101
|
Harada G, Bonadio RRDCC, de Araújo FCC, Victor CR, Sallum RAA, Junior UR, Cecconello I, Takeda FR, de Castria TB. Induction Chemotherapy for Locally Advanced Esophageal Cancer. J Gastrointest Cancer 2021; 51:498-505. [PMID: 31240598 DOI: 10.1007/s12029-019-00266-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Concurrent chemoradiotherapy followed by surgery is the standard treatment for locally advanced esophageal cancer (EC), and the role of induction chemotherapy (IC) remains unclear. We aimed to study if the addition of IC to standard treatment increases the rate of pathologic complete response (pCR). METHODS We assembled a retrospective analysis of patients (pts) diagnosed with locally advanced EC and treated with preoperative chemoradiotherapy followed by esophagectomy (CRT+S), preceded or not by IC, between 2009 and 2017. Patients' characteristics, tumor variables, and treatment outcomes were evaluated. The Kaplan-Meier method was used to estimate overall survival and the Cox proportional hazard model to evaluate prognostic factors. RESULTS One hundred and three patients were studied, with a median age of 62 years (range 37-84). Seventy-five patients (73%) were male, 67 (65%) had squamous cell carcinoma, and 31 (30%) had adenocarcinoma. Forty-three patients (41.7%) received IC followed by CRT+S (IC+CRT+S). The most frequent IC consisted of paclitaxel and platinum chemotherapy (90%), and the median number of cycles was 2. All patients received CRT+S. Concurrent chemotherapy was a combination of paclitaxel and platinum in 94 patients (91%). There was no statistically significant difference in pCR between the IC group and the standard CRT+S group. The pCR was 41.9% and 46.7% in the IC+CRT+S and CRT+S groups (p = 0.628), respectively. In the multivariate analysis, pCR was an independent prognostic factor for time to treatment failure (TTF) (HR 0.35, p = 0.021), but not for overall survival (OS) (p = 0.863). The factor that significantly affected OS in the multivariate analysis was positive lymph node (HR 5.9, 95%, p = 0.026). CONCLUSIONS Our data suggest that the addition of IC to standard CRT + S does not increase the pCR rate in locally advanced EC. No difference in OS was observed between pts. that received or not IC. Regardless of the treatment received, pts. achieving a pCR presented improved TTF.
Collapse
Affiliation(s)
- Guilherme Harada
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil.
- Centro de Oncologia, Hospital Sírio-Libanês, Sao Paulo, Brazil.
| | | | | | - Carolina Ribeiro Victor
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Rubens Antonio Aissar Sallum
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Division of Digestive Surgery - Department of Gastroenterology, São Paulo State Cancer Institute - ICESP-HCFMUSP, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Ulysses Ribeiro Junior
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Division of Digestive Surgery - Department of Gastroenterology, São Paulo State Cancer Institute - ICESP-HCFMUSP, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Ivan Cecconello
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Division of Digestive Surgery - Department of Gastroenterology, São Paulo State Cancer Institute - ICESP-HCFMUSP, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Flávio Roberto Takeda
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Division of Digestive Surgery - Department of Gastroenterology, São Paulo State Cancer Institute - ICESP-HCFMUSP, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Tiago Biachi de Castria
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Centro de Oncologia, Hospital Sírio-Libanês, Sao Paulo, Brazil
| |
Collapse
|
102
|
Tian Y, Li P, Xiao Z, Zhou J, Xue X, Jiang N, Peng C, Wu L, Tian H, Popper H, Poh ME, Marcucci F, Zhang C, Zhao X. Triptolide inhibits epithelial-mesenchymal transition phenotype through the p70S6k/GSK3/β-catenin signaling pathway in taxol-resistant human lung adenocarcinoma. Transl Lung Cancer Res 2021; 10:1007-1019. [PMID: 33718039 PMCID: PMC7947389 DOI: 10.21037/tlcr-21-145] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Chemotherapy is one of the primary treatments for both small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), however, chemoresistance develops over time and is a bottleneck to effective chemotherapy worldwide. Therefore, the development of new potent therapeutic agents to overcome chemoresistance is of utmost importance. Triptolide is a natural component extracted from Tripterygium Wilfordii, a Chinese plant; our study aimed to evaluate its anti-tumor effects in taxol-resistant human lung adenocarcinoma and investigate its molecular mechanisms of chemoresistance. Methods Triptolide’s inhibition of cell viability was detected by sulforhodamine B (SRB) assay. Cell cycle was measured by flow cytometry and cell apoptosis was assessed by flow cytometry and western blot. Expression of β-catenin was analyzed by western blot and immunofluorescence (IF). The anti-tumor effects of triptolide were determined using a subcutaneous in-vivo model. Cell proliferation and apoptosis were evaluated by immunohistochemistry (IHC) and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay, respectively. The expression level of p-p70S6K and p-GSK-3α/β was evaluated by western blot and IHC. Results Triptolide inhibited cell proliferation, induced S-phase cell cycle arrest and apoptosis in taxol-resistant A549 (A549/TaxR) cells. Moreover, intraperitoneal injection of triptolide resulted in a significant delay of tumor growth without obvious systemic toxicity in mice. Additionally, triptolide reversed epithelial-mesenchymal transition (EMT) through repression of the p70S6K/GSK3/β-catenin signaling pathway. Conclusions Our study provides evidence that triptolide can reverse EMT in taxol-resistant lung adenocarcinoma cells and impairs tumor growth by inhibiting the p70S6K/GSK3/β-catenin pathway, indicating that triptolide has potential to be used as a new therapeutic agent for taxol-resistant lung adenocarcinoma.
Collapse
Affiliation(s)
- Yu Tian
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peiwei Li
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhaohua Xiao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Zhou
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xia Xue
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ning Jiang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuanliang Peng
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Licun Wu
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, China.,Latner Thoracic Surgery Research Laboratories and Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Hui Tian
- Department of Thoracic Surgery, Cheeloo Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Helmut Popper
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Mau-Ern Poh
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Trentacoste 2, Milan, Italy
| | - Chengke Zhang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
103
|
Marcucci F, Rumio C. The tumor-promoting effects of the adaptive immune system: a cause of hyperprogressive disease in cancer? Cell Mol Life Sci 2021; 78:853-865. [PMID: 32940721 PMCID: PMC11072297 DOI: 10.1007/s00018-020-03606-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Adaptive antitumor immune responses, either cellular or humoral, aim at eliminating tumor cells expressing the cognate antigens. There are some instances, however, where these same immune responses have tumor-promoting effects. These effects can lead to the expansion of antigen-negative tumor cells, tumor cell proliferation and tumor growth, metastatic dissemination, resistance to antitumor therapy and apoptotic stimuli, acquisition of tumor-initiating potential and activation of various forms of survival mechanisms. We describe the basic mechanisms that underlie tumor-promoting adaptive immune responses and try to identify the variables that induce the switching of a tumor-inhibitory, cellular or humoral immune response, into a tumor-promoting one. We suggest that tumor-promoting adaptive immune responses may be at the origin of at least a fraction of hyperprogressive diseases (HPD) that are observed in cancer patients during therapy with immune checkpoint inhibitors (ICI) and, less frequently, with single-agent chemotherapy. We also propose the use of non-invasive biomarkers allowing to predict which patients may undergo HPD during ICI and other forms of antitumor therapy. Eventually, we suggest possibilities of therapeutic intervention allowing to inhibit tumor-promoting adaptive immune responses.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Trentacoste 2, Milan, Italy.
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Trentacoste 2, Milan, Italy
| |
Collapse
|
104
|
Qi Y, Yang W, Liu S, Han F, Wang H, Zhao Y, Zhou Y, Zhou D. Cisplatin loaded multiwalled carbon nanotubes reverse drug resistance in NSCLC by inhibiting EMT. Cancer Cell Int 2021; 21:74. [PMID: 33494783 PMCID: PMC7836500 DOI: 10.1186/s12935-021-01771-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/11/2021] [Indexed: 12/25/2022] Open
Abstract
Background Lung cancer is one of the important health threats worldwide, of which 5-year survival rate is less than 15%. Non-small-cell lung cancer (NSCLC) accounts for about 80% of all lung cancer with high metastasis and mortality. Methods Cisplatin loaded multiwalled carbon nanotubes (Pt-MWNTS) were synthesized and used to evaluate the anticancer effect in our study. The NSCLC cell lines A549 (cisplatin sensitive) and A549/DDP (cisplatin resistant) were used in our in vitro assays. MTT was used to determine Cancer cells viability and invasion were measured by MTT assay and Transwell assay, respectively. Apoptosis and epithelial-mesenchymal transition related marker proteins were measured by western blot. The in vivo anti-cancer effect of Pt-MWNTs were performed in male BALB/c nude mice (4-week old). Results Pt-MWNTS were synthesized and characterized by X-ray diffraction, Raman, FT-IR spectroscopy and scan electron microscopy. No significant cytotoxicity of MWNTS was detected in both A549/DDP and A549 cell lines. However, Pt-MWNTS showed a stronger inhibition effect on cell growth than free cisplatin, especially on A549/DDP. We found Pt-MWNTS showed higher intracellular accumulation of cisplatin in A549/DDP cells than free cisplatin and resulted in enhanced the percent of apoptotic cells. Western blot showed that application of Pt-MWNTS can significantly upregulate the expression level of Bax, Bim, Bid, Caspase-3 and Caspase-9 while downregulate the expression level of Bcl-2, compared with free cisplatin. Moreover, the expression level of mesenchymal markers like Vimentin and N-cadherin was more efficiently reduced by Pt-MWNTS treatment in A549/DDP cells than free cisplatin. In vivo study in nude mice proved that Pt-MWNTS more effectively inhibited tumorigenesis compared with cisplatin, although both of them had no significant effect on body weight. Conclusion Pt-MWNT reverses the drug resistance in the A549/DDP cell line, underlying its possibility of treating NSCLC with cisplatin resistance.
Collapse
Affiliation(s)
- Yuxin Qi
- Department of Respiratory Medicine, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan, 271199, China
| | - Wenping Yang
- Department of Respiratory Medicine, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan, 271199, China
| | - Shuang Liu
- Department of Respiratory Medicine, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan, 271199, China
| | - Fanjie Han
- Department of Respiratory Medicine, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan, 271199, China
| | - Haibin Wang
- Department of Respiratory Medicine, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan, 271199, China
| | - Yonghong Zhao
- Department of Respiratory Medicine, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan, 271199, China
| | - Yufa Zhou
- Department of Respiratory Medicine, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan, 271199, China
| | - Daijun Zhou
- Department of Oncology, General Hospital of Western Theater Command of PLA, Chengdu, 610083, China.
| |
Collapse
|
105
|
Yuan W, Tao R, Huang D, Yan W, Shen G, Ning Q. Transcriptomic characterization reveals prognostic molecular signatures of sorafenib resistance in hepatocellular carcinoma. Aging (Albany NY) 2021; 13:3969-3993. [PMID: 33495404 PMCID: PMC7906139 DOI: 10.18632/aging.202365] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/25/2020] [Indexed: 04/19/2023]
Abstract
Sorafenib is the first-line treatment for patients with advanced unresectable hepatocellular carcinoma (HCC); however, only a small number of patients benefit from sorafenib, and many develop sorafenib resistance (SR) and severe side effects. To identify biomarkers for SR, we systematically analyzed the molecular alterations in both sorafenib-resistant HCC specimens and cultured cells. By combining bioinformatics tools and experimental validation, four genes (C2orf27A, insulin-like growth factor 2 receptor, complement factor B, and paraoxonase 1) were identified as key genes related to SR in HCC and as independent prognostic factors significantly associated with clinical cancer stages and pathological tumor grades of liver cancer. These genes can affect the cytotoxicity of sorafenib to regulate the proliferation and invasion of Huh7 cells in vitro. Additionally, immune-cell infiltration according to tumor immune dysfunction and exclusion, a biomarker integrating the mechanisms of dysfunction and exclusion of T cells showed good predictive power for SR, with an AUC of 0.869. These findings suggest that immunotherapy may be a potential strategy for treating sorafenib-resistant HCC. Furthermore, the results enhance the understanding of the underlying molecular mechanisms of SR in HCC and will facilitate the development of precision therapy for patients with liver cancer.
Collapse
Affiliation(s)
- Wei Yuan
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Tao
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Da Huang
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Yan
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
106
|
GLI3 Promotes Invasion and Predicts Poor Prognosis in Colorectal Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8889986. [PMID: 33506047 PMCID: PMC7814942 DOI: 10.1155/2021/8889986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/02/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022]
Abstract
Purpose The epithelial–mesenchymal transition (EMT) is a key hallmark of cancer which promotes malignant progression, especially during the process of cancer invasion. A better understanding of EMT will help elucidate the molecular mechanism underlying colorectal cancer (CRC) metastasis and may provide new insights into the identification of potential biomarkers and therapeutic targets. Methods A series of bioinformatic approaches were combined and identify GLI3 as a potential key regulator in EMT. In vitro experiments were performed to knockdown GLI3 expression in two CRC cell lines and to reveal the oncogenic role of GLI3 in CRC. qRT-PCR and western blot were performed to show the influence of GLI3 in EMT and downstream pathways. The Kaplan-Meier analysis and log-rank test were used to evaluate the prognostic value of GLI3 in CRC patients. Results GLI3 was identified as a key regulator in coexpression and protein-protein interaction (PPI) networks involved in EMT. Bioinformatic analyses indicated that GLI3 had a high correlation with EMT markers in CRC. In vitro experiments showed that GLI3 knockdown attenuated the migratory and invasive capacities of CRC cells via influencing EMT property, especially by regulating phosphorylation of ERK signaling pathway. In addition, higher expression of GLI3 predicts worse prognosis in CRC patients. Conclusions In summary, we presented the first evidence that GLI3 could promote the migratory and invasive capacities of CRC cells by regulating the EMT process. Our study might provide some useful clues to a better understanding of GLI3 in EMT during CRC progression.
Collapse
|
107
|
Determining Factors in the Therapeutic Success of Checkpoint Immunotherapies against PD-L1 in Breast Cancer: A Focus on Epithelial-Mesenchymal Transition Activation. J Immunol Res 2021; 2021:6668573. [PMID: 33506060 PMCID: PMC7808819 DOI: 10.1155/2021/6668573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/17/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common neoplasm diagnosed in women around the world. Checkpoint inhibitors, targeting the programmed death receptor-1 or ligand-1 (PD-1/PD-L1) axis, have dramatically changed the outcome of cancer treatment. These therapies have been recently considered as alternatives for treatment of breast cancers, in particular those with the triple-negative phenotype (TNBC). A further understanding of the regulatory mechanisms of PD-L1 expression is required to increase the benefit of PD-L1/PD-1 checkpoint immunotherapy in breast cancer patients. In this review, we will compile the most recent studies evaluating PD-1/PD-L1 checkpoint inhibitors in breast cancer. We review factors that determine the therapeutic success of PD-1/PD-L1 immunotherapies in this pathology. In particular, we focus on pathways that interconnect the epithelial-mesenchymal transition (EMT) with regulation of PD-L1 expression. We also discuss the relationship between cellular metabolic pathways and PD-L1 expression that are involved in the promotion of resistance in TNBC.
Collapse
|
108
|
Marcucci F, Rumio C. Glycolysis-induced drug resistance in tumors-A response to danger signals? Neoplasia 2021; 23:234-245. [PMID: 33418276 PMCID: PMC7804361 DOI: 10.1016/j.neo.2020.12.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor cells often switch from mitochondrial oxidative metabolism to glycolytic metabolism even under aerobic conditions. Tumor cell glycolysis is accompanied by several nonenzymatic activities among which induction of drug resistance has important therapeutic implications. In this article, we review the main aspects of glycolysis-induced drug resistance. We discuss the classes of antitumor drugs that are affected and the components of the glycolytic pathway (transporters, enzymes, metabolites) that are involved in the induction of drug resistance. Glycolysis-associated drug resistance occurs in response to stimuli, either cell-autonomous (e.g., oncoproteins) or deriving from the tumor microenvironment (e.g., hypoxia or pseudohypoxia, mechanical cues, etc.). Several mechanisms mediate the induction of drug resistance in response to glycolytic metabolism: inhibition of apoptosis, induction of epithelial-mesenchymal transition, induction of autophagy, inhibition of drug influx and increase of drug efflux. We suggest that drug resistance in response to glycolysis comes into play in presence of qualitative (e.g., expression of embryonic enzyme isoforms, post-translational enzyme modifications) or quantitative (e.g., overexpression of enzymes or overproduction of metabolites) alterations of glycolytic metabolism. We also discern similarities between changes occurring in tumor cells in response to stimuli inducing glycolysis-associated drug resistance and those occurring in cells of the innate immune system in response to danger signals and that have been referred to as danger-associated metabolic modifications. Eventually, we briefly address that also mitochondrial oxidative metabolism may induce drug resistance and discuss the therapeutic implications deriving from the fact that the main energy-generating metabolic pathways may be both at the origin of antitumor drug resistance.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
109
|
Gil J, Jordà M, Soldevila B, Puig-Domingo M. Epithelial-Mesenchymal Transition in the Resistance to Somatostatin Receptor Ligands in Acromegaly. Front Endocrinol (Lausanne) 2021; 12:646210. [PMID: 33790868 PMCID: PMC8006574 DOI: 10.3389/fendo.2021.646210] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/22/2021] [Indexed: 01/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a dynamic process by which epithelial cells loss their phenotype and acquire mesenchymal traits, including increased migratory and invasive capacities. EMT is involved in physiological processes, such as embryogenesis and wound healing, and in pathological processes such as cancer, playing a pivotal role in tumor progression and metastasis. Pituitary tumors, although typically benign, can be locally invasive. Different studies have shown the association of EMT with increased tumor size and invasion in pituitary tumors, and in particular with a poor response to Somatostatin Receptor Ligands (SRLs) treatment in GH-producing pituitary tumors, the main cause of acromegaly. This review will summarize the current knowledge regarding EMT and SRLs resistance in acromegaly and, based on this relation, will suggest new biomarkers and possible therapies to SRLs resistant tumors.
Collapse
Affiliation(s)
- Joan Gil
- Endocrine Tumours Lab, Program of Predictive and Personalized Medicine of Cancer (PMPPC), Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Mireia Jordà
- Endocrine Tumours Lab, Program of Predictive and Personalized Medicine of Cancer (PMPPC), Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- *Correspondence: Manel Puig-Domingo, ; Mireia Jordà,
| | - Berta Soldevila
- Department of Endocrinology and Nutrition, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Manel Puig-Domingo
- Endocrine Tumours Lab, Program of Predictive and Personalized Medicine of Cancer (PMPPC), Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Endocrinology and Nutrition, Germans Trias i Pujol University Hospital, Badalona, Spain
- Department of Medicine, Autonomous University of Barcelona, Bellaterra, Spain
- *Correspondence: Manel Puig-Domingo, ; Mireia Jordà,
| |
Collapse
|
110
|
Wernitznig D, Meier-Menches SM, Cseh K, Theiner S, Wenisch D, Schweikert A, Jakupec MA, Koellensperger G, Wernitznig A, Sommergruber W, Keppler BK. Plecstatin-1 induces an immunogenic cell death signature in colorectal tumour spheroids. Metallomics 2020; 12:2121-2133. [PMID: 33295928 DOI: 10.1039/d0mt00227e] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Organometallic metal(arene) anticancer agents were believed to confer low selectivity for potential cellular targets. However, the ruthenium(arene) pyridinecarbothioamide (plecstatin-1) showed target selectivity for plectin, a scaffold protein and cytolinker. We employed a three-dimensional cancer spheroid model and showed that plecstatin-1 limited spheroid growth, induced changes in the morphology and in the architecture of tumour spheroids by disrupting the cytoskeletal organization. Additionally, we demonstrated that plecstatin-1 induced oxidative stress, followed by the induction of an immunogenic cell death signature through phosphorylation of eIF2α, exposure of calreticulin, HSP90 and HSP70 on the cell membrane and secretion of ATP followed by release of high mobility group box-1.
Collapse
Affiliation(s)
- Debora Wernitznig
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Li W, Yan P, Meng X, Zhang J, Yang Y. The microRNA cluster miR-214/miR-3120 prevents tumor cell switching from an epithelial to a mesenchymal-like phenotype and inhibits autophagy in gallbladder cancer. Cell Signal 2020; 80:109887. [PMID: 33340658 DOI: 10.1016/j.cellsig.2020.109887] [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: 07/20/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Tumor cells switch from an epithelial to a mesenchymal-like phenotype, which represents a key hallmark of human cancer metastasis, including gallbladder cancer (GBC). A large set of microRNAs (miRNAs/miRs) have been studied to elucidate their functions in initiating or inhibiting this phenotypic switching in GBC cells. In this paper, we attempted to identify the expression pattern of the miR-214/-3120 cluster and its mode of action in the context of GBC, with a specific focus being placed on their effects on EMT and autophagy in GBC cells. Human GBC cells GBC-SD were assayed for their migration, invasion, and autophagy using the Transwell chamber system, MDC staining, and transmission electron microscopy. The tumorigenicity and metastatic behavior of GBC-SD cells were tested in nude mice. The expression of EMT- and autophagy-specific markers (E-cadherin, N-cadherin, vimentin, ATG5, LC3II/LC3I, and Beclin1) was analyzed in cultured GBC-SD cells and in human GBC-SD xenografts. The E2F3 luciferase reporter activity in the presence of miR-214/-3120 was evaluated by a dual luciferase assay. The miR-214/-3120 was downregulated in GBC. Exogenous miR-214/-3120 inhibited the phenotypic switching of GBC cells from epithelial to mesenchymal, prevented autophagy, and suppressed the tumorigenicity and metastatic behavior of GBC-SD cells in vitro and in vivo. E2F3 was demonstrated to be the target gene of miR-214/-3120, and its knockdown in part mimicked the effect of miR-214/-3120 on the EMT, autophagy, tumorigenicity, and metastatic behavior of GBC-SD cells. These results demonstrated that the miR-214/-3120 cluster blocks the process of EMT and autophagy to limit GBC metastasis by repressing E2F3 expression.
Collapse
Affiliation(s)
- Wujun Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China; Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, PR China
| | - Pu Yan
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, PR China
| | - Xiaofen Meng
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China
| | - Jinpei Zhang
- Department of Encephalopathy, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, PR China.
| | - Yi Yang
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, PR China.
| |
Collapse
|
112
|
Cai L, Hu C, Yu S, Liu L, Zhao J, Zhao Y, Lin F, Du X, Yu Q, Xiao Q. Identification of EMT-Related Gene Signatures to Predict the Prognosis of Patients With Endometrial Cancer. Front Genet 2020; 11:582274. [PMID: 33343628 PMCID: PMC7738567 DOI: 10.3389/fgene.2020.582274] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background Endometrial cancer (EC) is one of the most common gynecological cancers. Epithelial–mesenchymal transition (EMT) is believed to be significantly associated with the malignant progression of tumors. However, there is no relevant study on the relationship between EMT-related gene (ERG) signatures and the prognosis of EC patients. Methods We extracted the mRNA expression profiles of 543 tumor and 23 normal tissues from The Cancer Genome Atlas database. Then, we selected differentially expressed ERGs (DEERGs) among these mRNAs. Next, univariate and multivariate Cox regression analyses were performed to select the ERGs with predictive ability for the prognosis of EC patients. In addition, risk score models were constructed based on the selected genes to predict patients’ overall survival (OS), progression-free survival (PFS), and disease-free survival (DFS). Finally, nomograms were constructed to estimate the OS and PFS of EC patients, and pan-cancer analysis was performed to further analyze the functions of a certain gene. Results Six OS-, ten PFS-, and five DFS-related ERGs were obtained. By constructing the prognostic risk score model, we found that the OS, PFS, and DFS of the high-risk group were notably poorer. Last, we found that AQP5 appeared in all three gene signatures, and through pan-cancer analysis, it was also found to play an important role in immunity in lower grade glioma (LGG), which may contribute to the poor prognosis of LGG patients. Conclusions We constructed ERG signatures to predict the prognosis of EC patients using bioinformatics methods. Our findings provide a thorough understanding of the effect of EMT in patients with EC and provide new targets and ideas for individualized treatment, which has important clinical significance.
Collapse
Affiliation(s)
- Luya Cai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chuan Hu
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shanshan Yu
- Department of Chemoradiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lixiao Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinduo Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ye Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fan Lin
- Department of Dermatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuedan Du
- Department of Chemoradiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiongjie Yu
- Department of Chemoradiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qinqin Xiao
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
113
|
Dong B, Qiu Z, Wu Y. Tackle Epithelial-Mesenchymal Transition With Epigenetic Drugs in Cancer. Front Pharmacol 2020; 11:596239. [PMID: 33343366 PMCID: PMC7746977 DOI: 10.3389/fphar.2020.596239] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/20/2020] [Indexed: 02/03/2023] Open
Abstract
Epithelial-mesenchymal Transition (EMT) is a de-differentiation process in which epithelial cells lose their epithelial properties to acquire mesenchymal features. EMT is essential for embryogenesis and wound healing but is aberrantly activated in pathological conditions like fibrosis and cancer. Tumor-associated EMT contributes to cancer cell initiation, invasion, metastasis, drug resistance and recurrence. This dynamic and reversible event is governed by EMT-transcription factors (EMT-TFs) with epigenetic complexes. In this review, we discuss recent advances regarding the mechanisms that modulate EMT in the context of epigenetic regulation, with emphasis on epigenetic drugs, such as DNA demethylating reagents, inhibitors of histone modifiers and non-coding RNA medication. Therapeutic contributions that improve epigenetic regulation of EMT will translate the clinical manifestation as treating cancer progression more efficiently.
Collapse
Affiliation(s)
- Bo Dong
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, KY, United States,Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY, United States
| | - Zhaoping Qiu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, KY, United States,Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY, United States
| | - Yadi Wu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, KY, United States,Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY, United States,*Correspondence: Yadi Wu,
| |
Collapse
|
114
|
Shinada M, Saeki K, Yoshitake R, Eto S, Tsuboi M, Chambers JK, Uchida K, Kato D, Yoshimoto S, Kamoto S, Ikeda N, Kinoshita R, Fujita N, Nishimura R, Nakagawa T. Evaluation of epithelial and mesenchymal cell markers in canine urinary bladder transitional cell carcinoma. Vet J 2020; 266:105571. [PMID: 33323173 DOI: 10.1016/j.tvjl.2020.105571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 10/21/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Canine transitional cell carcinoma (cTCC) is the most common malignant tumour in the urinary bladder: it is highly invasive and exhibits metastatic characteristics. Inflammation is also strongly related to cTCC. Epithelial tumours often exhibit a mesenchymal cell phenotype during tumour invasion and metastasis owing to epithelial-mesenchymal transition (EMT), which is often induced in chronic inflammation. The aim of this retrospective study was to investigate the expression of epithelial and mesenchymal cell markers in tumour cells and to evaluate its relationship with prognosis of cTCC. In this study, 29 dogs with cTCC who underwent surgical treatment were enrolled. Clinical parameters were reviewed using medical records. Tissue expression of epithelial and mesenchymal markers was evaluated by immunohistochemical analysis. The association between the expression of mesenchymal cell markers and clinical parameters, including prognosis, was statistically examined. In five normal bladder tissues used as controls, no expression of mesenchymal markers was observed, except for one tissue that expressed fibronectin. Conversely, epithelial tumour cells expressed vimentin and fibronectin in 23/29 and 19/28 cTCC tissues, respectively. Regarding clinical parameters, vimentin score in Miniature Dachshunds was significantly higher than those in other dog breeds (P < 0.001). Multivariate survival analyses revealed that age>12 years was related to shorter progression-free survival (P = 0.02). Higher vimentin score, lower fibronectin score, and advanced clinical T stage were significantly correlated with shorter median survival time (P < 0.05). The results of this study indicate that vimentin expression was associated with cTCC progression. Further studies are needed to examine the incidence and relevance of EMT in cTCC.
Collapse
Affiliation(s)
- M Shinada
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - K Saeki
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| | - R Yoshitake
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - S Eto
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - M Tsuboi
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - J K Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - K Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - D Kato
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - S Yoshimoto
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - S Kamoto
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - N Ikeda
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - R Kinoshita
- Veterinary Medical Center, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - N Fujita
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - R Nishimura
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - T Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
115
|
Rodríguez-Alonso A, Casas-Pais A, Roca-Lema D, Graña B, Romay G, Figueroa A. Regulation of Epithelial-Mesenchymal Plasticity by the E3 Ubiquitin-Ligases in Cancer. Cancers (Basel) 2020; 12:cancers12113093. [PMID: 33114139 PMCID: PMC7690828 DOI: 10.3390/cancers12113093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/08/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022] Open
Abstract
The epithelial-mesenchymal plasticity (EMP) is a process by which epithelial cells acquire the ability to dynamically switch between epithelial and mesenchymal phenotypic cellular states. Epithelial cell plasticity in the context of an epithelial-to-mesenchymal transition (EMT) confers increased cell motility, invasiveness and the ability to disseminate to distant sites and form metastasis. The modulation of molecularly defined targets involved in this process has become an attractive therapeutic strategy against cancer. Protein degradation carried out by ubiquitination has gained attention as it can selectively degrade proteins of interest. In the ubiquitination reaction, the E3 ubiquitin-ligases are responsible for the specific binding of ubiquitin to a small subset of target proteins, and are considered promising anticancer drug targets. In this review, we summarize the role of the E3 ubiquitin-ligases that control targeted protein degradation in cancer-EMT, and we highlight the potential use of the E3 ubiquitin-ligases as drug targets for the development of small-molecule drugs against cancer.
Collapse
Affiliation(s)
- Andrea Rodríguez-Alonso
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006 A Coruña, Spain; (A.R.-A.); (A.C.-P.); (D.R.-L.); (G.R.)
| | - Alba Casas-Pais
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006 A Coruña, Spain; (A.R.-A.); (A.C.-P.); (D.R.-L.); (G.R.)
| | - Daniel Roca-Lema
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006 A Coruña, Spain; (A.R.-A.); (A.C.-P.); (D.R.-L.); (G.R.)
| | - Begoña Graña
- Clinical Oncology Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006 A Coruña, Spain;
| | - Gabriela Romay
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006 A Coruña, Spain; (A.R.-A.); (A.C.-P.); (D.R.-L.); (G.R.)
| | - Angélica Figueroa
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006 A Coruña, Spain; (A.R.-A.); (A.C.-P.); (D.R.-L.); (G.R.)
- Correspondence:
| |
Collapse
|
116
|
Sistigu A, Musella M, Galassi C, Vitale I, De Maria R. Tuning Cancer Fate: Tumor Microenvironment's Role in Cancer Stem Cell Quiescence and Reawakening. Front Immunol 2020; 11:2166. [PMID: 33193295 PMCID: PMC7609361 DOI: 10.3389/fimmu.2020.02166] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cell dormancy is a common feature of human tumors and represents a major clinical barrier to the long-term efficacy of anticancer therapies. Dormant cancer cells, either in primary tumors or disseminated in secondary organs, may reawaken and relapse into a more aggressive disease. The mechanisms underpinning dormancy entry and exit strongly resemble those governing cancer cell stemness and include intrinsic and contextual cues. Cellular and molecular components of the tumor microenvironment persistently interact with cancer cells. This dialog is highly dynamic, as it evolves over time and space, strongly cooperates with intrinsic cell nets, and governs cancer cell features (like quiescence and stemness) and fate (survival and outgrowth). Therefore, there is a need for deeper insight into the biology of dormant cancer (stem) cells and the mechanisms regulating the equilibrium quiescence-versus-proliferation are vital in our pursuit of new therapeutic opportunities to prevent cancer from recurring. Here, we review and discuss microenvironmental regulations of cancer dormancy and its parallels with cancer stemness, and offer insights into the therapeutic strategies adopted to prevent a lethal recurrence, by either eradicating resident dormant cancer (stem) cells or maintaining them in a dormant state.
Collapse
Affiliation(s)
- Antonella Sistigu
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy.,Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Martina Musella
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Galassi
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo (TO), Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Ruggero De Maria
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| |
Collapse
|
117
|
Ozfiliz Kilbas P, Sonmez O, Uysal-Onganer P, Coker Gurkan A, Obakan Yerlikaya P, Arisan ED. Specific c-Jun N-Terminal Kinase Inhibitor, JNK-IN-8 Suppresses Mesenchymal Profile of PTX-Resistant MCF-7 Cells through Modulating PI3K/Akt, MAPK and Wnt Signaling Pathways. BIOLOGY 2020; 9:E320. [PMID: 33019717 PMCID: PMC7599514 DOI: 10.3390/biology9100320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/23/2020] [Accepted: 09/29/2020] [Indexed: 01/10/2023]
Abstract
Paclitaxel (PTX) is a widely used chemotherapeutic agent in the treatment of breast cancer, and resistance to PTX is a common failure of breast cancer therapy. Therefore, understanding the effective molecular targets in PTX-resistance gains importance in identifying novel strategies in successful breast cancer therapy approaches. The aim of the study was to investigate the functional role of PTX resistance on MCF-7 cell survival and proliferation related to PI3K/Akt and MAPK pathways. The generated PTX-resistant (PTX-res) MCF-7 cells showed enhanced cell survival, proliferation, and colony formation potential with decreased cell death compared to wt MCF-7 cells. PTX-res MCF-7 cells exhibited increased motility profile with EMT, PI3K/Akt, and MAPK pathway induction. According to the significant SAPK/JNK activation in PTX-res MCF-7 cells, specific c-Jun N-terminal kinase inhibitor, JNK-IN-8 is shown to suppress the migration potential of cells. Treatment of JNK inhibitor suppressed the p38 and SAPK/JNK and Vimentin expression. However, the JNK inhibitor further downregulated Wnt signaling members in PTX-res MCF-7 cells. Therefore, the JNK inhibitor JNK-IN-8 might be used as a potential therapy model to reverse PTX-resistance related to Wnt signaling.
Collapse
Affiliation(s)
- Pelin Ozfiliz Kilbas
- Department of Molecular Biology and Genetics, Istanbul Kultur University, 34158 Istanbul, Turkey; (P.O.K.); (O.S.); (A.C.G.); (P.O.Y.)
| | - Ozlem Sonmez
- Department of Molecular Biology and Genetics, Istanbul Kultur University, 34158 Istanbul, Turkey; (P.O.K.); (O.S.); (A.C.G.); (P.O.Y.)
| | - Pinar Uysal-Onganer
- Cancer Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK;
| | - Ajda Coker Gurkan
- Department of Molecular Biology and Genetics, Istanbul Kultur University, 34158 Istanbul, Turkey; (P.O.K.); (O.S.); (A.C.G.); (P.O.Y.)
| | - Pinar Obakan Yerlikaya
- Department of Molecular Biology and Genetics, Istanbul Kultur University, 34158 Istanbul, Turkey; (P.O.K.); (O.S.); (A.C.G.); (P.O.Y.)
| | - Elif Damla Arisan
- Institute of Biotechnology, Gebze Technical University, 41400 Kocaeli, Turkey
| |
Collapse
|
118
|
Liu H, Yao J, Guo H, Cai X, Jiang Y, Lin M, Jiang X, Leung W, Xu C. Tumor Microenvironment-Responsive Nanomaterials as Targeted Delivery Carriers for Photodynamic Anticancer Therapy. Front Chem 2020; 8:758. [PMID: 33134254 PMCID: PMC7550754 DOI: 10.3389/fchem.2020.00758] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Photodynamic therapy (PDT), as an alternative approach to treat tumors through reactive oxygen species (ROS) produced by the activated photosensitizers (PS) upon light irradiation, has attracted wide attention in recent years due to its low invasive and highly efficient features. However, the low hydrophilicity and poor targeting of PS limits the clinical application of PDT. Stimuli-responsive nanomaterials represent a major class of remarkable functional nanocarriers for drug delivery. In particular, tumor microenvironment-responsive nanomaterials (TMRNs) can respond to the special pathological microenvironment in tumor tissues to release the loaded drugs, that allows them to control the release of PS within tumor tissues. Recent studies have demonstrated that TMRNs can achieve the targeted release of PS at tumor sites, increase the concentration of PS in tumor tissues, and reduce side effects of PDT. Hence, in the present paper, we review TMRNs, mainly including pH-, redox-, enzymes-, and hypoxia-responsive smart nanomaterials, and focus on the application of these smart nanomaterials as targeted delivery carriers of PS in photodynamic anticancer therapy, to further boost the development of PDT in tumor therapy.
Collapse
Affiliation(s)
- Houhe Liu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Science & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiwen Yao
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Science & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huanhuan Guo
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Science & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaowen Cai
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Science & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuan Jiang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Science & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mei Lin
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Science & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xuejun Jiang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Science & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wingnang Leung
- Asia-Pacific Institute of Aging Studies, Lingnan University, Hong Kong, China
| | - Chuanshan Xu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Science & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
119
|
Vuoso DC, D'Angelo S, Ferraro R, Caserta S, Guido S, Cammarota M, Porcelli M, Cacciapuoti G. Annurca apple polyphenol extract promotes mesenchymal-to-epithelial transition and inhibits migration in triple-negative breast cancer cells through ROS/JNK signaling. Sci Rep 2020; 10:15921. [PMID: 32985606 PMCID: PMC7522716 DOI: 10.1038/s41598-020-73092-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022] Open
Abstract
Aberrant activation of epithelial-to-mesenchymal transition has been shown to correlate with triple-negative breast cancer (TNBC) progression and metastasis. Thus, the induction of the reverse process might offer promising opportunities to restrain TNBC metastatic spreading and related mortality. Recently, the Annurca apple polyphenol extract (APE) has been highlighted as a multi-faceted agent that selectively kills TNBC cells by ROS generation and sustained JNK activation. Here, by qualitatively and quantitatively monitoring the real-time movements of live cells we provided the first evidence that APE inhibited the migration of MDA-MB-231 and MDA-MB-468 TNBC cells and downregulated metalloproteinase-2 and metalloproteinase-9. In MDA-MB-231 cells APE decreased SMAD-2/3 and p-SMAD-2/3 levels, increased E-cadherin/N-cadherin protein ratio, induced the switch from N-cadherin to E-cadherin expression and greatly reduced vimentin levels. Confocal and scanning electron microscopy imaging of APE-treated MDA-MB-231 cells evidenced a significant cytoskeletal vimentin and filamentous actin reorganization and revealed considerable changes in cell morphology highlighting an evident transition from the mesenchymal to epithelial phenotype with decreased migratory features. Notably, all these events were reverted by N-acetyl-l-cysteine and JNK inhibitor SP600125 furnishing evidence that APE exerted its effects through the activation of ROS/JNK signaling. The overall data highlighted APE as a potential preventing agent for TNBC metastasis.
Collapse
Affiliation(s)
- Daniela Cristina Vuoso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 7, 80138, Naples, Italy
| | - Stefania D'Angelo
- Department of Motor Sciences and Wellness, "Parthenope" University, Via Medina 40, 80133, Naples, Italy
| | - Rosalia Ferraro
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, P.le Tecchio 80, 80125, Naples, Italy.,CEINGE Advanced Biotechnologies, 80145, Naples, Italy
| | - Sergio Caserta
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, P.le Tecchio 80, 80125, Naples, Italy.,CEINGE Advanced Biotechnologies, 80145, Naples, Italy
| | - Stefano Guido
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, P.le Tecchio 80, 80125, Naples, Italy.,CEINGE Advanced Biotechnologies, 80145, Naples, Italy
| | - Marcella Cammarota
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Luciano Armanni 5, 80138, Naples, Italy
| | - Marina Porcelli
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 7, 80138, Naples, Italy
| | - Giovanna Cacciapuoti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 7, 80138, Naples, Italy.
| |
Collapse
|
120
|
Luo H, Cong S, Dong J, Jin L, Jiang D, Wang X, Chen Q, Li F. Paired‑related homeobox 1 overexpression promotes multidrug resistance via PTEN/PI3K/AKT signaling in MCF‑7 breast cancer cells. Mol Med Rep 2020; 22:3183-3190. [PMID: 32945446 PMCID: PMC7453582 DOI: 10.3892/mmr.2020.11414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 06/19/2020] [Indexed: 12/17/2022] Open
Abstract
Multidrug resistance (MDR) is a major cause of disease relapse and mortality in breast cancer. Paired‑related homeobox 1 (PRRX1) is associated with the epithelial‑mesenchymal transition (EMT), which is involved in tumor development, including cell invasion and MDR. However, the effect of PRRX1 on MDR had not clearly established. The present study investigated the influence of PRRX1 on MDR and the underlying molecular mechanisms in MCF‑7 breast cancer cells. MCF‑7 cells were divided into PRRX1+ group (cells transfected with a recombinant plasmid carrying the PRRX1 gene), negative control group (cells transfected with a blank vector) and blank group (untreated cells). It was found that the relative protein and mRNA expression levels of PRRX1, N‑cadherin, vimentin and P‑glycoprotein were significantly higher in PRRX1‑overexpressing MCF‑7 cells compared with those in control cells. The half‑maximal inhibitory concentration of three groups after treatment with docetaxel and cis‑platinum complexes were significantly higher in PRRX1‑overexpressing MCF‑7 cells compared with those in control cells. Furthermore, relative PTEN expression decreased significantly and levels of phosphorylated PI3K and AKT increased substantially in PRRX1‑overexpressing MCF‑7 cells. These results indicated that PRRX1 overexpression may induce MDR via PTEN/PI3K/AKT signaling in breast cancer. It is highly recommended that PRRX1 gene expression detection should be performed in patients with breast cancer to aid the selection of more appropriate treatments, which will lead to an improved prognosis in clinical practice.
Collapse
Affiliation(s)
- Haoyue Luo
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Shaobo Cong
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Jiaojiao Dong
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Litao Jin
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Dandan Jiang
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Xingang Wang
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Qingfeng Chen
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Funian Li
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
121
|
Plygawko AT, Kan S, Campbell K. Epithelial-mesenchymal plasticity: emerging parallels between tissue morphogenesis and cancer metastasis. Philos Trans R Soc Lond B Biol Sci 2020; 375:20200087. [PMID: 32829692 PMCID: PMC7482222 DOI: 10.1098/rstb.2020.0087] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Many cells possess epithelial–mesenchymal plasticity (EMP), which allows them to shift reversibly between adherent, static and more detached, migratory states. These changes in cell behaviour are driven by the programmes of epithelial–mesenchymal transition (EMT) and mesenchymal–epithelial transition (MET), both of which play vital roles during normal development and tissue homeostasis. However, the aberrant activation of these processes can also drive distinct stages of cancer progression, including tumour invasiveness, cell dissemination and metastatic colonization and outgrowth. This review examines emerging common themes underlying EMP during tissue morphogenesis and malignant progression, such as the context dependence of EMT transcription factors, a central role for partial EMTs and the nonlinear relationship between EMT and MET. This article is part of a discussion meeting issue ‘Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Andrew T Plygawko
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Shohei Kan
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Kyra Campbell
- Department of Biomedical Science and Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
122
|
Lin X, Ping J, Wen Y, Wu Y. The Mechanism of Ferroptosis and Applications in Tumor Treatment. Front Pharmacol 2020; 11:1061. [PMID: 32774303 PMCID: PMC7388725 DOI: 10.3389/fphar.2020.01061] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/30/2020] [Indexed: 01/17/2023] Open
Abstract
Iron-dependent ferroptosis is a new form of cell death in recent years, which is driven by lipid peroxidation. The lethal lipid accumulation caused by glutathione depletion or inactivation of glutathione peroxidase 4 (GPX4) is characteristic of the ferroptosis process. In recent years, with the in-depth study of ferroptosis, various types of diseases have been reported to be related to ferroptosis. In other words, ferroptosis, which has attracted widespread attention in the fields of biochemistry, oncology, and especially materials science, can undoubtedly provide a new way for patients. This review introduces the relevant mechanisms of ferroptosis, the relationship between ferroptosis and various cancers, as well as the application of ferroptosis in tumor treatment. We also sorted out the genes and drugs that regulate ferroptosis. Moreover, small molecule compound-induced ferroptosis has a strong inhibitory effect on tumor growth in a drug-resistant environment, which can enhance the sensitivity of chemotherapeutic drugs, suggesting that ferroptosis is very important in the treatment of tumor drug resistance, but the details are still unclear. How to use ferroptosis to fight cancer, and how to prevent drug-resistant tumor cells have become the focus and direction of research. At the end of the article, some existing problems related to ferroptosis are summarized for future research.
Collapse
Affiliation(s)
- Xinyue Lin
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jieyi Ping
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yi Wen
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yan Wu
- School of Medicine, Jiangsu University, Zhenjiang, China.,Gynecology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
123
|
Ma F, Xie Y, Lei Y, Kuang Z, Liu X. The microRNA-130a-5p/RUNX2/STK32A network modulates tumor invasive and metastatic potential in non-small cell lung cancer. BMC Cancer 2020; 20:580. [PMID: 32571328 PMCID: PMC7310151 DOI: 10.1186/s12885-020-07056-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) remains a huge health burden for human health and life worldwide. Our study here was to illuminate the relevance of microRNA-130a-5p (miR-130a-5p) on growth and epithelial mesenchymal transition (EMT) in NSCLC cells along with metastasis in vivo, and to explore the underlying mechanism. Methods RT-qPCR was carried out for miR-130a-5p expression determination in NSCLC cells and tissue samples. Dual-luciferase reporter gene assay, RT-qPCR and western blot were carried out to study the potential targets of miR-130a-5p. Effects of miR-130a-5p, runt-related transcription factor 2 (RUNX2) and encoding serine/threonine kinase 32A (STK32A) on NSCLC proliferation, migration, invasion as well as EMT processes were assessed by cell counting kits-8, colony formation, Transwell and western blot assays. Results miR-130a-5p was diminished in NSCLC tissues and cells versus their counterparts. miR-130a-5p exerted its repressive role in NSCLC by curtailing cell viability, migration, invasion as well as EMT, while facilitating apoptosis. miR-130a-5p directly targeted RUNX2, a transcription factor, and conversely regulated its expression. RUNX2 was found to interact with STK32A to promote its expression. Following the validation of the supporting role of STK32A in NSCLC cells and NF-κB p65 phosphorylation, RUNX2 overexpression was monitored to reverse miR-130a-5p-inhibited NSCLC tumor volume and weight through enhancing STK32A expression in vivo. Conclusions miR-130a-5p diminished the growth and EMT of NSCLC cells by regulating the RUNX2/STK32A/NF-κB p65 axis, offering possible targets for the treatment for NSCLC.
Collapse
Affiliation(s)
- Fang Ma
- Department of Oncology, the Second Xiangya Hospital of Central South University, No. 139, Renmin Middle Road, Furong District, Changsha, 410000, Hunan, P.R. China
| | - Yangchun Xie
- Department of Oncology, the Second Xiangya Hospital of Central South University, No. 139, Renmin Middle Road, Furong District, Changsha, 410000, Hunan, P.R. China
| | - Yiyu Lei
- Department of Oncology, the Second Xiangya Hospital of Central South University, No. 139, Renmin Middle Road, Furong District, Changsha, 410000, Hunan, P.R. China
| | - Zengshuyu Kuang
- Department of Oncology, Zhuzhou 331 Hospital, Zhuzhou, 412000, Hunan, P.R. China
| | - Xianling Liu
- Department of Oncology, the Second Xiangya Hospital of Central South University, No. 139, Renmin Middle Road, Furong District, Changsha, 410000, Hunan, P.R. China.
| |
Collapse
|
124
|
SPOCK1 is a novel inducer of epithelial to mesenchymal transition in drug-induced gingival overgrowth. Sci Rep 2020; 10:9785. [PMID: 32555336 PMCID: PMC7300011 DOI: 10.1038/s41598-020-66660-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/22/2020] [Indexed: 11/28/2022] Open
Abstract
Few studies have investigated the role of extracellular-matrix proteoglycans in the pathogenesis of drug-induced gingival overgrowth (DIGO). SPOCK1 is an extracellular proteoglycan that induces epithelial to mesenchymal transition (EMT) in several cancer cell lines and exhibits protease-inhibitory activity. However, the role of SPOCK1 in non-cancerous diseases such as DIGO has not been well-addressed. We demonstrated that the expression of SPOCK1, TGF-β1, and MMP-9 in calcium channel blocker-induced gingival overgrowth is higher than that in non-overgrowth tissues. Transgenic mice overexpressing Spock1 developed obvious gingival-overgrowth and fibrosis phenotypes, and positively correlated with EMT-like changes. Furthermore, in vitro data indicated a tri-directional interaction between SPOCK1, TGF-β1, and MMP-9 that led to gingival overgrowth. Our study shows that SPOCK1 up-regulation in a noncancerous disease and SPOCK1-induced EMT in gingival overgrowth occurs via cooperation and crosstalk between several potential signaling pathways. Therefore, SPOCK1 is a novel therapeutic target for gingival overgrowth and its expression is a potential risk of EMT induction in cancerous lesions.
Collapse
|
125
|
Li XY, Wang JH, Gu LY, Yao XM, Cai FY, Jing M, Li XT, Ju RJ. Dual variable of drug loaded micelles in both particle and electrical charge on gastric cancer treatment. J Drug Target 2020; 28:1071-1084. [PMID: 32484364 DOI: 10.1080/1061186x.2020.1777419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Gastric cancer is a malignant tumour characterised by the uncontrolled cell growth. The incidence and mortality of gastric cancer remain high for the invasion and metastasis. We are urgently seeking a risk-free and effective treatment strategy for gastric cancer. In this study, paclitaxel and tetrandrine were encapsulated in the inner core of micelles, and DSPE-PEG2000-CPP and HA were modified on the micellar surface. HA/CPP modified paclitaxel plus tetrandrine micelles had a suitable particle size (90 nm) for permeating tumour tissue. The zeta potential of the targeting micelles was 8.37 mV after hydrolysis by HAase solution. Results of in vitro experiments indicated that HA/CPP modified paclitaxel plus tetrandrine micelles + HAase could enhance the intracellular uptake, inhibit the formation of neovascularization, block the process of EMT and destroy the invasion and metastasis. In vivo assays indicated that HA/CPP modified paclitaxel plus tetrandrine micelles could be selectively accumulated into tumour sites and exhibited the strong antitumor activity with negligible toxicity. These results suggested that HA/CPP modified paclitaxel plus tetrandrine micelles might provide a new strategy for treating gastric cancer.
Collapse
Affiliation(s)
- Xiu-Ying Li
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Jian-Hua Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Li-Yan Gu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xue-Min Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Fu-Yi Cai
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Ming Jing
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Rui-Jun Ju
- Department of Pharmaceutical Engineering, Beijing Institute of Petrochemical Technology, Beijing, China
| |
Collapse
|
126
|
Hakin-1, a New Specific Small-Molecule Inhibitor for the E3 Ubiquitin-Ligase Hakai, Inhibits Carcinoma Growth and Progression. Cancers (Basel) 2020; 12:cancers12051340. [PMID: 32456234 PMCID: PMC7281109 DOI: 10.3390/cancers12051340] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/22/2022] Open
Abstract
The requirement of the E3 ubiquitin-ligase Hakai for the ubiquitination and subsequent degradation of E-cadherin has been associated with enhanced epithelial-to-mesenchymal transition (EMT), tumour progression and carcinoma metastasis. To date, most of the reported EMT-related inhibitors were not developed for anti-EMT purposes, but indirectly affect EMT. On the other hand, E3 ubiquitin-ligase enzymes have recently emerged as promising therapeutic targets, as their specific inhibition would prevent wider side effects. Given this background, a virtual screening was performed to identify novel specific inhibitors of Hakai, targeted against its phosphotyrosine-binding pocket, where phosphorylated-E-cadherin specifically binds. We selected a candidate inhibitor, Hakin-1, which showed an important effect on Hakai-induced ubiquitination. Hakin-1 also inhibited carcinoma growth and tumour progression both in vitro, in colorectal cancer cell lines, and in vivo, in a tumour xenograft mouse model, without apparent systemic toxicity in mice. Our results show for the first time that a small molecule putatively targeting the E3 ubiquitin-ligase Hakai inhibits Hakai-dependent ubiquitination of E-cadherin, having an impact on the EMT process. This represents an important step forward in a future development of an effective therapeutic drug to prevent or inhibit carcinoma tumour progression.
Collapse
|
127
|
Yao XM, Niu FJ, Kong L, Cai FY, Jing M, Fu M, Liu JJ, He SY, Zhang L, Liu XZ, Ju RJ, Li XT. GGP modified daunorubicin plus dioscin liposomes inhibit breast cancer by suppressing epithelial-mesenchymal transition. Drug Dev Ind Pharm 2020; 46:916-930. [PMID: 32362146 DOI: 10.1080/03639045.2020.1763397] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor invasion and metastasis are the nodus of anti-tumor. Epithelial cell-mesenchymal transition is widely regarded as one of the key steps in the invasion and metastasis of breast cancer. In this study, GGP modified daunorubicin plus dioscin liposomes are constructed and characterized. GGP modified daunorubicin plus dioscin liposome has suitable particle size, narrow PDI, zeta potential of about -5 mV, long cycle effect, and enhanced cell uptake due to surface modification of GGP making the liposome could enter the inside of the tumor to fully exert its anti-tumor effect. The results of in vitro experiments show that the liposome has superior killing effect on tumor cells and invasion. In vivo results indicate that the liposome prolongs the drug's prolonged time in the body and accumulates at the tumor site with little systemic toxicity. In short, the targeted liposome can effectively inhibit tumor invasion and may provide a new strategy for the treatment of invasive breast cancer.
Collapse
Affiliation(s)
- Xue-Min Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Feng-Ju Niu
- Health Protection Center, Affiliated Hospital of Shandong Academy of Traditional Chinese Medicine, Jinan, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Fu-Yi Cai
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Ming Jing
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Min Fu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Jing-Jing Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Si-Yu He
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xin-Ze Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Rui-Jun Ju
- Department of Pharmaceutical Engineering, Beijing Institute of Petrochemical Technology, Beijing, China
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| |
Collapse
|
128
|
Liu H, Fu Q, Lu Y, Zhang W, Yu P, Liu Z, Sun X. Anti-tubulin agent vinorelbine inhibits metastasis of cancer cells by regulating epithelial-mesenchymal transition. Eur J Med Chem 2020; 200:112332. [PMID: 32473523 DOI: 10.1016/j.ejmech.2020.112332] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/12/2020] [Accepted: 04/12/2020] [Indexed: 02/06/2023]
Abstract
Cancer invasion and metastasis are the leading causes of death. The process of metastasis or tumor cell dissemination is still much of a mystery. Emerging evidence has shown that epithelial-mesenchymal transition (EMT) plays a vital role in the progression of malignant tumor including the inducing cell invasion and metastasis as well as promoting drug resistance. Vinorelbine is a traditional chemotherapeutic agent for treatment of lung cancer and breast cancer by the selectivity to mitotic microtubules. The aim of this study was to investigate the effect of vinorelbine on three metastatic cancer cells including lung cancer (H1975), liver cancer (HepG2), and colon cancer (HCT116) cells through inhibition of metastatic abilities and EMT program. Vinorelbine inhibited the cancer cell proliferation by MTT and colony formation assays and inducing G2/M arrest and cell apoptosis via regulation of Bax, Bcl-2, and Bcl-xL. Vinorelbine decrease the migration and invasion ability of the cancer cells by wound healing assay and Tran swell test. The molecular mechanisms of vinorelbine suppressing the metastatic phenotypes of cancer cells through modulation of E-cadherin, N-cadherin, vimentin and transcription factors Snail, MMP-2 and MMP-9. Our results demonstrated that vinorelbine inhibited the cancer cell metastasis through a reduction in metastatic mobility, such as migration, invasion, and the EMT. It provided the evidence that vinorelbine can be used alone or with other agents for treatment of metastatic lung cancer, liver cancer and colon cancer.
Collapse
Affiliation(s)
- Hongyu Liu
- School of Basic Medical Sciences, Guangzhou University of Traditional Chinese Medicine, Guangdong, 510000, PR China
| | - Qingshan Fu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Yao Lu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Wenqiang Zhang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China
| | - Zhen Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| | - Xiaosheng Sun
- School of Basic Medical Sciences, Guangzhou University of Traditional Chinese Medicine, Guangdong, 510000, PR China.
| |
Collapse
|
129
|
Li G, Wang Y, Tan G, Liu Y, Xu Z, Feng H, Xing W, Xu Z. [Preliminary Study on Detection of Circulating Tumor Cells in Lung Cancer by EGFR/Vimentin/Folic Acid Magnetic Sphere]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:351-359. [PMID: 32336066 PMCID: PMC7260381 DOI: 10.3779/j.issn.1009-3419.2020.103.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
背景与目的 循环肿瘤细胞(circulating tumor cell, CTC)在肺癌的筛查及预后方面发挥着重要的作用, 但较低的CTC分离效率和特异性对其临床应用有着明显的制约, 本研究旨在探讨非小细胞肺癌(non-small cell lung cancer, NSCLC)患者CTC的新型高效分离方法, 以期达到对NSCLC的早期诊断的目的。 方法 采用薄膜法制备表皮生长因子受体(epidermal growth factor receptor, EGFR)、波形蛋白(Vimentin)和叶酸(folic acid, FA)三种免疫脂质磁球, 表征后通过细胞系进行分选方案的探索, 构建对NSCLC CTC的最优分选方案, 初步研究了其在临床上的应用价值。 结果 EGFR、Vimentin和FA磁球磁球单独和联合使用对肺癌细胞株的平均捕获效率分别为78.0%、79.0%、82.0%和91.0%;在60例肺癌患者中, 以每7.5 mL血液2个CTC为cutoff值, EGFR、Vimentin、FA磁球单独和联合使用阳性率分别为65.0%、33.3%、93.3%和100.0%, 同时发现联合使用三种磁球检出的CTC数量与临床分期具有相关性(P < 0.05)。 结论 联合使用三种磁球可以分离EGFR+、Vimentin+和FA+表达且形态完整的CTC, 有利于的CTC相关下游分析, 本研究提供了一种提高NSCLC CTC捕获效率的新方法, 且验证了捕获的CTC计数方法可用于肺癌的辅助诊断。
Collapse
Affiliation(s)
- Guolei Li
- The First Surgery Department, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang 050051, China
| | - Yun Wang
- Department of Medical Ultrasonics, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Guoliang Tan
- The First Surgery Department, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang 050051, China
| | - Yuan Liu
- The First Surgery Department, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang 050051, China
| | - Zhao Xu
- The First Surgery Department, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang 050051, China
| | - Hao Feng
- The First Surgery Department, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang 050051, China
| | - Wei Xing
- The First Surgery Department, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang 050051, China
| | - Zhifeng Xu
- The First Surgery Department, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang 050051, China
| |
Collapse
|
130
|
Bhatia S, Wang P, Toh A, Thompson EW. New Insights Into the Role of Phenotypic Plasticity and EMT in Driving Cancer Progression. Front Mol Biosci 2020; 7:71. [PMID: 32391381 PMCID: PMC7190792 DOI: 10.3389/fmolb.2020.00071] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor cells demonstrate substantial plasticity in their genotypic and phenotypic characteristics. Epithelial-mesenchymal plasticity (EMP) can be characterized into dynamic intermediate states and can be orchestrated by many factors, either intercellularly via epigenetic reprograming, or extracellularly via growth factors, inflammation and/or hypoxia generated by the tumor stromal microenvironment. EMP has the capability to alter phenotype and produce heterogeneity, and thus by changing the whole cancer landscape can attenuate oncogenic signaling networks, invoke anti-apoptotic features, defend against chemotherapeutics and reprogram angiogenic and immune recognition functions. We discuss here the role of phenotypic plasticity in tumor initiation, progression and metastasis and provide an update of the modalities utilized for the molecular characterization of the EMT states and attributes of cellular behavior, including cellular metabolism, in the context of EMP. We also summarize recent findings in dynamic EMP studies that provide new insights into the phenotypic plasticity of EMP flux in cancer and propose therapeutic strategies to impede the metastatic outgrowth of phenotypically heterogeneous tumors.
Collapse
Affiliation(s)
- Sugandha Bhatia
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| | - Peiyu Wang
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| | - Alan Toh
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| | - Erik W Thompson
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
131
|
(20S)G-Rh2 Inhibits NF-κB Regulated Epithelial-Mesenchymal Transition by Targeting Annexin A2. Biomolecules 2020; 10:biom10040528. [PMID: 32244350 PMCID: PMC7225922 DOI: 10.3390/biom10040528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
(1) Background: Epithelial-mesenchymal transition (EMT) is an essential step for cancer metastasis; targeting EMT is an important path for cancer treatment and drug development. NF-κB, an important transcription factor, has been shown to be responsible for cancer metastasis by enhancing the EMT process. Our previous studies showed that (20S)Ginsenoside Rh2 (G-Rh2) inhibits NF-κB activity by targeting Anxa2, but it is still not known whether this targeted inhibition of NF-κB can inhibit the EMT process. (2) Methods: In vivo (20S)G-Rh2-Anxa2 interaction was assessed by cellular thermal shift assay. Protein interaction was determined by immuno-precipitation analysis. NF-κB activity was determined by dual luciferase reporter assay. Gene expression was determined by RT-PCR and immuno-blot. EMT was evaluated by wound healing and Transwell assay and EMT regulating gene expression. (3) Results: Anxa2 interacted with the NF-κB p50 subunit, promoted NF-κB activation, then accelerated mesenchymal-like gene expression and enhanced cell motility; all these cellular processes were inhibited by (20S)G-Rh2. In contrast, these (20S)G-Rh2 effect were completely eliminated by overexpression of Anxa2-K301A, an (20S)G-Rh2-binding-deficient mutant of Anxa2. (4) Conclusion: (20S)G-Rh2 inhibited NF-κB activation and related EMT by targeting Anxa2 in MDA-MB-231 cells.
Collapse
|
132
|
Wang YB, You ZH, Yang S, Yi HC, Chen ZH, Zheng K. A deep learning-based method for drug-target interaction prediction based on long short-term memory neural network. BMC Med Inform Decis Mak 2020; 20:49. [PMID: 32183788 PMCID: PMC7079345 DOI: 10.1186/s12911-020-1052-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background The key to modern drug discovery is to find, identify and prepare drug molecular targets. However, due to the influence of throughput, precision and cost, traditional experimental methods are difficult to be widely used to infer these potential Drug-Target Interactions (DTIs). Therefore, it is urgent to develop effective computational methods to validate the interaction between drugs and target. Methods We developed a deep learning-based model for DTIs prediction. The proteins evolutionary features are extracted via Position Specific Scoring Matrix (PSSM) and Legendre Moment (LM) and associated with drugs molecular substructure fingerprints to form feature vectors of drug-target pairs. Then we utilized the Sparse Principal Component Analysis (SPCA) to compress the features of drugs and proteins into a uniform vector space. Lastly, the deep long short-term memory (DeepLSTM) was constructed for carrying out prediction. Results A significant improvement in DTIs prediction performance can be observed on experimental results, with AUC of 0.9951, 0.9705, 0.9951, 0.9206, respectively, on four classes important drug-target datasets. Further experiments preliminary proves that the proposed characterization scheme has great advantage on feature expression and recognition. We also have shown that the proposed method can work well with small dataset. Conclusion The results demonstration that the proposed approach has a great advantage over state-of-the-art drug-target predictor. To the best of our knowledge, this study first tests the potential of deep learning method with memory and Turing completeness in DTIs prediction.
Collapse
Affiliation(s)
- Yan-Bin Wang
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China.,Department of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhu-Hong You
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China.
| | - Shan Yang
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China
| | - Hai-Cheng Yi
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China.,Department of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhan-Heng Chen
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China.,Department of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai Zheng
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China
| |
Collapse
|
133
|
Zhu X, Yu M, Wang K, Zou W, Zhu L. FoxM1 affects adhesive, migratory, and invasive abilities of human retinoblastoma Y-79 cells by targeting matrix metalloproteinase 2. Acta Biochim Biophys Sin (Shanghai) 2020; 52:294-301. [PMID: 32152631 DOI: 10.1093/abbs/gmz160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/01/2019] [Accepted: 11/06/2019] [Indexed: 12/22/2022] Open
Abstract
Forkhead box protein M1 (FoxM1) is an important transcription factor involved in various pathological processes including tumor metastasis. The changes of adhesive, migratory, and invasive abilities are considered as crucial events in tumor metastasis progression. In this study, we aimed to investigate the correlation between FoxM1 and retinoblastoma (Rb) metastasis and to explore the detailed mechanism. Wound healing, cell adhesion, and invasion assays showed that FoxM1 overexpression induced epithelial-mesenchymal transition in Y-79 cells and inhibited adhesion and subsequently promoted metastasis of Y-79 cells, while FoxM1 knockdown showed the opposite effects. A luciferase reporter assay and chromatin immunoprecipitation assay provided evidence that FoxM1 promoted matrix metalloproteinase 2 (MMP2) transcription by directly binding to and promoting MMP2 promoter. MMP2 knockdown by siRNA transfection attenuated cell metastasis of Y-79 cells induced by FoxM1 overexpression. Furthermore, the FoxM1-binding site mapped between -1167 and -1161 bp of the MMP2 promoter was identified. Our results suggested that the FoxM1-MMP2 axis plays an important role in Rb metastasis, which may be a novel target for designing therapeutic regimen to control Rb metastasis.
Collapse
Affiliation(s)
- Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Mengxi Yu
- Department of Ophthalmology, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, Jiangsu, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Wenjun Zou
- Department of Ophthalmology, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, Jiangsu, China
| | - Ling Zhu
- Save Sight Institute, University of Sydney, Sydney, New South Wales 2000, Australia
| |
Collapse
|
134
|
Mosca L, Minopoli M, Pagano M, Vitiello F, Carriero MV, Cacciapuoti G, Porcelli M. Effects of S‑adenosyl‑L‑methionine on the invasion and migration of head and neck squamous cancer cells and analysis of the underlying mechanisms. Int J Oncol 2020; 56:1212-1224. [PMID: 32319579 PMCID: PMC7115356 DOI: 10.3892/ijo.2020.5011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/13/2020] [Indexed: 02/07/2023] Open
Abstract
S-Adenosyl-L-methionine (AdoMet) is the principal methyl donor in transmethylation reactions fundamental to sustaining epigenetic modifications. Over the past decade, AdoMet has been extensively investigated for its anti- proliferative, pro-apoptotic and anti-metastatic roles in several types of human cancer. Head and neck squamous cell carcinoma (HNSCC) is the sixth most common type of cancer worldwide, and is an aggressive type of cancer that is associated with a high recurrence rate, metastasis and poor treatment outcomes. The present study demonstrates, for the first time, to the best of our knowledge, that AdoMet induces cell cycle arrest and inhibits the migratory and invasive ability of two different HNSCC cell lines, oral Cal-33 and laryngeal JHU-SCC-011 cells. In both cell lines, AdoMet attenuated cell cycle progression, decreased the protein level of several cyclins and downregulated the expression of p21 cell cycle inhibitor. Moreover, AdoMet was able to inhibit Cal-33 and JHU-SCC-011 cell migration in a dose-dependent manner after 24 and 48 h, respectively, and also induced a significant reduction in the cell invasive ability, as demonstrated by Matrigel invasion assay monitored by the xCELLigence RTCA system. Western blot analysis of several migration and invasion markers confirmed the inhibitory effects exerted by AdoMet on these processes and highlighted AKT, β-catenin and small mothers against decapentaplegic (SMAD) as the main signaling pathways modulated by AdoMet. The present study also demonstrated that the combination of AdoMet and cisplatin synergistically inhibited HNSCC cell migration. Taken together, these findings demonstrate that the physiological compound, AdoMet, affects the motility and extracellular matrix invasive capability in HNSCC. Thus, AdoMet may prove to be a good candidate for future drug development against metastatic cancer.
Collapse
Affiliation(s)
- Laura Mosca
- Dipartimento di Medicina di Precisione, Università della Campania 'Luigi Vanvitelli', I‑80138 Napoli, Italy
| | - Michele Minopoli
- Unità Progressione Neoplastica, Istituto Nazionale Tumori‑IRCCS 'Fondazione G. Pascale', I‑80131 Napoli, Italy
| | - Martina Pagano
- Dipartimento di Medicina di Precisione, Università della Campania 'Luigi Vanvitelli', I‑80138 Napoli, Italy
| | - Francesca Vitiello
- Dipartimento di Medicina di Precisione, Università della Campania 'Luigi Vanvitelli', I‑80138 Napoli, Italy
| | - Maria Vincenza Carriero
- Unità Progressione Neoplastica, Istituto Nazionale Tumori‑IRCCS 'Fondazione G. Pascale', I‑80131 Napoli, Italy
| | - Giovanna Cacciapuoti
- Dipartimento di Medicina di Precisione, Università della Campania 'Luigi Vanvitelli', I‑80138 Napoli, Italy
| | - Marina Porcelli
- Dipartimento di Medicina di Precisione, Università della Campania 'Luigi Vanvitelli', I‑80138 Napoli, Italy
| |
Collapse
|
135
|
Marcucci F, Romeo E, Caserta CA, Rumio C, Lefoulon F. Context-Dependent Pharmacological Effects of Metformin on the Immune System. Trends Pharmacol Sci 2020; 41:162-171. [DOI: 10.1016/j.tips.2020.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/14/2019] [Accepted: 01/06/2020] [Indexed: 12/20/2022]
|
136
|
Yu J, Shen W, Gao B, Xu J, Gong B. Metastasis suppressor 1 acts as a tumor suppressor by inhibiting epithelial-to-mesenchymal transition in triple-negative breast cancer. Int J Biol Markers 2020; 35:74-81. [PMID: 32052679 DOI: 10.1177/1724600820905114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study aimed to analyze the function of metastasis suppressor 1 (MTSS1) in triple negative breast cancer (TNBC). METHODS MTSS1 expression in 30 TNBC and paracancerous tissues was measured by quantitative reverse transcriptase polymerase chain reaction. The prognostic value of MTSS1 was assessed by Kaplan-Meier analysis followed by the log-rank test. MCF7 cells were transfected with si-MTSS1, while MDA-MB-231 cells were transfected with pcDNA3.1-MTSS1. Cell proliferation assay and transwell assay were performed to investigate the effects of MTSS1 on the biological behavior of breast cancer cells. Immunofluorescence and western blot were used to detect the influence of MTSS1 on epithelial-mesenchymal transition (EMT) markers. RESULTS MTSS1 expression was significantly lower in TNBC tissues compared with that in paracancerous tissues (0.012 vs. 0.370; P = 0.006). A lower MTSS1 expression level was also found in tumor tissues of patients with lymph node metastasis (P = 0.002) or tumor node metastasis stage (P = 0.010). Patients with low expression of MTSS1 (⩽ 0.009) had shorter disease-free survival (47.4 vs. 56.0 months; P = 0.012). The knockdown of MTSS1 in MCF7 cells inhibited cell proliferation, enhanced cell migration and invasion capacities, decreased the E-cadherin level, and increased the vimentin level, whereas overexpression of MTSS1 in MDA-MB-231 cells had the opposite effects (P < 0.05). CONCLUSIONS Our findings demonstrated that MTSS1 regulates proliferation, invasion, migration, and EMT in TNBC, and that decreased MTSS1 is associated with shorter disease-free survival.
Collapse
Affiliation(s)
- Jinling Yu
- Department of Breast Surgery, Shanghai Changning Maternity & Infant Health Hospital, Shanghai, China
| | - Weida Shen
- Department of Breast Surgery, Shanghai Changning Maternity & Infant Health Hospital, Shanghai, China
| | - Beimin Gao
- Department of Breast Surgery, Shanghai Changning Maternity & Infant Health Hospital, Shanghai, China
| | - Jinping Xu
- Department of Pathology, Shanghai Changning Maternity & Infant Health Hospital, Shanghai, China
| | - Bo Gong
- Department of Laboratory, Shanghai Changning Maternity & Infant Health Hospital, Shanghai, China
| |
Collapse
|
137
|
Yamamoto S, Miyama T, Komoda T, Sugawara M, Nonomura M, Nakanishi J. A Facile Assay of Epithelial-mesenchymal Transition Based on Cooperativity Quantification of Cellular Autonomous Motions. ANAL SCI 2020; 36:263-267. [PMID: 31588066 DOI: 10.2116/analsci.19p233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Epithelial-mesenchymal transition (EMT), a qualitative change in cell migration behavior during cancer invasion and metastasis, is becoming a new target for anticancer drugs. Therefore, it is crucial to develop in vitro assays for the evaluation of the abilities of drug candidates to control EMT progression. We herein report on a method for the quantification of the EMT based on particle image velocimetry and correlation functions. The exponential fitting of the correlation curve gives an index (λ), which represents transforming growth factor (TGF)-β1-induced EMT progression and its suppression by inhibitors. Moreover, real-time monitoring of the λ value illustrates a time-dependent EMT progressing process, which occurs earlier than the bio-chemical changes in an EMT marker protein expression. The results demonstrate the usefulness of the present method for kinetic studies of EMT progression as well as EMT inhibitor screening.
Collapse
Affiliation(s)
- Shota Yamamoto
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS)
| | - Tatsuya Miyama
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS).,Department of Mathematical Information Engineering, College of Industrial Technology, Nihon University
| | - Takafumi Komoda
- Department of Mechanical Engineering, Graduate School of Engineering, Chiba University
| | - Michiko Sugawara
- Department of Mechanical Engineering, Graduate School of Engineering, Chiba University
| | - Makiko Nonomura
- Department of Mathematical Information Engineering, College of Industrial Technology, Nihon University
| | - Jun Nakanishi
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS).,Graduate School of Advanced Science and Engineering, Waseda University
| |
Collapse
|
138
|
Guo Y, Fan Y, Pei X. Fangjihuangqi Decoction inhibits MDA-MB-231 cell invasion in vitro and decreases tumor growth and metastasis in triple-negative breast cancer xenografts tumor zebrafish model. Cancer Med 2020; 9:2564-2578. [PMID: 32037729 PMCID: PMC7131862 DOI: 10.1002/cam4.2894] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 01/11/2020] [Accepted: 01/19/2020] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a basal-like cancer which is considered to be more intrusive, have a poorer prognosis and chemoresistance. TNBC is characterized by the presence of epithelial to mesenchymal transition (EMT) that plays a major role in the progression of the cancer. In the present study, we first use a classic prescription of Chinese medicine Fangjihuangqi Decoction to treat TGFβ1-induced MDA-MB-231 cells in vitro. Our data showed that TGFβ1-induced MDA-MB-231 cell morphology change, promoted MDA-MB 231 invasion, increased Vimentin expression, and decreased E-cadherin expression. Further, Fangjihuangqi Decoction-medicated serum (FHS) treated both MDA-MB 231 cells and TGFβ1-induced MDA-MB-231 cells. Results showed that Fangjihuangqi Decoction could inhibit cell proliferation, reduce cell invasion, increase E-cadherin expression, and decrease EMT markers. Secondly, we established a xenograft tumor zebrafish model to assess Fangjihuangqi Decoction inhibition of cancer cell proliferation and invasion. Our results indicated that Fangjihuangqi Decoction could inhibit tumor growth, restrain the sprouts number of tumor neovascularization, and reduce the length of tumor neoplastic lymphatics by increasing E-cadherin expression and decreasing EMT markers in TNBC xenograft tumor zebrafish model. Overall, our studies provide evidences that Fangjihuangqi Decoction could inhibit TNBC, reverse EMT, and contribute to antimetastasis by increasing E-cadherin expression and decreasing EMT markers, which provide an experimental basis for clinical application of Fangjihuangqi Decoction on TNBC treatment.
Collapse
Affiliation(s)
- Yubo Guo
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yingyi Fan
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohua Pei
- Beijing University of Chinese Medicine Fangshan Traditional Medical Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
139
|
LETM1 is a potential biomarker that predicts poor prognosis in gastric adenocarcinoma. Exp Mol Pathol 2020; 112:104333. [DOI: 10.1016/j.yexmp.2019.104333] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/01/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022]
|
140
|
Zajac O, Leclere R, Nicolas A, Meseure D, Marchiò C, Vincent-Salomon A, Roman-Roman S, Schoumacher M, Dubois T. AXL Controls Directed Migration of Mesenchymal Triple-Negative Breast Cancer Cells. Cells 2020; 9:cells9010247. [PMID: 31963783 PMCID: PMC7016818 DOI: 10.3390/cells9010247] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/09/2020] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer with high risk of relapse and metastasis. TNBC is a heterogeneous disease comprising different molecular subtypes including those with mesenchymal features. The tyrosine kinase AXL is expressed in mesenchymal cells and plays a role in drug resistance, migration and metastasis. We confirm that AXL is more expressed in mesenchymal TNBC cells compared to luminal breast cancer cells, and that its invalidation impairs cell migration while having no or little effect on cell viability. Here, we found that AXL controls directed migration. We observed that AXL displays a polarized localization at the Golgi apparatus and the leading edge of migratory mesenchymal TNBC cells. AXL co-localizes with F-actin at the front of the cells. In migratory polarized cells, the specific AXL inhibitor R428 displaces AXL and F-actin from the leading edge to a lateral area localized between the front and the rear of the cells where both are enriched in protrusions. In addition, R428 treatment disrupts the polarized localization of the Golgi apparatus towards the leading edge in migratory cells. Immunohistochemical analysis of aggressive chemo-resistant TNBC samples obtained before treatment reveals inter- and intra-tumor heterogeneity of the percentage of AXL expressing tumor cells, and a preference of these cells to be in contact with the stroma. Taken together, our study demonstrates that AXL controls directed cell migration most likely by regulating cell polarity.
Collapse
Affiliation(s)
- Olivier Zajac
- Breast Cancer Biology Group, Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France;
| | - Renaud Leclere
- Department of Pathology, Platform of Investigative Pathology, Institut Curie, PSL Research University, 75005 Paris, France; (R.L.); (A.N.); (D.M.)
| | - André Nicolas
- Department of Pathology, Platform of Investigative Pathology, Institut Curie, PSL Research University, 75005 Paris, France; (R.L.); (A.N.); (D.M.)
| | - Didier Meseure
- Department of Pathology, Platform of Investigative Pathology, Institut Curie, PSL Research University, 75005 Paris, France; (R.L.); (A.N.); (D.M.)
| | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, Via Verdi 8, 10124 Torino TO, Italy;
- Department of Pathology, Institut Curie, PSL Research University, 75005 Paris, France;
| | - Anne Vincent-Salomon
- Department of Pathology, Institut Curie, PSL Research University, 75005 Paris, France;
| | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France;
| | - Marie Schoumacher
- Center for Therapeutic Innovation Oncology, Institut de Recherches Internationales SERVIER, 92284 Suresnes, France;
| | - Thierry Dubois
- Breast Cancer Biology Group, Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France;
- Correspondence: ; Tel.: +33-156246250
| |
Collapse
|
141
|
Directly targeting glutathione peroxidase 4 may be more effective than disrupting glutathione on ferroptosis-based cancer therapy. Biochim Biophys Acta Gen Subj 2020; 1864:129539. [PMID: 31958545 DOI: 10.1016/j.bbagen.2020.129539] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/11/2020] [Accepted: 01/16/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cancer is one of the major threats to human health and current cancer therapies have been unsuccessful in eradicating it. Ferroptosis is characterized by iron-dependence and lipid hydroperoxides accumulation, and its primary mechanism involves the suppression of system Xc--GSH (glutathione)-GPX4 (glutathione peroxidase 4) axis. Co-incidentally, cancer cells are also metabolically characterized by iron addiction and ROS tolerance, which makes them vulnerable to ferroptosis. This may provide a new tactic for cancer therapy. SCOPE OF REVIEW The general features and mechanisms of ferroptosis, and the basis that makes cancer cells vulnerable to ferroptosis are described. Further, we emphatically discussed that disrupting GSH may not be ideal for triggering ferroptosis of cancer cells in vivo, but directly inhibiting GPX4 and its compensatory members could be more effective. Finally, the various approaches to directly inhibit GPX4 without disturbing GSH were described. MAJOR CONCLUSIONS Targeting system Xc- or GSH may not effectively trigger cancer cells' ferroptosis in vivo the existence of other compensatory pathways. However, directly targeting GPX4 and its compensatory members without disrupting GSH may be more effective to induce ferroptosis in cancer cells in vivo, as GPX4 is essential in preventing ferroptosis. GENERAL SIGNIFICANCE Cancer is a severe threat to human health. Ferroptosis-based cancer therapy strategies are promising, but how to effectively induce ferroptosis in cancer cells in vivo is still a question without clear answers. Thus, the viewpoints raised in this review may provide some references and different perspectives for researchers working on ferroptosis-based cancer therapy.
Collapse
|
142
|
Integrated Bioinformatics Analysis Identifies Hub Genes Associated with the Pathogenesis and Prognosis of Esophageal Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2615921. [PMID: 31950035 PMCID: PMC6948276 DOI: 10.1155/2019/2615921] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 09/26/2019] [Indexed: 12/24/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) accounts for over 90% of all esophageal tumors. However, the molecular mechanism underlying ESCC development and prognosis remains unclear, and there are still no effective molecular biomarkers for diagnosing or predicting the clinical outcome of patients with ESCC. Here, using bioinformatics analyses, we attempted to identify potential biomarkers and therapeutic targets for ESCC. Differentially expressed genes (DEGs) between ESCC and normal esophageal tissue samples were obtained through comprehensive analysis of three publicly available gene expression profile datasets from the Gene Expression Omnibus database. The biological roles of the DEGs were identified by Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Moreover, the Cytoscape 3.7.1 platform and subsidiary tools such as Molecular Complex Detection (MCODE) and CytoHubba were used to visualize the protein-protein interaction (PPI) network of the DEGs and identify hub genes. A total of 345 DEGs were identified between normal esophageal and ESCC samples, which were enriched in the KEGG pathways of the cell cycle, endocytosis, pancreatic secretion, and fatty acid metabolism. Two of the highest scoring models were selected from the PPI network using Molecular Complex Detection. Moreover, CytoHubba revealed 21 hub genes with a valuable influence on the progression of ESCC in these patients. Among these, the high expression levels of five genes-SPP1, SPARC, BGN, POSTN, and COL1A2-were associated with poor disease-free survival of ESCC patients, as indicated by survival analysis. Taken together, we identified that elevated expression of five hub genes, including SPP1, is associated with poor prognosis in ESCC patients, which may serve as potential prognostic biomarkers or therapeutic target for ESCC.
Collapse
|
143
|
Wang LK, Yue HL, Peng XJ, Zhang SJ. GSTO1 regards as a meritorious regulator in cutaneous malignant melanoma cells. Mol Cell Probes 2019; 48:101449. [PMID: 31525447 DOI: 10.1016/j.mcp.2019.101449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Glutathione S-transferase omega 1 (GSTO1), as a member of the glutathione S-transferase (GST) family genes, has been discovered to be up-regulated in several cancer cell lines which exhibited strong aggressiveness. However, the function of GSTO1 on cutaneous malignant melanoma (CMM) has not been illuminated. METHODS Outcome of expression level and prognosis of GSTO1 were obtained from Oncomine and TCGA database. The specific effects of GSTO1 on the characteristics and regulatory mechanism of CMM cells were demonstrated by cell counting kit-8, colony formation, flow cytometry, and transwell assays in vitro. Western blot was employed to analyze the expression of proliferating cell nuclear antigen (PCNA), p53 and epithelial-to-mesenchymal (EMT) related proteins. RESULTS We observed that GSTO1 was up-regulated in CMM samples when compared with the corresponding controls. Moreover, patients in CMM with high expression of GSTO1 were more likely to have a poor prognosis. Through in vitro experiments, silenced GSTO1 resulted in inhibition of CMM cells growth and aggressiveness, increased cell apoptosis, and blocked cell cycle. Finally, the expression of PCNA, p53 and EMT-related proteins were changed due to reduction of GSTO1. CONCLUSIONS To sum up, our outcomes exhibited that weakening GSTO1 reduced the proliferation and mobility of CMM cells, increased the apoptosis ability of CMM cells, and arrested cell cycle at G1 phase, which can be achieved by affecting the expression of PCNA, p53 and the EMT process. This discovery provided a new perspective for elucidating the mechanism of CMM, and offered theoretical support for searching clinical therapeutic targets in the future.
Collapse
Affiliation(s)
- Li-Kun Wang
- Department of Dermatology, North China University of Science and Technology Affiliated Hospital, China.
| | - Hai-Long Yue
- Department of Anesthesiology, Kailuan General Hospital, China
| | - Xiao-Jing Peng
- Department of Anesthesiology, Kailuan General Hospital, China
| | - Shu-Juan Zhang
- Department of Anesthesiology, Kailuan General Hospital, China
| |
Collapse
|
144
|
Lee CH. Reversal of Epithelial-Mesenchymal Transition by Natural Anti-Inflammatory and Pro-Resolving Lipids. Cancers (Basel) 2019; 11:E1841. [PMID: 31766574 PMCID: PMC6966475 DOI: 10.3390/cancers11121841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 02/08/2023] Open
Abstract
Epithelial mesenchymal transition (EMT) is a key process in the progression of malignant cancer. Therefore, blocking the EMT can be a critical fast track for the development of anticancer drugs. In this paper, we update recent research output of EMT and we explore suppression of EMT by natural anti-inflammatory compounds and pro-resolving lipids.
Collapse
Affiliation(s)
- Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 100-715, Korea
| |
Collapse
|
145
|
Abraham AD, Esquer H, Zhou Q, Tomlinson N, Hamill BD, Abbott JM, Li L, Pike LA, Rinaldetti S, Ramirez DA, Lunghofer PJ, Gomez JD, Schaack J, Nemkov T, D'Alessandro A, Hansen KC, Gustafson DL, Messersmith WA, LaBarbera DV. Drug Design Targeting T-Cell Factor-Driven Epithelial-Mesenchymal Transition as a Therapeutic Strategy for Colorectal Cancer. J Med Chem 2019; 62:10182-10203. [PMID: 31675229 DOI: 10.1021/acs.jmedchem.9b01065] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Metastasis is the cause of 90% of mortality in cancer patients. For metastatic colorectal cancer (mCRC), the standard-of-care drug therapies only palliate the symptoms but are ineffective, evidenced by a low survival rate of ∼11%. T-cell factor (TCF) transcription is a major driving force in CRC, and we have characterized it to be a master regulator of epithelial-mesenchymal transition (EMT). EMT transforms relatively benign epithelial tumor cells into quasi-mesenchymal or mesenchymal cells that possess cancer stem cell properties, promoting multidrug resistance and metastasis. We have identified topoisomerase IIα (TOP2A) as a DNA-binding factor required for TCF-transcription. Herein, we describe the design, synthesis, biological evaluation, and in vitro and in vivo pharmacokinetic analysis of TOP2A ATP-competitive inhibitors that prevent TCF-transcription and modulate or reverse EMT in mCRC. Unlike TOP2A poisons, ATP-competitive inhibitors do not damage DNA, potentially limiting adverse effects. This work demonstrates a new therapeutic strategy targeting TOP2A for the treatment of mCRC and potentially other types of cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Dominique A Ramirez
- Clinical Sciences, School of Biomedical Engineering , Colorado State University , Fort Collins , Colorado 80523 , United States
| | - Paul J Lunghofer
- Clinical Sciences, School of Biomedical Engineering , Colorado State University , Fort Collins , Colorado 80523 , United States
| | | | | | | | | | | | - Daniel L Gustafson
- Clinical Sciences, School of Biomedical Engineering , Colorado State University , Fort Collins , Colorado 80523 , United States
| | | | | |
Collapse
|
146
|
Wang L, Liang Y, Li P, Liang Q, Sun H, Xu D, Hu W. Oncogenic Activities Of UBE2S Mediated By VHL/HIF-1α/STAT3 Signal Via The Ubiquitin-Proteasome System In PDAC. Onco Targets Ther 2019; 12:9767-9781. [PMID: 31814735 PMCID: PMC6863183 DOI: 10.2147/ott.s228522] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 10/22/2019] [Indexed: 01/12/2023] Open
Abstract
Purpose Ubiquitin-conjugating enzyme E2S (UBE2S) is important for the development and progression of several types of cancer. However, neither the role of UBE2S in pancreatic cancer nor its mechanism is clear. Methods We analyzed three GEO datasets to obtain 150 differentially expressed genes (DEGs) between pancreatic ductal adenocarcinoma (PDAC) and non-cancerous samples. Moreover, GSEA and mutation analysis were also done for UBE2S. The UBE2S expression in PDAC was measured by immunohistochemistry and qRT-PCR. Colony formation, scratch wound-healing and tumor growth assays were conducted to examine the effect of UBE2S on PDAC cells. Migration was detected using Transwell assay. UBE2S knockdown pancreatic cells were treated with proteasome inhibitor MG132. Immunofluorescence was undertaken for interaction between UBE2S and VHL. The expression of Snail and Twist1 and the changes of HIF-1α/STAT3 pathway were detected by Western blotting. Results The mRNA of UBE2S was significantly upregulated in human pancreatic cancer compared to normal tissues. Immunohistochemistry confirmed that the protein level of UBE2S increased in tissue microarrays (TMAs) and was associated with lymph nodes metastasis and distant metastasis. Conclusion UBE2S could enhance EMT by the VHL/HIF-1α/STAT3 pathway via the ubiquitin-proteasome system. Co-expression of CDC20 may represent a novel and promising therapeutic target for the patients with PDAC.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, Lianyungang Clinical College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang 222000, Jiangsu, People's Republic of China
| | - Yiyi Liang
- Department of Oncology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai Cancer Institute, Shanghai 200127, People's Republic of China
| | - Pengping Li
- Department of Bioinformatics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| | - Qingchun Liang
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, People's Republic of China
| | - Haijun Sun
- Department of Thoracic Surgery, Lianyungang Clinical College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang 222000, Jiangsu, People's Republic of China
| | - Dazhou Xu
- Department of Gastroenterology, Lianyungang Clinical College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang 222000, Jiangsu, People's Republic of China
| | - Wei Hu
- Department of Hepatobiliary Surgery, Lianyungang Clinical College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang 222000, Jiangsu, People's Republic of China
| |
Collapse
|
147
|
Proteomic Technology "Lens" for Epithelial-Mesenchymal Transition Process Identification in Oncology. Anal Cell Pathol (Amst) 2019; 2019:3565970. [PMID: 31781477 PMCID: PMC6855076 DOI: 10.1155/2019/3565970] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/20/2019] [Accepted: 09/10/2019] [Indexed: 02/08/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a complex transformation process that induces local and distant progression of many malignant tumours. Due to its complex array of proteins that are dynamically over-/underexpressed during this process, proteomic technologies gained their place in the EMT research in the last years. Proteomics has identified new molecular pathways of this process and brought important insights to develop new therapy targets. Various proteomic tools and multiple combinations were developed in this area. Out of the proteomic technology armentarium, mass spectrometry and array technologies are the most used approaches. The main characteristics of the proteomic technology used in this domain are high throughput and detection of minute concentration in small samples. We present herein, using various proteomic technologies, the identification in cancer cell lines and in tumour tissue EMT-related proteins, proteins that are involved in the activation of different cellular pathways. Proteomics has brought besides standard EMT markers (e.g., cell-cell adhesion proteins and transcription factors) other future potential markers for improving diagnosis, monitoring evolution, and developing new therapy targets. Future will increase the proteomic role in clinical investigation and validation of EMT-related biomarkers.
Collapse
|
148
|
Cao R, Yuan L, Ma B, Wang G, Qiu W, Tian Y. An EMT-related gene signature for the prognosis of human bladder cancer. J Cell Mol Med 2019; 24:605-617. [PMID: 31657881 PMCID: PMC6933372 DOI: 10.1111/jcmm.14767] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/06/2019] [Accepted: 09/23/2019] [Indexed: 12/24/2022] Open
Abstract
The transition from non-muscle-invasive bladder cancer (NMIBC) to muscle-invasive bladder cancer (MIBC) is detrimental to bladder cancer (BLCA) patients. Here, we aimed to study the underlying mechanism of the subtype transition. Gene set variation analysis (GSVA) revealed the epithelial-mesenchymal transition (EMT) signalling pathway with the most positive correlation in this transition. Then, we built a LASSO Cox regression model of an EMT-related gene signature in BLCA. The patients with high risk scores had significantly worse overall survival (OS) and disease-free survival (DFS) than those with low risk scores. The EMT-related gene signature also performed favourably in the accuracy of prognosis and in the subtype survival analysis. Univariate and multivariate Cox regression analyses demonstrated that the EMT-related gene signature, pathological N stage and age were independent prognostic factors for predicting survival in BLCA patients. Furthermore, the predictive nomogram model was able to effectively predict the outcome of BLCA patients by appropriately stratifying the risk score. In conclusion, we developed a novel EMT-related gene signature that has tumour-promoting effects, acts as a negative independent prognostic factor and might facilitate personalized counselling and treatment in BLCA.
Collapse
Affiliation(s)
- Rui Cao
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lushun Yuan
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Bo Ma
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Qiu
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ye Tian
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
149
|
Fan X, Jin S, Li Y, Khadaroo PA, Dai Y, He L, Zhou D, Lin H. Genetic And Epigenetic Regulation Of E-Cadherin Signaling In Human Hepatocellular Carcinoma. Cancer Manag Res 2019; 11:8947-8963. [PMID: 31802937 PMCID: PMC6801489 DOI: 10.2147/cmar.s225606] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/27/2019] [Indexed: 12/24/2022] Open
Abstract
E-cadherin is well known as a growth and invasion suppressor and belongs to the large cadherin family. Loss of E-cadherin is widely known as the hallmark of epithelial-to-mesenchymal transition (EMT) with the involvement of transcription factors such as Snail, Slug, Twist and Zeb1/2. Tumor cells undergoing EMT could migrate to distant sites and become metastases. Recently, numerous studies have revealed how the expression of E-cadherin is regulated by different kinds of genetic and epigenetic alteration, which are implicated in several crucial transcription factors and pathways. E-cadherin signaling plays an important role in hepatocellular carcinoma (HCC) initiation and progression considering the highly mutated frequency of CTNNB1 (27%). Combining the data from The Cancer Genome Atlas (TCGA) database and previous studies, we have summarized the roles of gene mutations, chromosome instability, DNA methylation, histone modifications and non-coding RNA in E-cadherin in HCC. In this review, we discuss the current understanding of the relationship between these modifications and HCC. Perspectives on E-cadherin-related research in HCC are provided.
Collapse
Affiliation(s)
- Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Shengxi Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yirun Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Parikshit Asutosh Khadaroo
- School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Yili Dai
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Lifeng He
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Daizhan Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hui Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
150
|
Lin S, Zhang R, An X, Li Z, Fang C, Pan B, Chen W, Xu G, Han W. LncRNA HOXA-AS3 confers cisplatin resistance by interacting with HOXA3 in non-small-cell lung carcinoma cells. Oncogenesis 2019; 8:60. [PMID: 31615976 PMCID: PMC6794325 DOI: 10.1038/s41389-019-0170-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022] Open
Abstract
Many studies have indicated that the aberrant expression of long noncoding RNAs (lncRNAs) is responsible for drug resistance, which represents a substantial obstacle for cancer therapy. In the present study, we aimed to investigate the role of the lncRNA HOXA-AS3 in drug resistance and elucidate its underlying mechanisms in non-small-cell lung carcinoma (NSCLC) cells. The role of HOXA-AS3 in drug resistance was demonstrated by the cell counting kit-8 assay (CCK-8), ethynyldeoxyuridine (EDU) assay, and flow cytometry analysis. Tumor xenografts in nude mice were established to evaluate the antitumor effects of HOXA-AS3 knockdown in vivo. Western blotting and quantitative real-time PCR were used to evaluate protein and RNA expression. RNA pull-down assays, mass spectrometry, and RNA immunoprecipitation were performed to confirm the molecular mechanism of HOXA-AS3 in the cisplatin resistance of NSCLC cells. We found that HOXA-AS3 levels increased with cisplatin treatment and knockdown of HOXA-AS3 enhance the efficacy of cisplatin in vitro and in vivo. Mechanistic investigations showed that HOXA-AS3 conferred cisplatin resistance by down-regulating homeobox A3 (HOXA3) expression. Moreover, HOXA-AS3 was demonstrated to interact with both the mRNA and protein forms of HOXA3. In addition, HOXA3 knockdown increased cisplatin resistance and induced epithelial-mesenchymal transition (EMT). Taken together, our findings suggested that additional research into HOXA-AS3 might provide a better understanding of the mechanisms of drug resistance and promote the development of a novel and efficient strategy to treat NSCLC.
Collapse
Affiliation(s)
- Shuang Lin
- Department of Lung Transplantation, Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Rui Zhang
- Department of Internal medicine, Hangzhou Wuyunshan Sanatorium, the Affiliated Hangzhou First People's Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoxia An
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhoubin Li
- Department of Lung Transplantation, Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cheng Fang
- Department of Lung Transplantation, Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Pan
- Department of Lung Transplantation, Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Chen
- Cancer Institute of Integrated traditional Chinese and Western Medicine, Key laboratory of cancer prevention and therapy combining traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310012, China
| | - Guodong Xu
- Department of Cardiovascular Surgery, The Affiliated Hospital, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, 315041, China
| | - Weili Han
- Department of Lung Transplantation, Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|