beta1. Each of these factors is capable, on its own, of inducing an EMT in the human mammary epithelial (HMLE) cell line. However, the interactions between these regulators are poorly understood. Overexpression of each of the above EMT inducers up-regulates a subset of other EMT-inducing TFs, with Twist, Zeb1, Zeb2, TGF-beta1, and FOXC2 being commonly induced. Up-regulation of Slug and FOXC2 by either Snail or Twist does not depend on TGF-beta1 signaling. Gene expression signatures (GESs) derived by overexpressing EMT-inducing TFs reveal that the Twist GES and Snail GES are the most similar, although the Goosecoid GES is the least similar to the others. An EMT core signature was derived from the changes in gene expression shared by up-regulation of Gsc, Snail, Twist, and TGF-beta1 and by down-regulation of E-cadherin, loss of which can also trigger an EMT in certain cell types. The EMT core signature associates closely with the claudin-low and metaplastic breast cancer subtypes and correlates negatively with pathological complete response. Additionally, the expression level of FOXC1, another EMT inducer, correlates strongly with poor survival of breast cancer patients.
Collapse
|
47
|
Yang J, Eddy JA, Pan Y, Hategan A, Tabus I, Wang Y, Cogdell D, Price ND, Pollock RE, Lazar AJF, Hunt KK, Trent JC, Zhang W. Integrated proteomics and genomics analysis reveals a novel mesenchymal to epithelial reverting transition in leiomyosarcoma through regulation of slug. Mol Cell Proteomics 2010; 9:2405-13. [PMID: 20651304 DOI: 10.1074/mcp.m110.000240] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Leiomyosarcoma is one of the most common mesenchymal tumors. Proteomics profiling analysis by reverse-phase protein lysate array surprisingly revealed that expression of the epithelial marker E-cadherin (encoded by CDH1) was significantly elevated in a subset of leiomyosarcomas. In contrast, E-cadherin was rarely expressed in the gastrointestinal stromal tumors, another major mesenchymal tumor type. We further sought to 1) validate this finding, 2) determine whether there is a mesenchymal to epithelial reverting transition (MErT) in leiomyosarcoma, and if so 3) elucidate the regulatory mechanism responsible for this MErT. Our data showed that the epithelial cell markers E-cadherin, epithelial membrane antigen, cytokeratin AE1/AE3, and pan-cytokeratin were often detected immunohistochemically in leiomyosarcoma tumor cells on tissue microarray. Interestingly, the E-cadherin protein expression was correlated with better survival in leiomyosarcoma patients. Whole genome microarray was used for transcriptomics analysis, and the epithelial gene expression signature was also associated with better survival. Bioinformatics analysis of transcriptome data showed an inverse correlation between E-cadherin and E-cadherin repressor Slug (SNAI2) expression in leiomyosarcoma, and this inverse correlation was validated on tissue microarray by immunohistochemical staining of E-cadherin and Slug. Knockdown of Slug expression in SK-LMS-1 leiomyosarcoma cells by siRNA significantly increased E-cadherin; decreased the mesenchymal markers vimentin and N-cadherin (encoded by CDH2); and significantly decreased cell proliferation, invasion, and migration. An increase in Slug expression by pCMV6-XL5-Slug transfection decreased E-cadherin and increased vimentin and N-cadherin. Thus, MErT, which is mediated through regulation of Slug, is a clinically significant phenotype in leiomyosarcoma.
Collapse
Affiliation(s)
- Jilong Yang
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tanno B, Sesti F, Cesi V, Bossi G, Ferrari-Amorotti G, Bussolari R, Tirindelli D, Calabretta B, Raschellà G. Expression of Slug is regulated by c-Myb and is required for invasion and bone marrow homing of cancer cells of different origin. J Biol Chem 2010; 285:29434-45. [PMID: 20622260 DOI: 10.1074/jbc.m109.089045] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In metastatic cancer cells, the process of invasion is regulated by several transcription factors that induce changes required for migration and resistance to apoptosis. Slug (SNAI2, Snail2) is involved in epithelial mesenchymal transition in physiological and in pathological contexts. We show here that in embryonic kidney, colon carcinoma, chronic myeloid leukemia-blast crisis, and in neuroblastoma cells, expression of Slug is transcriptionally regulated by c-Myb via Myb binding sites in the 5'-flanking region and in the first intron of the slug gene. In embryonic kidney and neuroblastoma cells, c-Myb induced vimentin, fibronectin, and N-cadherin expression and membrane ruffling via actin polymerization consistent with the acquisition of a mesenchymal-like phenotype. Furthermore, down-regulation of endogenous c-Myb levels in colon carcinoma cells led to increased expression of E-cadherin and reduced levels of vimentin. Some of these changes are predominantly Slug-dependent as Slug silencing via RNA interference (RNAi) reverts the cells to a quasi-parental condition. Changes in gene expression and morphology induced by c-Myb-activated Slug correlated with increased ability to migrate (embryonic kidney) and to invade through a Matrigel membrane (embryonic kidney, colon carcinoma, neuroblastoma). c-Myb-dependent Slug expression was also essential for the homing of chronic myeloid leukemia K562 cells to the bone marrow. In summary, we show here that the proto-oncogene c-Myb controls Slug transcription in tumor cells of different origin. Such a regulatory pathway contributes to the acquisition of invasive properties that are important for the metastatic process.
Collapse
Affiliation(s)
- Barbara Tanno
- Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), Research Center Casaccia, Laboratory of Radiation Biology and Biomedicine, 00123 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cha HS, Bae EK, Ahn JK, Lee J, Ahn KS, Koh EM. Slug suppression induces apoptosis via Puma transactivation in rheumatoid arthritis fibroblast-like synoviocytes treated with hydrogen peroxide. Exp Mol Med 2010; 42:428-36. [PMID: 20418652 PMCID: PMC2892596 DOI: 10.3858/emm.2010.42.6.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2010] [Indexed: 11/04/2022] Open
Abstract
Inadequate apoptosis contributes to synovial hyperplasia in rheumatoid arthritis (RA). Recent study shows that low expression of Puma might be partially responsible for the decreased apoptosis of fibroblast-like synoviocytes (FLS). Slug, a highly conserved zinc finger transcriptional repressor, is known to antagonize apoptosis of hematopoietic progenitor cells by repressing Puma transactivation. In this study, we examined the expression and function of Slug in RA FLS. Slug mRNA expression was measured in the synovial tissue (ST) and FLS obtained from RA and osteoarthritis patients. Slug and Puma mRNA expression in FLS by apoptotic stimuli were measured by real-time PCR analysis. FLS were transfected with control siRNA or Slug siRNA. Apoptosis was quantified by trypan blue exclusion, DNA fragmentation and caspase-3 assay. RA ST expressed higher level of Slug mRNA compared with osteoarthritis ST. Slug was significantly induced by hydrogen peroxide (H2O2) but not by exogenous p53 in RA FLS. Puma induction by H2O2 stimulation was significantly higher in Slug siRNA-transfected FLS compared with control siRNA-transfected FLS. After H2O2 stimulation, viable cell number was significantly lower in Slug siRNA-transfected FLS compared with control siRNA-transfected FLS. Apoptosis enhancing effect of Slug siRNA was further confirmed by ELISA that detects cytoplasmic histone-associated DNA fragments and caspase-3 assay. These data demonstrate that Slug is overexpressed in RA ST and that suppression of Slug gene facilitates apoptosis of FLS by increasing Puma transactivation. Slug may therefore represent a potential therapeutic target in RA.
Collapse
Affiliation(s)
- Hoon-Suk Cha
- Department of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | | | | | | | | | | |
Collapse
|
50
|
Wang Y, Ngo VN, Marani M, Yang Y, Wright G, Staudt LM, Downward J. Critical role for transcriptional repressor Snail2 in transformation by oncogenic RAS in colorectal carcinoma cells. Oncogene 2010; 29:4658-70. [PMID: 20562906 DOI: 10.1038/onc.2010.218] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Activating mutations in the KRAS gene are among the most prevalent genetic changes in human cancers. To identify synthetic lethal interactions in cancer cells harbouring mutant KRAS, we performed a large-scale screen in isogenic paired colon cancer cell lines that differ by a single allele of mutant KRAS using an inducible short hairpin RNA interference library. Snail2, a zinc finger transcriptional repressor encoded by the SNAI2 gene, was found to be selectively required for the long-term survival of cancer cells with mutant KRAS that have undergone epithelial-mesenchymal transition (EMT), a transdifferentiation event that is frequently seen in advanced tumours and is promoted by RAS activation. Snail2 expression is regulated by the RAS pathway and is required for EMT. Our findings support Snail2 as a possible target for the treatment of the broad spectrum of human cancers of epithelial origin with mutant RAS that have undergone EMT and are characterized by a high degree of chemoresistance and radioresistance.
Collapse
Affiliation(s)
- Y Wang
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, London, UK
| | | | | | | | | | | | | |
Collapse
|