1
|
Wang X, Xue X, Pang M, Yu L, Qian J, Li X, Tian M, Lyu A, Lu C, Liu Y. Epithelial-mesenchymal plasticity in cancer: signaling pathways and therapeutic targets. MedComm (Beijing) 2024; 5:e659. [PMID: 39092293 PMCID: PMC11292400 DOI: 10.1002/mco2.659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Currently, cancer is still a leading cause of human death globally. Tumor deterioration comprises multiple events including metastasis, therapeutic resistance and immune evasion, all of which are tightly related to the phenotypic plasticity especially epithelial-mesenchymal plasticity (EMP). Tumor cells with EMP are manifest in three states as epithelial-mesenchymal transition (EMT), partial EMT, and mesenchymal-epithelial transition, which orchestrate the phenotypic switch and heterogeneity of tumor cells via transcriptional regulation and a series of signaling pathways, including transforming growth factor-β, Wnt/β-catenin, and Notch. However, due to the complicated nature of EMP, the diverse process of EMP is still not fully understood. In this review, we systematically conclude the biological background, regulating mechanisms of EMP as well as the role of EMP in therapy response. We also summarize a range of small molecule inhibitors, immune-related therapeutic approaches, and combination therapies that have been developed to target EMP for the outstanding role of EMP-driven tumor deterioration. Additionally, we explore the potential technique for EMP-based tumor mechanistic investigation and therapeutic research, which may burst vigorous prospects. Overall, we elucidate the multifaceted aspects of EMP in tumor progression and suggest a promising direction of cancer treatment based on targeting EMP.
Collapse
Affiliation(s)
- Xiangpeng Wang
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoxia Xue
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Mingshi Pang
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Liuchunyang Yu
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Jinxiu Qian
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoyu Li
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Meng Tian
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Aiping Lyu
- School of Chinese MedicineHong Kong Baptist UniversityKowloonHong KongChina
| | - Cheng Lu
- Institute of Basic Research in Clinical MedicineChina Academy of Chinese Medical SciencesBeijingChina
| | - Yuanyan Liu
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
2
|
Wu J, Li W, Su J, Zheng J, Liang Y, Lin J, Xu B, Liu Y. Integration of single-cell sequencing and bulk RNA-seq to identify and develop a prognostic signature related to colorectal cancer stem cells. Sci Rep 2024; 14:12270. [PMID: 38806611 PMCID: PMC11133358 DOI: 10.1038/s41598-024-62913-3] [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: 01/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
The prognosis for patients with colorectal cancer (CRC) remains worse than expected due to metastasis, recurrence, and resistance to chemotherapy. Colorectal cancer stem cells (CRCSCs) play a vital role in tumor metastasis, recurrence, and chemotherapy resistance. However, there are currently no prognostic markers based on CRCSCs-related genes available for clinical use. In this study, single-cell transcriptome sequencing was employed to distinguish cancer stem cells (CSCs) in the CRC microenvironment and analyze their properties at the single-cell level. Subsequently, data from TCGA and GEO databases were utilized to develop a prognostic risk model for CRCSCs-related genes and validate its diagnostic performance. Additionally, functional enrichment, immune response, and chemotherapeutic drug sensitivity of the relevant genes in the risk model were investigated. Lastly, the key gene RPS17 in the risk model was identified as a potential prognostic marker and therapeutic target for further comprehensive studies. Our findings provide new insights into the prognostic treatment of CRC and offer novel perspectives for a systematic and comprehensive understanding of CRC development.
Collapse
Affiliation(s)
- Jiale Wu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Wanyu Li
- Well Lead Medical Co., Ltd., Guangzhou, 511434, Guangdong, China
| | - Junyu Su
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jiamin Zheng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yanwen Liang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jiansuo Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Bilian Xu
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Yi Liu
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| |
Collapse
|
3
|
Bawadud RS, Alkhatib MH. Growth and invasion inhibition of T47D ductal carcinoma cells by the association of docetaxel with a bioactive agent in neutral nanosuspension. BIOIMPACTS : BI 2023; 13:145-157. [PMID: 37193079 PMCID: PMC10182446 DOI: 10.34172/bi.2023.23515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 08/16/2022] [Accepted: 09/13/2022] [Indexed: 05/18/2023]
Abstract
Introduction: The approach for drug delivery has impressively developed with the emergence of nanosuspension, particularly the targeted nanoemulsions (NEs). It can potentially improve the bioavailability of drugs, enhancing their therapeutic efficiency. This study aims to examine the potential role of NE as a delivery system for the combination of docetaxel (DTX), a microtubule-targeting agent, and thymoquinone (TQ) in the treatment of human ductal carcinoma cells T47D. Methods: NEs were synthesized by ultra-sonication and characterized physically by dynamic light scattering (DLS). A sulforhodamine B assay was performed to evaluate cytotoxicity, and a flow cytometry analysis for cell cycle, apoptosis, autophagy, and cancer stem cell evaluations. A quantitative polymerase chain reaction further assessed the epithelial-mesenchymal transition gene expirations of SNAIL-1, ZEB-1, and TWIST-1. Results: The optimal sizes of blank-NEs and NE-DTX+TQ were found at 117.3 ± 8 nm and 373 ± 6.8 nm, respectively. The synergistic effect of the NE-DTX+TQ formulation significantly inhibited the in vitro proliferation of T47D cells. It caused a significant increase in apoptosis, accompanied by the stimulation of autophagy. Moreover, this formulation arrested T47D cells at the G2/M phase, promoted the reduction of the breast cancer stem cell (BCSC) population, and repressed the expression of TWIST-1 and ZEB-1. Conclusion: Co-delivery of NE-DTX+TQ may probably inhibit the proliferation of T47D via the induction of apoptosis and autophagy pathways and impede the migration by reducing the BCSC population and downregulating TWIST-1 expression to decrease the epithelial-to-mesenchymal transition (EMT) of breast cancer cells. Therefore, the study suggests the NE-DTX+TQ formula as a potential approach to inhibit breast cancer growth and metastasis.
Collapse
Affiliation(s)
- Raghdah S. Bawadud
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mayson H. Alkhatib
- Department of Biological Sciences & Chemistry, College of Arts and Sciences, University of Nizwa, Nizwa, Sultanate of Oman
| |
Collapse
|
4
|
Ding P, Wu J, Wu H, Sun C, Guo H, Lowe S, Yang P, Tian Y, Liu Y, Meng L, Zhao Q. Inflammation and nutritional status indicators as prognostic indicators for patients with locally advanced gastrointestinal stromal tumors treated with neoadjuvant imatinib. BMC Gastroenterol 2023; 23:23. [PMID: 36690935 PMCID: PMC9869595 DOI: 10.1186/s12876-023-02658-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Previous studies have confirmed that preoperative nutritional-inflammatory indicators can predict prognosis in various malignancies. However, to the best of our knowledge, no study has investigated the assessment of systemic inflammatory immunity index (SII) combined with prognostic nutritional index (PNI) scores to predict prognosis after neoadjuvant treatment with imatinib in locally advanced gastrointestinal stromal tumours (LA-GIST). The aim of this study was to evaluate the predictive value of pretreatment SII-PNI scores in predicting recurrence after neoadjuvant therapy with imatinib in patients with LA-GIST. METHODS We retrospectively analyzed 57 patients with LA-GIST who received imatinib neoadjuvant from January 2013 to March 2019. Patients were divided into recurrence and non-recurrence groups according to their follow-up status, and SII and PNI cut-offs were calculated by receiver operating characteristic. The SII-PNI score ranged from 0 to 2 and were categorized into the following: score of 2, high SII (≥ 544.6) and low PNI (≤ 47.2); score of 1, either high SII (≥ 544.6) or low PNI (≤ 47.2); score of 0, no high SII (≥ 544.6) nor low PNI (≤ 47.2). RESULTS All patients received imatinib neoadjuvant therapy for a median treatment period of 8.5 months (ranging from 3.2 to 12.6 months), with 8 patients (14.04%) and 49 patients (85.96%) developing recurrence and non-recurrence, respectively. Patients with a high SII-PNI score had a significantly worse recurrence-free survival time than those with a low SII-PNI score (P = 0.022, 0.046), and had a poorer pathological response (P = 0.014). Multivariate analysis demonstrated that the SII-PNI score was an independent prognostic factor for prediction of recurrence-free survival (P = 0.002). CONCLUSION The pre-treatment SII-PNI score can be used to predict the efficacy after neoadjuvant treatment with imatinib in patients with LA-GIST, which may be a promising predictor of recurrence-free survival time for patients.
Collapse
Affiliation(s)
- Ping’an Ding
- grid.452582.cThe Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei China ,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China
| | - Jiaxiang Wu
- grid.452582.cThe Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei China ,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China
| | - Haotian Wu
- grid.452582.cThe Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei China ,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China
| | - Chenyu Sun
- grid.488798.20000 0004 7535 783XAMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL 60657 USA
| | - Honghai Guo
- grid.452582.cThe Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei China ,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China
| | - Scott Lowe
- grid.258405.e0000 0004 0539 5056College of Osteopathic Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106 USA
| | - Peigang Yang
- grid.452582.cThe Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei China ,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China
| | - Yuan Tian
- grid.452582.cThe Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei China ,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China
| | - Yang Liu
- grid.452582.cThe Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei China ,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China
| | - Lingjiao Meng
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China ,grid.452582.cResearch Center of the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 China
| | - Qun Zhao
- grid.452582.cThe Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011 Hebei China ,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011 China
| |
Collapse
|
5
|
Poonaki E, Kahlert UD, Meuth SG, Gorji A. The role of the ZEB1–neuroinflammation axis in CNS disorders. J Neuroinflammation 2022; 19:275. [PMCID: PMC9675144 DOI: 10.1186/s12974-022-02636-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/31/2022] [Indexed: 11/21/2022] Open
Abstract
Zinc finger E-box binding homeobox 1 (ZEB1) is a master modulator of the epithelial–mesenchymal transition (EMT), a process whereby epithelial cells undergo a series of molecular changes and express certain characteristics of mesenchymal cells. ZEB1, in association with other EMT transcription factors, promotes neuroinflammation through changes in the production of inflammatory mediators, the morphology and function of immune cells, and multiple signaling pathways that mediate the inflammatory response. The ZEB1–neuroinflammation axis plays a pivotal role in the pathogenesis of different CNS disorders, such as brain tumors, multiple sclerosis, cerebrovascular diseases, and neuropathic pain, by promoting tumor cell proliferation and invasiveness, formation of the hostile inflammatory micromilieu surrounding neuronal tissues, dysfunction of microglia and astrocytes, impairment of angiogenesis, and dysfunction of the blood–brain barrier. Future studies are needed to elucidate whether the ZEB1–neuroinflammation axis could serve as a diagnostic, prognostic, and/or therapeutic target for CNS disorders.
Collapse
Affiliation(s)
- Elham Poonaki
- grid.411327.20000 0001 2176 9917Department of Neurology, Faculty of Medicine, Heinrich-Heine-University, Düsseldorf, Germany ,grid.5949.10000 0001 2172 9288Epilepsy Research Center, Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Domagkstr. 11, 48149 Münster, Germany
| | - Ulf Dietrich Kahlert
- grid.5807.a0000 0001 1018 4307Molecular and Experimental Surgery, Faculty of Medicine, University Clinic for General-, Visceral-, Vascular- and Transplantation Surgery, Otto-Von-Guericke-University, Magdeburg, Germany
| | - Sven G. Meuth
- grid.411327.20000 0001 2176 9917Department of Neurology, Faculty of Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ali Gorji
- grid.5949.10000 0001 2172 9288Epilepsy Research Center, Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Domagkstr. 11, 48149 Münster, Germany ,grid.512981.60000 0004 0612 1380Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran ,grid.411583.a0000 0001 2198 6209Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Khazravi M, Shirkhoda M, Saffar H, Jalaeefar A. Prognostic Value of Lymph Node Density Compared to Pre-operative Platelet-to-Lymphocyte Ratio and Neutrophil-to-Lymphocyte Ratio in Patients with Tongue Squamous Cell Carcinoma. J Maxillofac Oral Surg 2022; 21:845-855. [PMID: 36274869 PMCID: PMC9474769 DOI: 10.1007/s12663-021-01524-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/03/2021] [Indexed: 10/22/2022] Open
Abstract
Background We aimed to investigate the prognostic significance of lymph node density (LND), and pre-operative neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) for outcome measuring in tongue squamous cell carcinoma (TSCC)' patients. Methods A total of 129 patients who underwent surgery for TSCC were enrolled in this retrospective study. LND and pre-operative NLR and PLR were used as outcome measures and their correlations with different clinicopathological features were examined. Results The cutoff values for NLR, PLR, and LND were obtained 1.21, 97.81, and 0.02, respectively, by receiver operating characteristic (ROC) curve approach. Only LND was found to be significantly associated with decreased overall survival (HR = 4.24; 95% CI 1.49-12.10; P = 0.007) and disease-free survival (HR = 3.48; 95% CI 1.43-8.45; P = 0.006) both in univariate and multivariate analyzes. Conclusion Based on the findings, the LND has superiority over pre-operative NLR and PLR in predicting outcomes for the patients with TSCC.
Collapse
Affiliation(s)
- Mona Khazravi
- Department of Surgical Oncology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Research Center of Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shirkhoda
- Department of Surgical Oncology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hana Saffar
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirmohsen Jalaeefar
- Department of Surgical Oncology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Research Center of Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
EMT and Inflammation: Crossroads in HCC. J Gastrointest Cancer 2022; 54:204-212. [PMID: 35020133 DOI: 10.1007/s12029-021-00801-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
Abstract
Hepatocellular carcinoma is one of the major causes of cancer-related deaths worldwide and is associated with several inflammatory mediators, since 90% of HCCs occur based on chronic hepatitis B or C, alcoholism or increasingly metabolic syndrome-associated inflammation. EMT is a physiological process, with coordinated changes in epithelial gene signatures and is regulated by multiple factors, including cytokines and growth factors such as TGFβ, EGF, and FGF. Recent reports propose a strong association between EMT and inflammation, which is also correlated with tumor aggressiveness and poor outcomes. Cellular heterogeneity results collectively as an outcome of EMT, inflammation, and the tumor microenvironment, and it plays a fundamental role in the progression, complexity of cancer, and chemoresistance. In this review, we highlight recent developments concerning the association of EMT and inflammation in the context of HCC progression. Identifying potential EMT-related biomarkers and understanding EMT regulatory molecules will likely contribute to promising developments in clinical practice and will be a valuable tool for predicting metastasis in general and specifically in HCC.
Collapse
|
8
|
OUP accepted manuscript. Glycobiology 2022; 32:556-579. [DOI: 10.1093/glycob/cwac014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/22/2022] [Accepted: 03/09/2022] [Indexed: 11/12/2022] Open
|
9
|
Grigolato R, Accorona R, Lombardo G, Corrocher G, Garagiola U, Massari F, Nicoli S, Rossi S, Calabrese L. Oral cancer in non-smoker non-drinker patients. Could comparative pet oncology help to understand risk factors and pathogenesis? Crit Rev Oncol Hematol 2021; 166:103458. [PMID: 34461267 DOI: 10.1016/j.critrevonc.2021.103458] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/03/2021] [Accepted: 08/22/2021] [Indexed: 01/16/2023] Open
Abstract
During the last decades there has been a progressive increase in proportion of incidence of oral cancer not related to a known etiologic factor, such as the so-called "oral cancer in young", a relevant tumor in non-smoker non-drinker (NSND) patients. The topic is matter of long standing debate, and adequate study models to analyze this entity are lacking. Spontaneous oral cancer in companion animals such as dogs and cats, presents more clinical and biological similarities with the human oral cancer than any other animal model. In our review we analyze how the study of spontaneous oral cancer in common pets can prospectively prove to be of double usefulness in unraveling the question about the origin of oral cancer in NSND patients, allowing both the analysis of environmental and behavioral risk factors, and the study of how carcinogenic viruses, chronic inflammation, and changes in immunity can influence pre-tumoral and tumoral microenvironment.
Collapse
Affiliation(s)
- Roberto Grigolato
- Division of Otorhinolaryngology, "San Maurizio" Hospital, Bolzano, Italy
| | - Remo Accorona
- Department of Otorhinolaryngology - Head and Neck Surgery, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy.
| | - Giorgio Lombardo
- School of Dentistry, Department of Surgery, Dentistry, Pediatrics, and Gynecology (DIPSCOMI), University of Verona, Verona, Italy
| | - Giovanni Corrocher
- School of Dentistry, Department of Surgery, Dentistry, Pediatrics, and Gynecology (DIPSCOMI), University of Verona, Verona, Italy
| | - Umberto Garagiola
- Biomedical, Surgical and Dental Sciences Department, Maxillofacial and Odontostomatology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | | | | | - Sabrina Rossi
- Department of Oncology and Hematology, Humanitas Cancer Center, Rozzano, Milano, Italy
| | - Luca Calabrese
- Division of Otorhinolaryngology, "San Maurizio" Hospital, Bolzano, Italy
| |
Collapse
|
10
|
Exploring the Crosstalk between Inflammation and Epithelial-Mesenchymal Transition in Cancer. Mediators Inflamm 2021; 2021:9918379. [PMID: 34220337 PMCID: PMC8219436 DOI: 10.1155/2021/9918379] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
Tumor cells undergo invasion and metastasis through epithelial-to-mesenchymal cell transition (EMT) by activation of alterations in extracellular matrix (ECM) protein-encoding genes, enzymes responsible for the breakdown of ECM, and activation of genes that drive the transformation of the epithelial cell to the mesenchymal type. Inflammatory cytokines such as TGFβ, TNFα, IL-1, IL-6, and IL-8 activate transcription factors such as Smads, NF-κB, STAT3, Snail, Twist, and Zeb that drive EMT. EMT drives primary tumors to metastasize in different parts of the body. T and B cells, dendritic cells (DCs), and tumor-associated macrophages (TAMs) which are present in the tumor microenvironment induce EMT. The current review elucidates the interaction between EMT tumor cells and immune cells under the microenvironment. Such complex interactions provide a better understanding of tumor angiogenesis and metastasis and in defining the aggressiveness of the primary tumors. Anti-inflammatory molecules in this context may open new therapeutic options for the better treatment of tumor progression. Targeting EMT and the related mechanisms by utilizing natural compounds may be an important and safe therapeutic alternative in the treatment of tumor growth.
Collapse
|
11
|
GP88/PGRN Serum Levels Are Associated with Prognosis for Oral Squamous Cell Carcinoma Patients. BIOLOGY 2021; 10:biology10050400. [PMID: 34064411 PMCID: PMC8147813 DOI: 10.3390/biology10050400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/12/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary An oral squamous cell carcinoma (OSCC) is a tumor of the oral cavity that has a five-year survival rate of only around 50%. As this rate has not increased in recent decades, despite improvements in diagnosis and therapy, novel, easily accessible biomarkers for prognosis assessment are still needed. In our study, we measured the growth factor protein progranulin/GP88 in the serum of OSCC patients and demonstrated that an increased serum GP88 level is associated with a better prognosis for the OSCC patients in our study group. Furthermore, serum GP88 levels were not significantly associated with age, sex, or the tumor’s histological features, indicating that serum GP88 levels may be an independent predictor of an individual OSCC patient’s prognosis. These findings may help to improve therapy management of an OSCC in personalized medicine. Abstract Progranulin (PGRN)/GP88 is a growth factor that is expressed in a wide range of tumor tissues. The secreted form is involved in various biological processes including proliferation and inflammation. In several tumor types, the serum GP88 level is associated with a patient’s prognosis; however, data for oral squamous cell carcinomas (OSCCs) have not yet been reported. We measured the serum GP88 levels in 96 OSCC patients by an enzyme immunosorbent assay (EIA) and correlated these data with clinicopathological parameters and patient outcomes. The GP88 levels in the serum of OSCC patients and healthy volunteers were comparable. In OSCC patients, the levels did not correlate with age, sex, or TNM status. In a Kaplan–Meier survival analysis, a serum GP88 level < 68 ng/mL was significantly associated with worsened survival (p = 0.0005, log-rank-test) as well as in uni- and multivariate Cox regression analyses (RR = 4.6 [1.6–12.9], p = 0.004 and RR = 4.2 [1.2–12.0], p = 0.008). This effect was predominant in OSCC patients older than 60.5 years (p = 0.027), while in younger patients no significant association between serum GP88 levels and prognosis could be observed. Altogether, lower serum GP88 levels are significantly associated with a worsened outcome for an OSCC and may be an interesting candidate for risk stratification during OSCC therapy.
Collapse
|
12
|
Ishida M, Takebayashi A, Kimura F, Nakamura A, Kitazawa J, Morimune A, Hanada T, Tsuta K, Murakami T. Induction of the epithelial-mesenchymal transition in the endometrium by chronic endometritis in infertile patients. PLoS One 2021; 16:e0249775. [PMID: 33826645 PMCID: PMC8026044 DOI: 10.1371/journal.pone.0249775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
Background The purpose of the present study was to evaluate the relationship between chronic endometritis and the epithelial-mesenchymal transition in the endometrium of infertile patients in the implantation phase. Methods Endometrial biopsy specimens from 66 infertility patients were analyzed. The presence of chronic endometritis was investigated by immunostaining for CD138. Immunohistochemical staining for E-cadherin, N-cadherin, Slug, and Snail was performed, and the expression profiles were statistically analyzed according to the presence of chronic endometritis. When the loss of E-cadherin expression and/or the positive expression of N-cadherin was detected, the specimen was considered epithelial-mesenchymal transition-positive. Epithelial-mesenchymal transition-positive cases were also statistically analyzed according to the presence of chronic endometritis. The characteristics of the patients in the epithelial-mesenchymal transition-positive and epithelial-mesenchymal transition-negative groups were compared. The association between variables, including age, body mass index, gravidity, parity, and each causative factor of infertility and epithelial-mesenchymal transition positivity was analyzed. Results The rates of the loss of E-cadherin expression, the gain of N-cadherin and epithelial-mesenchymal transition positivity were significantly higher in chronic endometritis patients. The expression of Slug, cytoplasmic Snail, and nuclear Snail was also detected at significantly higher rates in chronic endometritis patients. Chronic endometritis were related to the epithelial-mesenchymal transition. Conclusion The epithelial-mesenchymal transition was frequently detected in the endometrium in infertile patients with chronic endometritis. Since the epithelial-mesenchymal transition is associated with chronic endometritis, the epithelial-mesenchymal transition appears to be involved in the alteration of mechanisms of implantation.
Collapse
Affiliation(s)
- Mitsuaki Ishida
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Osaka, Japan
- * E-mail: (MI); (FK)
| | - Akie Takebayashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Fuminori Kimura
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Shiga, Japan
- * E-mail: (MI); (FK)
| | - Akiko Nakamura
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Jun Kitazawa
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Aina Morimune
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Koji Tsuta
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Osaka, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
13
|
Espinosa-Sánchez A, Suárez-Martínez E, Sánchez-Díaz L, Carnero A. Therapeutic Targeting of Signaling Pathways Related to Cancer Stemness. Front Oncol 2020; 10:1533. [PMID: 32984007 PMCID: PMC7479251 DOI: 10.3389/fonc.2020.01533] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
The theory of cancer stem cells (CSCs) proposes that the different cells within a tumor, as well as metastasis deriving from it, are originated from a single subpopulation of cells with self-renewal and differentiation capacities. These cancer stem cells are supposed to be critical for tumor expansion and metastasis, tumor relapse and resistance to conventional therapies, such as chemo- and radiotherapy. The acquisition of these abilities has been attributed to the activation of alternative pathways, for instance, WNT, NOTCH, SHH, PI3K, Hippo, or NF-κB pathways, that regulate detoxification mechanisms; increase the metabolic rate; induce resistance to apoptotic, autophagic, and senescence pathways; promote the overexpression of drug transporter proteins; and activate specific stem cell transcription factors. The elimination of CSCs is an important goal in cancer therapeutic approaches because it could decrease relapses and metastatic dissemination, which are main causes of mortality in oncology patients. In this work, we discuss the role of these signaling pathways in CSCs along with their therapeutic potential.
Collapse
Affiliation(s)
- Asunción Espinosa-Sánchez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Elisa Suárez-Martínez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Laura Sánchez-Díaz
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
- CIBER de Cancer, Madrid, Spain
| |
Collapse
|
14
|
Richardson LS, Menon PR, Menon R. The effects of extracellular matrix rigidity on 3-dimensional cultures of amnion membrane cells. Placenta 2019; 90:82-89. [PMID: 32056556 DOI: 10.1016/j.placenta.2019.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION To determine 3D growth of amnion membrane cells using soft substrate plates of various rigidities. METHODS Amnion epithelial (AEC) and mesenchymal cells (AMC) were cultured on 6-well soft substrate plates coated with matrigel and elastomer with rigidities of 0.5, 2, 8, 16, and 64 kPa (n = 3 each). Controls were cells in standard culture conditions. Cell morphology, spheroids' and sheets' formations and viability (bright field microscopy and crystal violet staining), and cellular transitions (vimentin/cytokeratin-18 [CK-18] ratios) were analyzed. A Student t-test was used for statistical analyses. RESULTS AECs in substrate rigidities between 2 and 8 kPa formed 3D features (spheroids and sheets) while retaining viability. Two kPa produced spheroids with epithelial characteristics (decrease in vimentin), and 8 kPa favored sheets. Transplantation and culture of AEC sheets with no matrix or elastomers, retained AECs' viability and maintained their epithelial characteristics. Optimum AMC growth was also between 2 and 8 kP A, with predominance of vimentin; however, AMCs did not form 3D structures. Lower and higher rigidities transitioned AMCs into AECs (decrease in vimentin). DISCUSSION Matrix rigidities between 2 and 8 kPa produced 3D structures of AECs (spheroids and sheets), resembling amnion membranes' morphology and exhibiting regenerative capacity in utero. Although AMCs grew in similar rigidities, a lack of 3D structures support their dispersed character in the membrane matrix. Extreme rigidities transitioned AMCs into AECs, suggesting that AMCs are transient cells (reservoirs) in the matrix required for remodeling. Compromises in matrix rigidity can cause membrane dysfunction and lead to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Lauren S Richardson
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA; Department of Neuroscience, Cell Biology & Anatomy, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Poorna R Menon
- Clear Falls High School, 4380 Village Way, League City, TX, 77573, USA
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
15
|
Sedaghat F, Cheraghpour M, Hosseini SA, Pourvali K, Teimoori-Toolabi L, Mehrtash A, Talaei R, Zand H. Hypomethylation of NANOG promoter in colonic mucosal cells of obese patients: a possible role of NF- κB. Br J Nutr 2019; 122:499-508. [PMID: 30157990 DOI: 10.1017/s000711451800212x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Obesity and particularly central obesity are the main risk factors of colon cancer. All intestinal cell populations including stem cells, their progenitors and differentiated colonocytes seem to be the origin of colorectal cancer. However, recent data support the role of differentiated cells as cancer origin especially during inflammation. Based on Yamanaka's seminal work, re-expression of few transcription factors in terminally differentiated cells creates stemness properti'es. Although these transcription factors are involved in tumorigenesis, they are epigenetically repressed in adult tissues. We proposed that obesity might regulate methylation of stemness genes in colonocytes via inflammatory signalling. Obesity-associated inflammation was analysed using Western blot analysis of phospho-IκB (inhibitor of NF-κB). Methylation-sensitive high-resolution melting analysis was performed on colonic mucosal samples of twenty obese and twenty normal-weight men to analyse promoter methylation of POU5F1 (OCT4), NANOG, MYC and CDKN2A. TNF-treated HT-29 cells were used to recapitulate the effect of NF-κB activation on stemness genes methylation. Our results showed that colonic phosphorylation of IκB, as a signal of NF-κB activation, was higher in obese subjects compared with their normal-weight counterparts. Moreover, promoter methylation of NANOG was likely to be lower in obese subjects and correlated with central obesity. HT-29 cells incubated by TNF-α showed hypomethylation of POU5F1 and MYC genes in addition to the NANOG. These results suggest that obesity-induced inflammation might be involved in the regulation of DNA methylation of oncogenic genes such as NANOG in differentiated colonocytes and thus predispose them to later oncogenic alterations.
Collapse
Affiliation(s)
- Fatemeh Sedaghat
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, 1981619573 Tehran, Iran
| | - Makan Cheraghpour
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, 6135715794 Ahvaz, Iran
| | - Seyed Ahmad Hosseini
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, 6135715794 Ahvaz, Iran
| | - Katayoun Pourvali
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, 1981619573 Tehran, Iran
| | - Ladan Teimoori-Toolabi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Amirhosein Mehrtash
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Ramin Talaei
- Faculty of Medicine, Shahid Modaress Hospital, Shahid Beheshti University of Medical Sciences, 1998734383 Tehran, Iran
| | - Hamid Zand
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, 1981619573 Tehran, Iran
| |
Collapse
|
16
|
Xia H, Xue J, Xu H, Lin M, Shi M, Sun Q, Xiao T, Dai X, Wu L, Li J, Xiang Q, Tang H, Bian Q, Liu Q. Andrographolide antagonizes the cigarette smoke-induced epithelial-mesenchymal transition and pulmonary dysfunction through anti-inflammatory inhibiting HOTAIR. Toxicology 2019; 422:84-94. [PMID: 31128153 DOI: 10.1016/j.tox.2019.05.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023]
Abstract
In cells of the lung surface, cigarette smoke (CS) induces inflammatory and epithelial-mesenchymal transition (EMT), effects that are related to pulmonary dysfunction and Chronic obstructive pulmonary disease (COPD). However, the molecular mechanisms involved remain largely unknown, and potential therapeutic approaches are under development. In the present study, with cell culture and animal studies, we showed that CS exposure causes pulmonary dysfunction and airway remodeling with inflammatory cell infiltration. Consistent with these pulmonary lesions, the inflammatory factors interleukin-6 (IL-6) and interleukin-8 (IL-8) were increased in mice exposed to CS for 4 days. Accordingly, downstream signal transducer and activator of transcription 3 (STAT3) was activated, which up-regulated expression of the lncRNA HOTAIR, and enhancer of zeste homolog 2 (EZH2). In addition, CS exposure led to decreased levels of E-cadherin and to increased N-cadherin, vimentin, and α-SMA, indicating that the EMT was induced in mouse lung tissues. These effects, including increases of IL-6 and HOTAIR, were confirmed in human bronchial epithelial (HBE) cells treated with cigarette smoke extract (CSE). Finally, we established that, in HBE cells, andrographolide reversed the CSE-induced EMT via decreasing IL-6 levels and, in an animal model, prevented CS-induced lung inflammation and small airway remodeling, indicating that it has potential clinical application for CS-induced pulmonary dysfunction and COPD.
Collapse
Affiliation(s)
- Haibo Xia
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Junchao Xue
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Hui Xu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Min Lin
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Ming Shi
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Qian Sun
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Tian Xiao
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Xiangyu Dai
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Lu Wu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Junjie Li
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Quanyong Xiang
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Huanwen Tang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong, People's Republic of China
| | - Qian Bian
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, Jiangsu, People's Republic of China.
| | - Qizhan Liu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
17
|
Huang WQ, Long WQ, Mo XF, Zhang NQ, Luo H, Lin FY, Huang J, Zhang CX. Direct and indirect associations between dietary magnesium intake and breast cancer risk. Sci Rep 2019; 9:5764. [PMID: 30962499 PMCID: PMC6453912 DOI: 10.1038/s41598-019-42282-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/27/2019] [Indexed: 01/07/2023] Open
Abstract
This study aimed to explore the effect of dietary magnesium intake on breast cancer risk both directly and indirectly via its effect on inflammatory markers C-reactive protein (CRP) and interleukin-6 (IL-6). This case-control study recruited 1050 case patients and 1229 control subjects. Inflammatory marker levels of 322 cases and 322 controls, randomly selected, were measured using ELISA, and data on dietary magnesium intake were collected using a food frequency questionnaire. Multivariable logistic regression was used to estimate the odds ratio (OR) and 95% confidence interval (CI), and path analysis was used to investigate the mediating effect. A higher magnesium intake was associated with a lower breast cancer risk (adjusted OR = 0.80, 95% CI = 0.65, 0.99). A positive association was found between the CRP level and breast cancer risk (adjusted OR = 1.43, 95% CI = 1.02-2.01). However, IL-6 was not found to be associated with breast cancer risk. Path analysis revealed that dietary magnesium affected breast cancer risk both directly and indirectly by influencing the CRP level. The results indicate that a direct negative association and an indirect association through influencing the CRP level were observed between dietary magnesium intake and breast cancer risk.
Collapse
Affiliation(s)
- Wu-Qing Huang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wei-Qing Long
- Department of Clinical Laboratory, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiong-Fei Mo
- Department of Vascular Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Nai-Qi Zhang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hong Luo
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fang-Yu Lin
- Nursing Department, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing Huang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Cai-Xia Zhang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
18
|
Induction of the Epithelial-to-Mesenchymal Transition of Human Colorectal Cancer by Human TNF-β (Lymphotoxin) and its Reversal by Resveratrol. Nutrients 2019; 11:nu11030704. [PMID: 30917533 PMCID: PMC6471988 DOI: 10.3390/nu11030704] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022] Open
Abstract
Objective: Tumor necrosis factor-beta (TNF-β), as an inflammatory mediator that has been shown to promote tumorigenesis, induces NF-κB. Natural multi-targeted agent resveratrol in turn shows anti-inflammatory and anti-cancer properties. Epithelial-to-mesenchymal transition (EMT) allows cancer cells to turn into a motile state with invasive capacities and is associated with metastasis and development of cancer stem cells (CSC). However, TNF-β-induced EMT and the anti-invasion mechanism of resveratrol on CRC are not yet completely understood. Methods: We investigated the underlying molecular mechanisms of resveratrol on TNF-β/TNF-βR-induced EMT and migration of CRC cells (HCT116, RKO, SW480) in monolayer or 3D alginate cultures. Results: TNF-β, similar to TNF-α, induced significant cell proliferation, morphological change, from an epithelial to a spindle-like mesenchymal shape with the formation of filopodia and lamellipodia associated with the expression of EMT parameters (elevated vimentin and slug, reduced E-cadherin), increased migration/invasion, and formation of CSC in all CRC cells. Interestingly, these effects were dramatically decreased in the presence of resveratrol or anti-TNF-βR with TNF-β co-treatment, inducing biochemical changes to the mesenchymal-epithelial transition (MET), with a planar cell surface and suppressed formation of CSC cells. This was associated with a significant increase in apoptosis. Furthermore, we found that resveratrol suppressed TNF-β-induced NF-κB and NF-κB-regulated gene biomarkers associated with growth, proliferation, and invasion. Finally, TNF-βR interacts directly with focal adhesion kinase (FAK) and NF-κB. Conclusion: These results suggest that resveratrol down-regulates TNF-β/TNF-βR-induced EMT, at least in part via specific suppression of NF-κΒ and FAK in CRC cells.
Collapse
|
19
|
Over-expressed miRNA-200b ameliorates ulcerative colitis-related colorectal cancer in mice through orchestrating epithelial-mesenchymal transition and inflammatory responses by channel of AKT2. Int Immunopharmacol 2018; 61:346-354. [PMID: 29933193 DOI: 10.1016/j.intimp.2018.06.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/25/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
Abstract
Our study was to explore the potential role of miRNA-200b in modulating tumorigenesis in the model of ulcerative colitis-related colorectal cancer (UCRCC) and, further, to decipher the underlying mechanisms associated with this effect. In this study, we examined a greater number of polyps or adenomas, a higher grade of epithelial dysplasia accompanied with a decrease in survival ratio in azoxymethane (AOM)/dextran sulfate sodium (DSS) model mice compared to mice treated with over-expressed miRNA-200b. Surprisingly, enforced miRNA-200b expression significantly suppressed AOM/DSS-induced up-regulation of oncologic markers including β-catenin and CD133. Independent of this, treatment with miRNA-200b obviously attenuated inflammatory responses, as indicated by down-regulating tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β) and blockade of AKT2-mediated NF-κB/IL-6/STAT3 signaling pathway. Furthermore, a simultaneous shift in epithelial-mesenchymal transition (EMT) markers such as E-cadherin and N-cadherin were observed to be increased and decreased, respectively. Coupled with the associated influence of over-expressed miRNA-200b were change in colorectal cell morphology shown by Transmission electron microscope (TEM) and a decrease in expression of rho-kinase2 (ROCK2) together with AKT2 phosphorylation (p-AKT2). Moreover, mice which were transfected with negative control of miRNA-200b possessed results that were in line with that obtained from AOM/DSS model mice. Additionally, we demonstrated that the 3'untranslated region (UTR) of AKT2 was a direct target of miRNA-200b through bioinformatics analysis and dual-luciferase assay. Collectively, these findings suggest that miRNA-200b's contribution to tumor-suppressing program was correlated with EMT and inflammatory responses in a AKT2-dependent manner.
Collapse
|
20
|
Baker K. Organoids Provide an Important Window on Inflammation in Cancer. Cancers (Basel) 2018; 10:E151. [PMID: 29883385 PMCID: PMC5977124 DOI: 10.3390/cancers10050151] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 02/08/2023] Open
Abstract
Inflammation is a primary driver of cancer initiation and progression. However, the complex and dynamic nature of an inflammatory response make this a very difficult process to study. Organoids are a new model system where complex multicellular structures of primary cells can be grown in a 3D matrix to recapitulate the biology of the parent tissue. This experimental model offers several distinct advantages over alternatives including the ability to be genetically engineered, implanted in vivo and reliably derived from a wide variety of normal and cancerous tissue from patients. Furthermore, long-term organoid cultures reproduce many features of their source tissue, including genetic and epigenetic alterations and drug sensitivity. Perhaps most significantly, cancer organoids can be cocultured in a variety of different systems with a patients’ own immune cells, uniquely permitting the study of autologous cancer-immune cell interactions. Experiments with such systems promise to shed light on the mechanisms governing inflammation-associated cancer while also providing prognostic information on an individual patient’s responsiveness to immunotherapeutic anti-cancer drugs. Thanks to their ability to capture important features of the complex relationship between a cancer and its microenvironment, organoids are poised to become an essential tool for unraveling the mechanisms by which inflammation promotes cancer.
Collapse
Affiliation(s)
- Kristi Baker
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada.
| |
Collapse
|
21
|
Chen W, Ren X, Wu J, Gao X, Cen X, Wang S, Sheng S, Chen Q, Tang Y, Liang X, Tang Y. HSP27 associates with epithelial-mesenchymal transition, stemness and radioresistance of salivary adenoid cystic carcinoma. J Cell Mol Med 2018; 22:2283-2298. [PMID: 29424489 PMCID: PMC5867171 DOI: 10.1111/jcmm.13510] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/26/2017] [Indexed: 02/05/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been shown to associate with cancer stem cells and radioresistance. However, it is obscure whether EMT itself or specific EMT regulators play causal roles in these properties of salivary adenoid cystic carcinoma (SACC). Here, we exhibited that overexpression of HSP27 drove the migration and invasion, induced EMT, as well as mediated TGF-β1-induced EMT in SACC cells, accompanying the up-regulation of Snail1 and Prrx1. Conversely, HSP27 silencing reduced the migration and invasion and contributed to MET of SACC cells. HSP27 indirectly down-regulates the expression of E-cadherin through activating Snail1 and Prrx1 expressions. Overexpression of Snail1 or Prrx1 restored the migration and invasion in HSP27 knockdown cells. Enforced expression of HSP27 enhanced colony formation, CD133+ /CD44+ population and radioresistance of SACC cell lines. In addition, HSP27 expression was positively associated with radioresistance and poor prognosis of SACC patients as well as with the expression of Prrx1 or Snail1 in SACC tissues. The data confirm an important function for HSP27 in SACC progression through regulating EMT and stemness, and they imply the possible association between EMT and radioresistance of SACC.
Collapse
Affiliation(s)
- Wei Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
- Department of Oral and Maxillofacial SurgeryStomatological Hospital of Nankai UniversityTianjinChina
| | - Xiaohua Ren
- Department of StomatologySichuan Academy of Medical Sciences and Sichuan People's HospitalChengduChina
| | - Jiashun Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xiaolei Gao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xiao Cen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Shasha Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Surui Sheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Qianming Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ya‐jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education)Hubei Provincial Cooperative Innovation Center of Industrial FermentationHubei Key Laboratory of Industrial MicrobiologyHubei University of TechnologyWuhanChina
| | - Xin‐hua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ya‐ling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
- Department of Oral PathologyWest China Hospital of Stomatology (Sichuan University)ChengduChina
| |
Collapse
|
22
|
Resistin facilitates breast cancer progression via TLR4-mediated induction of mesenchymal phenotypes and stemness properties. Oncogene 2017; 37:589-600. [PMID: 28991224 DOI: 10.1038/onc.2017.357] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022]
Abstract
Growing evidence indicates that resistin-an obesity-related cytokine-is upregulated in breast cancer patients, yet its impact on breast cancer behavior remains to be ascertained. Similarly, Toll-like receptor 4 (TLR4) has been implicated in breast cancer progression, however, its clinically relevant endogenous ligand remains elusive. In this study, we observed that high serum resistin levels in breast cancer patients positively correlated with tumor stage, size and lymph node metastasis. These findings were replicated in animal models of breast cancer tumorigenesis and metastasis. Resistin was found to promote epithelial-mesenchymal transition and stemness in breast cancer cells-mechanisms critical to tumorigenesis and metastasis-through a TLR4/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/signal transducer and activator of transcription 3 (STAT3) signaling pathway and negated by TLR4-specific antibody and antagonist. These findings provide clear evidence that resistin is a clinically relevant endogenous ligand for TLR4, which promotes tumor progression via TLR4/NF-κB/STAT3 signaling, providing insights into a novel therapeutic target in breast cancer.
Collapse
|
23
|
Zeeshan R, Mutahir Z. Cancer metastasis - tricks of the trade. Bosn J Basic Med Sci 2017; 17:172-182. [PMID: 28278128 DOI: 10.17305/bjbms.2017.1908] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 01/21/2017] [Accepted: 01/22/2017] [Indexed: 12/18/2022] Open
Abstract
Decades of cancer research have unraveled genetic, epigenetic and molecular pathways leading to plausible therapeutic targets; many of which hold great promise in improving clinical outcomes. Metastatic tumors become evident early on and are one of the major causes of cancer-related fatalities worldwide. This review depicts the sequential events of cancer metastasis. Genetic and epigenetic heterogeneity influences local tumor cell invasion, intravasation, survival in circulation, extravasation and colonization to distant sites. Each sequential event is associated with heterogeneous tumor microenvironment, gain of competence, unique population of cancer stem cells (CSCs), circulatory pathway, compatible niche and immune system support. A tight regulation of metastasis-promoting mechanisms and, in parallel, evading inhibitory mechanisms contribute to the severity and site of metastasis. A comprehensive understanding of tumor cell fate as an individual entity, as well as in combination with different promoting factors and associated molecular mechanisms, is anticipated in the coming years. This will enable scientists to depict design strategies for targeted cancer therapies.
Collapse
Affiliation(s)
- Rabia Zeeshan
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS Institute of Information Technology, Lahore, Pakistan.
| | | |
Collapse
|
24
|
Obesity and p16 INK4A Downregulation Activate Breast Adipocytes and Promote Their Protumorigenicity. Mol Cell Biol 2017. [PMID: 28630279 DOI: 10.1128/mcb.00101-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Obesity is increasingly recognized as a risk factor for breast cancer development. However, the molecular basis of obesity-related breast carcinogenesis remains elusive. In this study, we have shown that obesity reduces the level of the tumor suppressor p16INK4A protein in breast adipocytes, which showed active features and strong procarcinogenic potential both in vitro and in orthotopic tumor xenografts compared to mature adipocytes from lean women. Furthermore, obesity triggered epithelial-to-mesenchymal transition (EMT) in breast ductal epithelial cells. Interestingly, specific downregulation of p16INK4A increased the expression/secretion levels of various adipokines, including leptin, and activated breast adipocytes from lean women. Consequently, like breast adipocytes from obese women, p16-deficient adipocytes induced EMT in normal primary breast luminal cells in a leptin-dependent manner and enhanced tumor growth. Additionally, we have shown that p16INK4A negatively controls leptin at the mRNA level through microRNAs 141 and 146b-5p (miR-141 and miR-146b-5p), which bind the leptin mRNA at a specific sequence in the 3' untranslated region (UTR). These results show that obesity activates breast stromal adipocytes through p16 downregulation, which upregulates leptin and promotes procarcinogenic processes.
Collapse
|
25
|
Kim JH, Shim JW, Eum DY, Kim SD, Choi SH, Yang K, Heo K, Park MT. Downregulation of UHRF1 increases tumor malignancy by activating the CXCR4/AKT-JNK/IL-6/Snail signaling axis in hepatocellular carcinoma cells. Sci Rep 2017; 7:2798. [PMID: 28584306 PMCID: PMC5459852 DOI: 10.1038/s41598-017-02935-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 04/20/2017] [Indexed: 12/19/2022] Open
Abstract
UHRF1 (ubiquitin-like, with PHD and RING finger domains 1) plays a crucial role in DNA methylation, chromatin remodeling and gene expression and is aberrantly upregulated in various types of human cancers. However, the precise role of UHRF1 in cancer remains controversial. In this study, we observed that hypoxia-induced downregulation of UHRF1 contributes to the induction of the epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma cells. By negatively modulating UHRF1 expression, we further showed that UHRF1 deficiency in itself is sufficient to increase the migratory and invasive properties of cells via inducing EMT, increasing the tumorigenic capacity of cells and leading to the expansion of cancer stem-like cells. Epigenetic changes caused by UHRF1 deficiency triggered the upregulation of CXCR4, thereby activating AKT and JNK to increase the expression and secretion of IL-6. In addition, IL-6 readily activated the JAK/STAT3/Snail signaling axis, which subsequently contributed to UHRF1 deficiency-induced EMT. Our results collectively demonstrate that UHRF1 deficiency may play a pivotal role in the malignant alteration of cancer cells.
Collapse
Affiliation(s)
- Ji-Hyun Kim
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Republic of Korea
| | - Jae-Woong Shim
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Republic of Korea
| | - Da-Young Eum
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Republic of Korea
| | - Sung Dae Kim
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Republic of Korea
| | - Si Ho Choi
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Republic of Korea
| | - Kwangmo Yang
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Republic of Korea
| | - Kyu Heo
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Republic of Korea.
| | - Moon-Taek Park
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, 46033, Republic of Korea.
| |
Collapse
|
26
|
Khalafalla FG, Khan MW. Inflammation and Epithelial-Mesenchymal Transition in Pancreatic Ductal Adenocarcinoma: Fighting Against Multiple Opponents. CANCER GROWTH AND METASTASIS 2017; 10:1179064417709287. [PMID: 28579826 PMCID: PMC5436837 DOI: 10.1177/1179064417709287] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and one of the most lethal human cancers. Inflammation is a critical component in PDAC initiation and progression. Inflammation also contributes to the aggressiveness of PDAC indirectly via induction of epithelial-mesenchymal transition (EMT), altogether leading to enhanced resistance to chemotherapy and poor survival rates. This review gives an overview of the key pro-inflammatory signaling pathways involved in PDAC pathogenesis and discusses the role of inflammation in induction of EMT and development of chemoresistance in patients with PDAC.
Collapse
|
27
|
Interplay between Inflammation and Stemness in Cancer Cells: The Role of Toll-Like Receptor Signaling. J Immunol Res 2016; 2016:4368101. [PMID: 28116318 PMCID: PMC5223024 DOI: 10.1155/2016/4368101] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 11/22/2016] [Accepted: 12/05/2016] [Indexed: 02/08/2023] Open
Abstract
Cancer stem cells (CSCs) are a small population of cancer cells that exhibit stemness. These cells contribute to cancer metastasis, treatment resistance, and relapse following therapy; therefore, they may cause malignancy and reduce the success of cancer treatment. Nuclear factor kappa B- (NF-κB-) mediated inflammatory responses increase stemness in cancer cells, and CSCs constitutively exhibit higher NF-κB activation, which in turn increases their stemness. These opposite effects form a positive feedback loop that further amplifies inflammation and stemness in cancer cells, thereby expanding CSC populations in the tumor. Toll-like receptors (TLRs) activate NF-κB-mediated inflammatory responses when stimulated by carcinogenic microbes and endogenous molecules released from cells killed during cancer treatment. NF-κB activation by extrinsic TLR ligands increases stemness in cancer cells. Moreover, it was recently shown that increased NF-κB activity and inflammatory responses in CSCs may be caused by altered TLR signaling during the enrichment of stemness in cancer cells. Thus, the activation of TLR signaling by extrinsic and intrinsic factors drives a positive interplay between inflammation and stemness in cancer cells.
Collapse
|
28
|
Mei J, Li MQ, Li DJ, Sun HX. MicroRNA expression profiles and networks in CXCL12‑stimulated human endometrial stromal cells. Mol Med Rep 2016; 15:249-255. [PMID: 27959395 PMCID: PMC5355667 DOI: 10.3892/mmr.2016.5997] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/10/2016] [Indexed: 02/02/2023] Open
Abstract
The chemokine stromal cell-derived factor-1 (C-X-C motif chemokine ligand 12; CXCL12) is important in the recruitment of leukocytes to the peritoneal cavity and the regulation of endometriotic tissue growth in endometriosis patients. However, the alterations in microRNA (miRNA) expression induced by CXCL12 remain to be fully elucidated. The present study evaluated key miRNAs in CXCL12‑stimulated endometrial stromal cells (ESCs), and investigated the underlying cellular regulatory mechanisms of CXCL12 in endometriosis by building networks between miRNAs, genes and gene ontologies (GOs). Differential expression of miRNAs and mRNAs induced by CXCL12 stimulation in ESCs was measured using miRNA and gene chips, and it was observed that 35 miRNAs and 1,671 mRNAs were differentially expressed. Using potential target genes of the 35 miRNAs, intersections of these genes were examined and 63 intersection genes were identified. A total of 39 GOs were obtained for these intersection genes, based on information from GO databases, including immune cell chemoattractants, inflammatory and immune responses, and pathological processes of endometriotic lesions in endometriosis. In addition, miRNA‑gene networks were built according to the GO and Kyoto Encyclopedia of Genes and Genomes databases. The present study, to the best of our knowledge, provides the most complete miRNAome and mRNAome profiles, and the most detailed investigation of the underlying cellular regulatory mechanisms, of the effects of CXCL12 in endometriosis. These results may facilitate the complete elucidation of the role of CXCL12 in endometriosis, and its underlying epigenetic mechanisms.
Collapse
Affiliation(s)
- Jie Mei
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, P.R. China
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, P.R. China
| | - Hai-Xiang Sun
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
29
|
Abstract
BACKGROUND Metastasis is the main cause of mortality in cancer patients. Two major routes of cancer cell spread are currently being recognized: dissemination via blood vessels (hematogenous spread) and dissemination via the lymphatic system (lymphogenous spread). Here, our current knowledge on the role of both blood and lymphatic vessels in cancer cell metastasis is summarized. In addition, I will discuss why cancer cells select one or both of the two routes to disseminate and I will provide a short description of the passive and active models of intravasation. Finally, lymphatic vessel density (LVD), blood vessel density (BVD), interstitial fluid pressure (IFP) and tumor hypoxia, as well as regional lymph node metastasis and the recently discovered primo vascular system (PVS) will be highlighted as important factors influencing tumor cell motility and spread and, ultimately, clinical outcome. CONCLUSIONS Lymphangiogenesis and angiogenesis are important phenomena involved in the spread of cancer cells and they are associated with a poor prognosis. It is anticipated that new discoveries and advancing knowledge on these phenomena will allow an improvement in the treatment of cancer patients.
Collapse
Affiliation(s)
- Roman Paduch
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland.
- Department of General Ophthalmology, Medical University of Lublin, Chmielna 1, 20-079, Lublin, Poland.
| |
Collapse
|
30
|
Liu B, Yan S, Jia Y, Ma J, Wu S, Xu Y, Shang M, Mao A. TLR2 promotes human intrahepatic cholangiocarcinoma cell migration and invasion by modulating NF-κB pathway-mediated inflammatory responses. FEBS J 2016; 283:3839-3850. [PMID: 27616304 DOI: 10.1111/febs.13894] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 08/25/2016] [Accepted: 09/08/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Bingyan Liu
- Department of Interventional Radiology; Tongren Hospital; Shanghai Jiaotong University School of Medicine; China
| | - Shuo Yan
- Department of Interventional Radiology; Tongren Hospital; Shanghai Jiaotong University School of Medicine; China
| | - Yiping Jia
- Department of Interventional Radiology; Tongren Hospital; Shanghai Jiaotong University School of Medicine; China
| | - Jun Ma
- Department of Interventional Radiology; Tongren Hospital; Shanghai Jiaotong University School of Medicine; China
| | - Shaoqiu Wu
- Department of Interventional Radiology; Tongren Hospital; Shanghai Jiaotong University School of Medicine; China
| | - Yuyao Xu
- Department of Interventional Radiology; Tongren Hospital; Shanghai Jiaotong University School of Medicine; China
| | - Mingyi Shang
- Department of Interventional Radiology; Tongren Hospital; Shanghai Jiaotong University School of Medicine; China
| | - Aiwu Mao
- Department of Interventional Radiology; Tongren Hospital; Shanghai Jiaotong University School of Medicine; China
| |
Collapse
|
31
|
Giladi ND, Ziv-Av A, Lee HK, Finniss S, Cazacu S, Xiang C, Waldman Ben-Asher H, deCarvalho A, Mikkelsen T, Poisson L, Brodie C. RTVP-1 promotes mesenchymal transformation of glioma via a STAT-3/IL-6-dependent positive feedback loop. Oncotarget 2016; 6:22680-97. [PMID: 26267319 PMCID: PMC4673191 DOI: 10.18632/oncotarget.4205] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/06/2015] [Indexed: 01/06/2023] Open
Abstract
Glioblastomas (GBMs), the most aggressive primary brain tumors, exhibit increased invasiveness and resistance to anti-tumor treatments. We explored the role of RTVP-1, a glioma-associated protein that promotes glioma cell migration, in the mesenchymal transformation of GBM. Analysis of The Cancer Genome Atlas (TCGA) demonstrated that RTVP-1 expression was higher in mesenchymal GBM and predicted tumor recurrence and poor clinical outcome. ChiP analysis revealed that the RTVP-1 promoter binds STAT3 and C/EBPβ, two master transcription factors that regulate mesenchymal transformation of GBM. In addition, IL-6 induced RTVP-1 expression in a STAT3-dependent manner. RTVP-1 increased the migration and mesenchymal transformation of glioma cells. Similarly, overexpression of RTVP-1 in human neural stem cells induced mesenchymal differentiation, whereas silencing of RTVP-1 in glioma stem cells (GSCs) decreased the mesenchymal transformation and stemness of these cells. Silencing of RTVP-1 also increased the survival of mice bearing GSC-derived xenografts. Using gene array analysis of RTVP-1 silenced glioma cells we identified IL-6 as a mediator of RTVP-1 effects on the mesenchymal transformation and migration of GSCs, therefore acting in a positive feedback loop by upregulating RTVP-1 expression via the STAT3 pathway. Collectively, these results implicate RTVP-1 as a novel prognostic marker and therapeutic target in GBM.
Collapse
Affiliation(s)
- Nis David Giladi
- Everard and Mina Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Amotz Ziv-Av
- Everard and Mina Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Hae Kyung Lee
- Department of Neurosurgery, Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Susan Finniss
- Department of Neurosurgery, Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Simona Cazacu
- Department of Neurosurgery, Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Cunli Xiang
- Department of Neurosurgery, Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Hiba Waldman Ben-Asher
- Everard and Mina Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Ana deCarvalho
- Department of Neurosurgery, Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Tom Mikkelsen
- Department of Neurosurgery, Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| | - Laila Poisson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Chaya Brodie
- Everard and Mina Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.,Department of Neurosurgery, Davidson Laboratory of Cell Signaling and Tumorigenesis, Hermelin Brain Tumor Center, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
32
|
Wang D, Fu L, Sun H, Guo L, DuBois RN. Prostaglandin E2 Promotes Colorectal Cancer Stem Cell Expansion and Metastasis in Mice. Gastroenterology 2015; 149:1884-1895.e4. [PMID: 26261008 PMCID: PMC4762503 DOI: 10.1053/j.gastro.2015.07.064] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 07/09/2015] [Accepted: 07/30/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Inflammation may contribute to the formation, maintenance, and expansion of cancer stem cells (CSCs), which have the capacity for self-renewal, differentiation, and resistance to cytotoxic agents. We investigated the effects of the inflammatory mediator prostaglandin E2 (PGE2) on colorectal CSC development and metastasis in mice and the correlation between levels of PGE2 and CSC markers in human colorectal cancer (CRC) specimens. METHODS Colorectal carcinoma specimens and matched normal tissues were collected from patients at the Mayo Clinic (Scottsdale, AZ) and analyzed by mass spectrometry and quantitative polymerase chain reaction. Human primary CRC cells and mouse tumor cells were isolated using microbeads or flow cytometry and analyzed for sphere-formation and by flow cytometry assays. LS-174T cells were sorted by flow cytometry (for CD133(+)CD44(+) and CD133(-)CD44(-) cells) and also used in these assays. NOD-scidIL-2Rγ(-/-) (NSG) mice were given cecal or subcutaneous injections of LS-174T or human primary CRC cells. Apc(Min/+) mice and NSG mice with orthotopic cecal tumors were given vehicle (controls), PGE2, celecoxib, and/or Ono-AE3-208. PGE2 downstream signaling pathways were knocked down with small hairpin RNAs, expressed from lentiviral vectors in LS-174T cells, or blocked with inhibitors in human primary CRC cells. RESULTS Levels of PGE2 correlated with colonic CSC markers (CD133, CD44, LRG5, and SOX2 messenger RNAs) in human colorectal carcinoma samples. Administration of PGE2 to Apc(Min/+) mice increased tumor stem cells and tumor burden, compared with controls. NSG mice given PGE2 had increased numbers of cecal CSCs and liver metastases compared with controls after intracecal injection of LS-174T or human primary CRC cells. Alternatively, celecoxib, an inhibitor of prostaglandin-endoperoxide synthase 2, reduced polyp numbers in Apc(Min/+) mice, liver metastasis in NSG mice with orthotopic tumors, and numbers of CSCs in Apc(Min/+) and NSG mice. Inhibitors or knockdown of PGE2 receptor 4 (EP4), phosphoinositide 3-kinase (PI3K) p85α, extracellular signal-regulated kinase 1 (ERK1), or nuclear factor (NF)-κB reduced PGE2-induced sphere formation and expansion of LS-174T and/or human primary CRC cells. Knockdown of ERK1 or PI3K p85α also attenuated PGE2-induced activation of NF-κB in LS-174T cells. An EP4 antagonist reduced the ability of PGE2 to induce CSC expansion in orthotopic tumors and to accelerate the formation of liver metastases. Knockdown experiments showed that NF-κB was required for PGE2 induction of CSCs and metastasis in mice. CONCLUSIONS PGE2 induces CSC expansion by activating NF-κB, via EP4-PI3K and EP4-mitogen-activated protein kinase signaling, and promotes the formation of liver metastases in mice. The PGE2 signaling pathway therefore might be targeted therapeutically to slow CSC expansion and colorectal cancer progression.
Collapse
Affiliation(s)
- Dingzhi Wang
- Laboratory for Inflammation and Cancer, Biodesign Institute of Arizona State University, Tempe, AZ 85287
| | - Lingchen Fu
- Laboratory for Inflammation and Cancer, Biodesign Institute of Arizona State University, Tempe, AZ 85287
| | - Haiyan Sun
- Laboratory for Inflammation and Cancer, Biodesign Institute of Arizona State University, Tempe, AZ 85287
| | - Lixia Guo
- Laboratory for Inflammation and Cancer, Biodesign Institute of Arizona State University, Tempe, AZ 85287,Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905 (the present affiliation)
| | - Raymond N. DuBois
- Laboratory for Inflammation and Cancer, Biodesign Institute of Arizona State University, Tempe, AZ 85287,Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287,Department of Research and Division of Gastroenterology, Mayo Clinic, Scottsdale, AZ 85259,Correspondence to: Raymond N. DuBois, MD. Ph.D., Executive Director of the Biodesign Institute at Arizona State University, PO Box 875001, 1001 S. McAllister Ave., Tempe, AZ 85287, Tel: 480-965-1228 and Fax: 480-727-9550,
| |
Collapse
|
33
|
Liu R, Zheng H, Li W, Guo Q, He S, Hirasaki Y, Hou W, Hua B, Li C, Bao Y, Gao Y, Qi X, Pei Y, Zhang Y. Anti-tumor enhancement of Fei-Liu-Ping ointment in combination with celecoxib via cyclooxygenase-2-mediated lung metastatic inflammatory microenvironment in Lewis lung carcinoma xenograft mouse model. J Transl Med 2015; 13:366. [PMID: 26597177 PMCID: PMC4656184 DOI: 10.1186/s12967-015-0728-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/10/2015] [Indexed: 12/19/2022] Open
Abstract
Background Fei-Liu-Ping (FLP) ointment is an oral prescription medication that has been widely applied to treat lung cancer patients in China. Regulation of the metastatic microenvironment is an important therapeutic approach for prevention and treatment of tumor recurrence and metastasis. The advantage of Traditional Chinese Medicine management of lung cancer lies in the prevention of recurrence and metastasis. Our previous study has demonstrated that FLP ointment could regulate lung inflammatory microenvironment in vitro. However, the effects of FLP on the tumor microenvironment in vivo are still poorly understood. The objective of this study is to investigate the effect of FLP alone or in combination with celecoxib in the prevention of lung cancer progression by Cyclooxygenase (Cox)-2 mediated tumor inflammatory microenvironment in vivo. Methods 120 Lewis lung carcinoma xenograft mice were divided equally into four groups: vehicle, FLP, celecoxib, and FLP plus celecoxib. The dynamic growth of the xenografted tumors was observed using an in vivo fluorescence imaging system. Mice were sacrificed on day 14, day 21, and day 28, and tumor specimens and lung tissues were harvested to detect the metastasis-associated protein expression. Results Tumor inhibition rate was 15.4, 44.2, 47.4 % at day 14, 37.3, 34.7, 61.5 % at day 21, and 15.5, 10.3, 32.5 % at day 28 after treatment of FLP, celecoxib, and FLP plus celecoxib, respectively. Upon treatment of FLP and celecoxib together, lung metastasis rate was 30 % (8 metastatic nodules) lower than other groups. FLP inhibited Cox-2 expression in a time-dependent manner. Moreover, FLP inhibited N-cadherin, matrix metalloproteinases (MMP)-9, and Vimentin expression. Treatment of FLP in combination with celecoxib was more effective than FLP or celecoxib alone in inhibiting vascular endothelial growth factor, platelet-derived growth factor receptors β, microsomal Prostaglandin E synthase-1, MMP-2, MMP-9, N-cadherin, and Vimentin expression, but increased E-cadherin expression. Conclusions FLP inhibited tumor growth and metastasis in a Lewis lung xenograft mice model through the Cox-2 pathway. FLP in combination with celecoxib enhanced the antitumor growth and anti-metastasis effects. Traditional Chinese herbs combined with anti-inflammatory drugs might offer a promising strategy to prevent tumor metastasis.
Collapse
Affiliation(s)
- Rui Liu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Weidong Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Qiujun Guo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shulin He
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yoshiro Hirasaki
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Department of Japanese-Oriental (Kampo) Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | - Wei Hou
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Conghuang Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yebo Gao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China. .,Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xin Qi
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yingxia Pei
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yun Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Number 5 Beixiange, Xicheng District, Beijing, 100053, China. .,Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
34
|
Yeh DW, Chen YS, Lai CY, Liu YL, Lu CH, Lo JF, Chen L, Hsu LC, Luo Y, Xiang R, Chuang TH. Downregulation of COMMD1 by miR-205 promotes a positive feedback loop for amplifying inflammatory- and stemness-associated properties of cancer cells. Cell Death Differ 2015; 23:841-52. [PMID: 26586569 PMCID: PMC4832103 DOI: 10.1038/cdd.2015.147] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 09/22/2015] [Accepted: 10/02/2015] [Indexed: 12/16/2022] Open
Abstract
Sustained activation of nuclear factor-κB (NF-κB) in cancer cells has been shown to promote inflammation, expansion of cancer stem cell (CSC) population, and tumor development. In contrast, recent studies reveal that CSCs exhibit increased inflammation due to constitutive NF-κB activation; however, the underlying molecular mechanism remains unclear. In the present study, the analysis of microarray data revealed upregulation of NF-κB-regulated pro-inflammatory genes and downregulation of copper metabolism MURR1 domain-containing 1 (COMMD1) during the enrichment for stemness in SAS head and neck squamous-cell carcinoma (HNSCC) cells. The 3′-UTR of COMMD1 mRNA contains microRNA (miR)-205 target site. Parallel studies with HNSCC and NSCLC cells indicated that miR-205 is upregulated upon NF-κB activation and suppresses COMMD1 expression in stemness-enriched cancer cells. COMMD1 negatively regulates the inflammatory responses induced by TLR agonists, IL-1β, and TNF-α by targeting RelA for degradation. The shRNA-mediated downregulation of COMMD1 in cancer cells enhanced inflammatory response, generating favorable conditions for macrophage recruitment. In addition, genes associated with stemness were also upregulated in these cells, which exhibited increased potential for anchorage-independent growth. Furthermore, COMMD1 downregulation promoted in vivo tumorigenesis and tumor growth, and tumors derived from COMMD1-knockdown cells displayed elevated level of NF-κB activation, increased expression of inflammatory- and stemness-associated genes, and contain expanded population of tumor-associated leukocytes and stemness-enriched cancer cells. These results suggest that COMMD1 downregulation by miR-205 promotes tumor development by modulating a positive feedback loop that amplifies inflammatory- and stemness-associated properties of cancer cells.
Collapse
Affiliation(s)
- D-W Yeh
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan.,Institute of Molecular Medicine, National Tsing-Hua University, Hsinchu, Taiwan
| | - Y-S Chen
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - C-Y Lai
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Y-L Liu
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - C-H Lu
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - J-F Lo
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - L Chen
- Institute of Molecular Medicine, National Tsing-Hua University, Hsinchu, Taiwan
| | - L-C Hsu
- Institute of Molecular Medicine, National Taiwan University, Taipei, Taiwan
| | - Y Luo
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - R Xiang
- School of Medicine, University of Nankai, Tianjin, PR China
| | - T-H Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan.,Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Research and Development Center for Immunology, China Medical University, Taichung, Taiwan
| |
Collapse
|
35
|
Caldeira PC, de Andrade Sousa A, de Aguiar MCF. Differential infiltration of neutrophils in T1-T2 versus T3-T4 oral squamous cell carcinomas: a preliminary study. BMC Res Notes 2015; 8:569. [PMID: 26467671 PMCID: PMC4607015 DOI: 10.1186/s13104-015-1541-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/05/2015] [Indexed: 12/17/2022] Open
Abstract
Background Recent studies have pointed towards a role of tumour-infiltrating neutrophils in cancer biology. Investigations on oral squamous cell carcinoma have indicated a possible association with clinical characteristics. This study aimed to evaluate neutrophil infiltration and the neutrophil/lymphocyte ratio in the central areas and invasive front of oral squamous cell carcinomas at different T stages, and their association with clinicopathological features and patient outcome. Methods Clinical information was retrieved from the charts of patients who had undergone surgical treatment. Samples of the excised tumours were subjected to immunohistochemical analysis for CD66b and CD3. Semi-quantitative analysis was performed in the intratumoural region and in the invasive front. Appropriate statistical tests were used for evaluating the data, including Kaplan–Meier survival analysis and the log-rank test. A p value of less than 0.05 was considered significant. Results T3–T4 tumours presented higher CD66b infiltration in the intratumoural region and higher CD66b/CD3 ratios in the invasive front than T1–T2 lesions (p < 0.05). There was a strong inverse correlation between CD66b and CD3 in the invasive front of T3–T4 tumours (r = −0.712, p < 0.05). Comparisons of CD66b and the CD66b/CD3 ratio according to N status, tumour location, recurrence, inflammation grade, and histological grade did not reach statistical significance. Survival analysis also did not show any significant differences. Conclusions The present study showed different degrees of neutrophil infiltration between T1–T2 and T3–T4 oral cancers, with higher indexes in the advanced lesions. However, there was no association with clinicopathological features or with time to recurrence. Electronic supplementary material The online version of this article (doi:10.1186/s13104-015-1541-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patrícia Carlos Caldeira
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, CEP: 31.270-901, Belo Horizonte, MG, Brazil.
| | - Alexandre de Andrade Sousa
- Department of Surgery of the School of Medicine and Head and Neck Surgery, Instituto Alfa de Gastroenterologia, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Maria Cássia Ferreira de Aguiar
- Department of Oral Pathology and Surgery, School of Dentistry, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, CEP: 31.270-901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
36
|
Lu X, Luo F, Liu Y, Zhang A, Li J, Wang B, Xu W, Shi L, Liu X, Lu L, Liu Q. The IL-6/STAT3 pathway via miR-21 is involved in the neoplastic and metastatic properties of arsenite-transformed human keratinocytes. Toxicol Lett 2015; 237:191-9. [DOI: 10.1016/j.toxlet.2015.06.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/10/2015] [Accepted: 06/13/2015] [Indexed: 12/19/2022]
|
37
|
Xue G, Zippelius A, Wicki A, Mandala M, Tang F, Massi D, Hemmings BA. Integrated Akt/PKB Signaling in Immunomodulation and Its Potential Role in Cancer Immunotherapy. J Natl Cancer Inst 2015; 107:djv171. [DOI: 10.1093/jnci/djv171] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/22/2015] [Indexed: 12/17/2022] Open
|
38
|
Choi YJ, Kim N, Chang H, Lee HS, Park SM, Park JH, Shin CM, Kim JM, Kim JS, Lee DH, Jung HC. Helicobacter pylori-induced epithelial-mesenchymal transition, a potential role of gastric cancer initiation and an emergence of stem cells. Carcinogenesis 2015; 36:553-563. [DOI: 10.1093/carcin/bgv022] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
39
|
Is salivary epidermal growth factor a biomarker for oral leukoplakia? A preliminary study. Oral Surg Oral Med Oral Pathol Oral Radiol 2015; 119:451-8. [DOI: 10.1016/j.oooo.2014.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 01/27/2023]
|
40
|
Tan W, Madhavan K, Hunter KS, Park D, Stenmark KR. Vascular stiffening in pulmonary hypertension: cause or consequence? (2013 Grover Conference series). Pulm Circ 2014; 4:560-80. [PMID: 25610594 PMCID: PMC4278618 DOI: 10.1086/677370] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/27/2014] [Indexed: 12/24/2022] Open
Abstract
Recent studies have indicated that systemic arterial stiffening is a precursor to hypertension and that hypertension, in turn, can perpetuate arterial stiffening. Pulmonary artery (PA) stiffening is also well documented to occur in pulmonary hypertension (PH), and there is evidence that pulmonary vascular stiffness (PVS) may be a better predictor of outcome than pulmonary vascular resistance (PVR). We have hypothesized that the decreased flow-damping function of elastic PAs in PH likely initiates and/or perpetuates dysfunction of pulmonary microvasculature. Recent studies have shown that large-vessel stiffening increases flow pulsatility in the distal pulmonary vasculature, leading to endothelial dysfunction within a proinflammatory, vasoconstricting, and profibrogenic environment. The intricate role of stiffening-stimulated high pulsatile flow in endothelial cell dysfunction includes stepwise molecular events underlying PA hypertrophy, inflammation, endothelial-mesenchymal transition, and fibrosis. In addition to contributing to microenvironmental alterations of the distal vasculature, disordered proximal-distal PA coupling likely also plays a role in increasing ventricular afterload, ultimately causing right ventricle (RV) dysfunction and death. Current therapeutic treatments do not provide a realistic approach to destiffening arteries and, thus, to potentially abrogating the effects of high pulsatile flow on the distal pulmonary vasculature or the increased work imposed by stiffening on the RV. Scrutinizing the effect of PA stiffening on high pulsatile flow-induced cellular and molecular changes, and vice versa, might lead to important new therapeutic options that abrogate PA remodeling and PH development. With a clear understanding that PA stiffening may contribute to the progression of PH to an irreversible state by contributing to chronic microvascular damage in lungs, future studies should be aimed first at defining the underlying mechanisms leading to PA stiffening and then at improved treatment approaches based on these findings.
Collapse
Affiliation(s)
- Wei Tan
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, USA
- Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver, Aurora, Colorado, USA
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado, USA
| | - Krishna Madhavan
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, USA
- Department of Bioengineering, University of Colorado Denver, Aurora, Colorado, USA
| | - Kendall S. Hunter
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, USA
- Department of Bioengineering, University of Colorado Denver, Aurora, Colorado, USA
| | - Daewon Park
- Department of Bioengineering, University of Colorado Denver, Aurora, Colorado, USA
| | - Kurt R. Stenmark
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, USA
- Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
41
|
Liu Y, Luo F, Xu Y, Wang B, Zhao Y, Xu W, Shi L, Lu X, Liu Q. Epithelial-mesenchymal transition and cancer stem cells, mediated by a long non-coding RNA, HOTAIR, are involved in cell malignant transformation induced by cigarette smoke extract. Toxicol Appl Pharmacol 2014; 282:9-19. [PMID: 25447409 DOI: 10.1016/j.taap.2014.10.022] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/27/2014] [Accepted: 10/31/2014] [Indexed: 12/24/2022]
Abstract
The incidence of lung diseases, including cancer, caused by cigarette smoke is increasing, but the molecular mechanisms of gene regulation induced by cigarette smoke remain unclear. This report describes a long noncoding RNA (lncRNA) that is induced by cigarette smoke extract (CSE) and experiments utilizing lncRNAs to integrate inflammation with the epithelial-mesenchymal transition (EMT) in human bronchial epithelial (HBE) cells. The present study shows that, induced by CSE, IL-6, a pro-inflammatory cytokine, leads to activation of STAT3, a transcription activator. A ChIP assay determined that the interaction of STAT3 with the promoter regions of HOX transcript antisense RNA (HOTAIR) increased levels of HOTAIR. Blocking of IL-6 with anti-IL-6 antibody, decreasing STAT3, and inhibiting STAT3 activation reduced HOTAIR expression. Moreover, for HBE cells cultured in the presence of HOTAIR siRNA for 24h, the CSE-induced EMT, formation of cancer stem cells (CSCs), and malignant transformation were reversed. Thus, IL-6, acting on STAT3 signaling, which up-regulates HOTAIR in an autocrine manner, contributes to the EMT and to CSCs induced by CSE. These data define a link between inflammation and EMT, processes involved in the malignant transformation of cells caused by CSE. This link, mediated through lncRNAs, establishes a mechanism for CSE-induced lung carcinogenesis.
Collapse
Affiliation(s)
- Yi Liu
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China
| | - Fei Luo
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China
| | - Yuan Xu
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China
| | - Bairu Wang
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China
| | - Yue Zhao
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China
| | - Wenchao Xu
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China
| | - Le Shi
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China
| | - Xiaolin Lu
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China
| | - Qizhan Liu
- Institute of Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, P. R. China.
| |
Collapse
|
42
|
He Z, Deng R, Huang X, Ni Y, Yang X, Wang Z, Hu Q. Lipopolysaccharide enhances OSCC migration by promoting epithelial-mesenchymal transition. J Oral Pathol Med 2014; 44:685-92. [PMID: 25367215 DOI: 10.1111/jop.12285] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2014] [Indexed: 01/19/2023]
Abstract
BACKGROUND This study was performed to examine whether lipopolysaccharide can influence cell migration and epithelial-mesenchymal transition of oral squamous cell carcinoma. METHODS Three oral squamous cell carcinoma cell lines (HSC3, CAL27, and SCC4) were obtained for the study. TLR4 expression in three cell lines was analyzed by Q-PCR and Western blot. After cells treated with LPS, cell migration was analyzed by wound-healing and chemotaxis cell migration assay. Changes of E-cadherin and vimentin expression were tested by Western blot and immunofluorescence staining. To examine NF-κB activation, NF-κB nuclear translocation was investigated. RESULTS TLR4 was expressed in all three cell lines and was highest in HSC3 while lowest in SCC4. TLR4 ligand lipopolysaccharide accelerated wound healing and enhanced cell migration. Also, it stimulated epithelial-mesenchymal transition demonstrated by decreased E-cadherin and increased vimentin expression. Lipopolysaccharide also provoked NF-κB nuclear translocation. Either TLR4 or NF-κB blocking reverted these effects. CONCLUSIONS Lipopolysaccharide can induce TLR4-mediated epithelial-mesenchymal transition and cell migration in oral squamous cell carcinoma. These responses could further affect tumor progressing by inducing tumor cell metastasis.
Collapse
Affiliation(s)
- Zhifeng He
- Central Laboratory of Stomatology, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Runzhi Deng
- Department of Oral and Maxillofacial Surgery, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xiaofeng Huang
- Central Laboratory of Stomatology, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yanhong Ni
- Central Laboratory of Stomatology, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xudong Yang
- Department of Oral and Maxillofacial Surgery, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Zhiyong Wang
- Department of Oral and Maxillofacial Surgery, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
43
|
Chen W, Gao Q, Han S, Pan F, Fan W. The CCL2/CCR2 axis enhances IL-6-induced epithelial-mesenchymal transition by cooperatively activating STAT3-Twist signaling. Tumour Biol 2014; 36:973-81. [PMID: 25318604 DOI: 10.1007/s13277-014-2717-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 10/07/2014] [Indexed: 12/18/2022] Open
Abstract
The pattern of secreted factors in the tumor microenvironment has been shown to initiate tumor epithelial-mesenchymal transition (EMT); however, little is known about their interplay undergoing this phenotypic switch. In this study, we revealed obvious coactions of cytokine IL-6 and chemokine CCL2 during EMT induction. We found that IL-6 effectively induced EMT and promoted tumor cell invasion, which could be markedly enhanced by addition of CCL2 in a CCR2-dependent manner. IL-6 and CCL2 induced each other and cooperatively elicited STAT3 phosphorylation; conversely, STAT3 regulated the production of IL-6 and CCL2, thus constituting a positive feedback loop to maintain and amplify STAT3 signaling, consequently promoting additional EMT events. Furthermore, CCL2 greatly enhanced IL-6-induced EMT events mainly by upregulating the expression of Twist. Genetic or pharmacological inhibition of STAT3 disrupted STAT3-centered loop and markedly suppressed Twist expression as well as IL-6/CCL2-mediated EMT induction. Thus, our findings highlighted the synergy of the two secreted factors of tumor microenvironment, in regulating transformed properties of non-small cell lung cancer (NSCLC).
Collapse
Affiliation(s)
- Wei Chen
- Department of Respiratory, Navy General Hospital, Beijing, 100048, China
| | | | | | | | | |
Collapse
|
44
|
Yang L, Han S, Sun Y. An IL6-STAT3 loop mediates resistance to PI3K inhibitors by inducing epithelial-mesenchymal transition and cancer stem cell expansion in human breast cancer cells. Biochem Biophys Res Commun 2014; 453:582-7. [PMID: 25285632 DOI: 10.1016/j.bbrc.2014.09.129] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 09/29/2014] [Indexed: 01/20/2023]
Abstract
Recently, a new generation of PI3K-specific inhibitors, such as GDC0941 and BKM120, are being investigated in clinical trials for treatment against tumors harboring PIK3CA mutations. Nevertheless, not all patients benefit from such treatment, suggesting that their tumors may be resistant to PI3K inhibitors. The investigation of the underlying mechanisms and efficacious personalized treatment remain a large unmet need. In this study, we revealed an IL6-STAT3 positive feedback loop that mediated the resistance to PI3K inhibitors. We found that breast cancer cells with acquired resistance to PI3K inhibitors displayed epithelial-mesenchymal transition (EMT) features and an highly enriched cancer stem cells (CSCs), secreting ∼1000-fold more IL6 than parental cells. Further studies elucidated that activation of the IL6-STAT3 signaling effectively triggered EMT action, expanded the CSCs population, and reduced sensitivity to PI3K inhibitors. Pharmacological inhibition of STAT3 disrupted the IL6-STAT3 signaling and overcome resistance to PI3K inhibitors partially due to increased apoptosis induction. Taken together, our results demonstrated that feedback activation of the IL6-STAT3 loop lead to acquired resistance to PI3K inhibitors by promoting EMT and CSC-like features, and suggested that targeting this loop may be an efficient strategy to overcome resistance to PI3K inhibitors.
Collapse
Affiliation(s)
- Lin Yang
- Department of Clinical Laboratory, Hubei Maternal and Child Health Hospital, Wuhan 430070, China
| | - Siqi Han
- Department of Medical Oncology, Jinling Hospital, Nanjing 210002, China
| | - Yong Sun
- Department of Burn and Plastic Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an 223300, China.
| |
Collapse
|
45
|
Lu Y, Liu C, Xu YF, Cheng H, Shi S, Wu CT, Yu XJ. Stathmin destabilizing microtubule dynamics promotes malignant potential in cancer cells by epithelial-mesenchymal transition. Hepatobiliary Pancreat Dis Int 2014; 13:386-94. [PMID: 25100123 DOI: 10.1016/s1499-3872(14)60038-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Stathmin is a ubiquitous cytosolic regulatory phosphoprotein and is overexpressed in different human malignancies. The main physiological function of stathmin is to interfere with microtubule dynamics by promoting depolymerization of microtubules or by preventing polymerization of tubulin heterodimers. Stathmin plays important roles in regulating many cellular functions as a result of its microtubule-destabilizing activity. Currently, the critical roles of stathmin in cancer cells, as well as in lymphocytes have been valued. This review discusses stathmin and microtubule dynamics in cancer development, and hypothesizes their possible relationship with epithelial-mesenchymal transition (EMT). DATA SOURCES A PubMed search using such terms as "stathmin", "microtubule dynamics", "epithelial-mesenchymal transition", "EMT", "malignant potential" and "cancer" was performed to identify relevant studies published in English. More than 100 related articles were reviewed. RESULTS The literature clearly documented the relationship between stathmin and its microtubule-destabilizing activity of cancer development. However, the particular mechanism is poorly understood. Microtubule disruption is essential for EMT, which is a crucial process during cancer development. As a microtubule-destabilizing protein, stathmin may promote malignant potential in cancer cells by initiating EMT. CONCLUSIONS We propose that there is a stathmin-microtubule dynamics-EMT (S-M-E) axis during cancer development. By this axis, stathmin together with its microtubule-destabilizing activity contributes to EMT, which stimulates the malignant potential in cancer cells.
Collapse
Affiliation(s)
- Yu Lu
- Pancreatic Cancer Institute, Fudan University; Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | | | | | | | | | | | | |
Collapse
|
46
|
Insights into the role of components of the tumor microenvironment in oral carcinoma call for new therapeutic approaches. Exp Cell Res 2014; 325:58-64. [DOI: 10.1016/j.yexcr.2013.12.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 12/28/2013] [Accepted: 12/31/2013] [Indexed: 11/19/2022]
|
47
|
Sun Y, Song GD, Sun N, Chen JQ, Yang SS. Slug overexpression induces stemness and promotes hepatocellular carcinoma cell invasion and metastasis. Oncol Lett 2014; 7:1936-1940. [PMID: 24932263 PMCID: PMC4049749 DOI: 10.3892/ol.2014.2037] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 02/04/2014] [Indexed: 02/06/2023] Open
Abstract
Detection of metastasis of hepatocellular carcinoma (HCC) is crucial for early diagnosis. Epithelial-mesenchymal transition (EMT) is a common event in the metastasis of tumor cells. Slug and Snail are homologous proteins, which play an important role in EMT. The present study aimed to investigate whether Slug and Snail overexpression is associated with the invasiveness of HCC in vitro and in vivo. Invasion, colony formation and wound healing assays, as well as flow cytometry analysis, were performed to examine the invasiveness and proliferation capabilities of HepG2 cells following transfection with cNDA or the siRNA of Slug or Snail. The effects of Slug on HCC in vivo were examined using a xenograft model. Slug upregulation increased the percentage of cluster of differentiation (CD)133+ cells among HepG2 cells, and induced cell invasion and proliferation; whereas Snail upregulation did not affect the cells in vitro. The Slug overexpression group exhibited the highest rate of tumor growth compared with the Snail overexpression and control groups in vivo. These findings demonstrated that Slug increases the percentage of CD133+ cells, promotes the clonigenicity of HCC cells and induces a stronger stemness in Slug-overexpressing cells. These changes activate dormant developmental pathways in invading tumor cells. Thus, Slug may serve as a novel target for HCC prognosis and therapy.
Collapse
Affiliation(s)
- Yu Sun
- Department of Surgery, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Guo-Dong Song
- Department of Surgery, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Ning Sun
- Department of Surgery, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Jian-Qiu Chen
- Department of Surgery, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Shao-Shi Yang
- Department of Surgery, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| |
Collapse
|
48
|
Arsenite evokes IL-6 secretion, autocrine regulation of STAT3 signaling, and miR-21 expression, processes involved in the EMT and malignant transformation of human bronchial epithelial cells. Toxicol Appl Pharmacol 2013; 273:27-34. [DOI: 10.1016/j.taap.2013.08.025] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/25/2013] [Accepted: 08/26/2013] [Indexed: 01/26/2023]
|
49
|
Zhao Y, Xu Y, Li Y, Xu W, Luo F, Wang B, Pang Y, Xiang Q, Zhou J, Wang X, Liu Q. NF-κB-mediated inflammation leading to EMT via miR-200c is involved in cell transformation induced by cigarette smoke extract. Toxicol Sci 2013; 135:265-76. [PMID: 23824089 DOI: 10.1093/toxsci/kft150] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cigarette smoking constitutes a major human health hazard because it is the most important risk factor for lung cancer. Although evidence for smoking-induced lung cancer in humans is strong, the molecular mechanisms by which smoking causes cancer remain to be established. In this investigation, we evaluated the roles of inflammation and the epithelial-mesenchymal transition (EMT) in cigarette smoke extract (CSE)-induced transformation of human bronchial epithelial (HBE) cells. The results showed that chronic exposure to CSE induced EMT and transformation of these cells. Activation of nuclear factor-κB (NF-κB) by CSE increased levels of the proinflammatory interleukin-6 (IL-6), and acute and chronic exposures to CSE caused decreases in miR-200c levels. By blocking NF-κB with Bay11-7082 and IL-6 with anti-IL-6 antibody and enhancement of IL-6 with human recombinant IL-6, we found that the NF-κB signal pathway was involved in CSE-induced increases of IL-6, which suppressed miR-200c expression and promoted EMT. Moreover, IL-6 was necessary for maintenance of CSE-induced transformation and for malignant progression of HBE cells. Finally, blocking of NF-κB with Bay11-7082 prevented CSE-induced EMT and malignant transformation due to decreases of E-cadherin and miR-200c and elevations of IL-6, N-cadherin, and vimentin. Thus, we have defined a link between inflammation and EMT, processes involved in the malignant transformation of cells caused by CSE. This link, mediated through miRNAs, establishes a mechanism for CSE-induced lung carcinogenesis.
Collapse
|
50
|
Sinha N, Mukhopadhyay S, Das DN, Panda PK, Bhutia SK. Relevance of cancer initiating/stem cells in carcinogenesis and therapy resistance in oral cancer. Oral Oncol 2013; 49:854-862. [PMID: 23876628 DOI: 10.1016/j.oraloncology.2013.06.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 06/18/2013] [Accepted: 06/22/2013] [Indexed: 12/31/2022]
Abstract
Oral squamous cell carcinoma (OSCC) acquires the top most position among the other malignancies and patients die with this disease complication within 5years. One of the causes behind this scenario is the identified sub-population in heterogeneous tumor mass that are purported as cancer stem cells (CSCs) or tumor-initiating cells (TICs). Oral CSCs populations show upregulation of the stem cell related genes Oct-4, Nanog, Nestin, CK19, BMI-1, CD117 (c-kit), CD44 and CD133 with sunken expression of involucrin and CK13. This small proportion of tumor cells can sustain tumor growth, proliferation, invasion and distant metastasis playing a pivotal role in relapse of oral cancer. Unanimous risk factors include prevalent use of cigarette smoking, tobacco chewing with less explored HPV infection play an important role in origin of CSCs. Moreover, highly apoptotic resistant oral CSCs show enhanced protective autophagy for survival. Several studies report them to be more chemo and radiation resistant than non-stem cell population implicating the failure of the present cancer therapy. This resistance associated with normal stem cell protective mechanisms including increased expression of drug efflux pumps, alteration in program cell death, cell cycle, and DNA repair mechanisms. Notably, CSCs appear to play a major role in tumor recurrence and metastatic spread, common causes of the high morbidity and ultimately the death of the majority of patients with oral cancer. In this review we would like to highlight the intricate crosstalk of the cancer initiating/stem cells involved in carcinogenesis and potential hurdle to oral cancer therapy.
Collapse
Affiliation(s)
- Niharika Sinha
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Subhadip Mukhopadhyay
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Durgesh Nandini Das
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Prashanta Kumar Panda
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| | - Sujit K Bhutia
- Department of Life Science, National Institute of Technology Rourkela, Odisha, India.
| |
Collapse
|