1
|
Araújo D, Ribeiro E, Amorim I, Vale N. Repurposed Drugs in Gastric Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010319. [PMID: 36615513 PMCID: PMC9822219 DOI: 10.3390/molecules28010319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
Gastric cancer (GC) is one of the major causes of death worldwide, ranking as the fifth most incident cancer in 2020 and the fourth leading cause of cancer mortality. The majority of GC patients are in an advanced stage at the time of diagnosis, presenting a poor prognosis and outcome. Current GC treatment approaches involve endoscopic detection, gastrectomy and chemotherapy or chemoradiotherapy in an adjuvant or neoadjuvant setting. Drug development approaches demand extreme effort to identify molecular mechanisms of action of new drug candidates. Drug repurposing is based on the research of new therapeutic indications of drugs approved for other pathologies. In this review, we explore GC and the different drugs repurposed for this disease.
Collapse
Affiliation(s)
- Diana Araújo
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Irina Amorim
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Correspondence: ; Tel.: +351-220426537
| |
Collapse
|
2
|
Kong F, Ma L, Wang X, You H, Zheng K, Tang R. Regulation of epithelial-mesenchymal transition by protein lysine acetylation. Cell Commun Signal 2022; 20:57. [PMID: 35484625 PMCID: PMC9052664 DOI: 10.1186/s12964-022-00870-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/20/2022] [Indexed: 01/01/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a vital driver of tumor progression. It is a well-known and complex trans-differentiation process in which epithelial cells undergo morphogenetic changes with loss of apical-basal polarity, but acquire spindle-shaped mesenchymal phenotypes. Lysine acetylation is a type of protein modification that favors reversibly altering the structure and function of target molecules via the modulation of lysine acetyltransferases (KATs), as well as lysine deacetylases (KDACs). To date, research has found that histones and non-histone proteins can be acetylated to facilitate EMT. Interestingly, histone acetylation is a type of epigenetic regulation that is capable of modulating the acetylation levels of distinct histones at the promoters of EMT-related markers, EMT-inducing transcription factors (EMT-TFs), and EMT-related long non-coding RNAs to control EMT. However, non-histone acetylation is a post-translational modification, and its effect on EMT mainly relies on modulating the acetylation of EMT marker proteins, EMT-TFs, and EMT-related signal transduction molecules. In addition, several inhibitors against KATs and KDACs have been developed, some of which can suppress the development of different cancers by targeting EMT. In this review, we discuss the complex biological roles and molecular mechanisms underlying histone acetylation and non-histone protein acetylation in the control of EMT, highlighting lysine acetylation as potential strategy for the treatment of cancer through the regulation of EMT. Video Abstract
Collapse
Affiliation(s)
- Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lihong Ma
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xing Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
3
|
Kałafut J, Czerwonka A, Anameriç A, Przybyszewska-Podstawka A, Misiorek JO, Rivero-Müller A, Nees M. Shooting at Moving and Hidden Targets-Tumour Cell Plasticity and the Notch Signalling Pathway in Head and Neck Squamous Cell Carcinomas. Cancers (Basel) 2021; 13:6219. [PMID: 34944837 PMCID: PMC8699303 DOI: 10.3390/cancers13246219] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
Head and Neck Squamous Cell Carcinoma (HNSCC) is often aggressive, with poor response to current therapies in approximately 40-50% of the patients. Current therapies are restricted to operation and irradiation, often combined with a small number of standard-of-care chemotherapeutic drugs, preferentially for advanced tumour patients. Only very recently, newer targeted therapies have entered the clinics, including Cetuximab, which targets the EGF receptor (EGFR), and several immune checkpoint inhibitors targeting the immune receptor PD-1 and its ligand PD-L1. HNSCC tumour tissues are characterized by a high degree of intra-tumour heterogeneity (ITH), and non-genetic alterations that may affect both non-transformed cells, such as cancer-associated fibroblasts (CAFs), and transformed carcinoma cells. This very high degree of heterogeneity likely contributes to acquired drug resistance, tumour dormancy, relapse, and distant or lymph node metastasis. ITH, in turn, is likely promoted by pronounced tumour cell plasticity, which manifests in highly dynamic and reversible phenomena such as of partial or hybrid forms of epithelial-to-mesenchymal transition (EMT), and enhanced tumour stemness. Stemness and tumour cell plasticity are strongly promoted by Notch signalling, which remains poorly understood especially in HNSCC. Here, we aim to elucidate how Notch signal may act both as a tumour suppressor and proto-oncogenic, probably during different stages of tumour cell initiation and progression. Notch signalling also interacts with numerous other signalling pathways, that may also have a decisive impact on tumour cell plasticity, acquired radio/chemoresistance, and metastatic progression of HNSCC. We outline the current stage of research related to Notch signalling, and how this pathway may be intricately interconnected with other, druggable targets and signalling mechanisms in HNSCC.
Collapse
Affiliation(s)
- Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodzki 1, 20-093 Lublin, Poland; (J.K.); (A.C.); (A.A.); (A.P.-P.); (A.R.-M.)
| | - Arkadiusz Czerwonka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodzki 1, 20-093 Lublin, Poland; (J.K.); (A.C.); (A.A.); (A.P.-P.); (A.R.-M.)
| | - Alinda Anameriç
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodzki 1, 20-093 Lublin, Poland; (J.K.); (A.C.); (A.A.); (A.P.-P.); (A.R.-M.)
| | - Alicja Przybyszewska-Podstawka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodzki 1, 20-093 Lublin, Poland; (J.K.); (A.C.); (A.A.); (A.P.-P.); (A.R.-M.)
| | - Julia O. Misiorek
- Department of Molecular Neurooncology, Institute of Bioorganic Chemistry Polish Academy of Sciences, ul. Noskowskiego 12/14, 61-704 Poznan, Poland;
| | - Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodzki 1, 20-093 Lublin, Poland; (J.K.); (A.C.); (A.A.); (A.P.-P.); (A.R.-M.)
| | - Matthias Nees
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodzki 1, 20-093 Lublin, Poland; (J.K.); (A.C.); (A.A.); (A.P.-P.); (A.R.-M.)
- Western Finland Cancer Centre (FICAN West), Institute of Biomedicine, University of Turku, 20101 Turku, Finland
| |
Collapse
|
4
|
Nowak E, Bednarek I. Aspects of the Epigenetic Regulation of EMT Related to Cancer Metastasis. Cells 2021; 10:3435. [PMID: 34943943 PMCID: PMC8700111 DOI: 10.3390/cells10123435] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/28/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) occurs during the pathological process associated with tumor progression and is considered to influence and promote the metastatic cascade. Characterized by loss of cell adhesion and apex base polarity, EMT enhances cell motility and metastasis. The key markers of the epithelial to mesenchymal transition are proteins characteristic of the epithelial phenotype, e.g., E-cadherin, cytokeratins, occludin, or desmoplakin, the concentration and activity of which are reduced during this process. On the other hand, as a result of acquiring the characteristics of mesenchymal cells, an increased amount of N-cadherin, vimentin, fibronectin, or vitronectin is observed. Importantly, epithelial cells undergo partial EMT where some of the cells show both epithelial and mesenchymal characteristics. The significant influence of epigenetic regulatory mechanisms is observed in the gene expression involved in EMT. Among the epigenetic modifications accompanying incorrect genetic reprogramming in cancer are changes in the level of DNA methylation within the CpG islands and posttranslational covalent changes of histone proteins. All observed modifications, which are stable but reversible changes, affect the level of gene expression leading to the development and progression of the disease, and consequently affect the uncontrolled growth of the population of cancer cells.
Collapse
Affiliation(s)
- Ewa Nowak
- Department of Biotechnology and Genetic Engineering, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland;
| | | |
Collapse
|
5
|
Jahani M, Khanahmad H, Nikpour P. Evaluation of the Effects of Valproic Acid Treatment on Cell Survival and Epithelial-Mesenchymal Transition-Related Features of Human Gastric Cancer Cells. J Gastrointest Cancer 2021; 52:676-681. [PMID: 32621111 DOI: 10.1007/s12029-019-00332-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Metastasis is the most important feature of gastric cancer accounting for more than 90% of tumor-related mortality. As one of the main modulators of epithelial-mesenchymal transition (EMT), histone deacetylase inhibitors (HDACI) are considered rational candidates for cancer therapy. Valproic acid (VPA) is a HDACI with reported controversial effects on the EMT. The main aim of the current study was to evaluate the effects of VPA treatment on cell survival and EMT-related features of human gastric cancer cells (AGS). METHODS Methyl-thiazoltetrazolium (MTT) assay was utilized to assess the effect of VPA on the proliferation rate of cells. Apoptotic cell death was detected with Annexin V/PI staining. Migratory ability of cells following VPA treatment was assessed using a Boyden chamber test. The expression of EMT markers in AGS cells was analyzed using quantitative real-time RT-PCR. RESULTS Treatment with VPA significantly inhibited AGS cell proliferation compared with control. An increased rate of early and late apoptotic cells was observed following VPA exposure. It was demonstrated that VPA significantly diminished the cell migratory ability in AGS gastric cancer cells. Furthermore, treatment with VPA significantly decreased the expression of E-cadherin but increased the Vimentin expression. CONCLUSIONS Our results showed that VPA induces apoptosis and inhibits the cell proliferation and the migratory ability of AGS gastric cancer cells and may prove useful in the development of therapeutic agents for human gastric cancer. However, these preliminary findings call for further investigations to clarify the precise molecular mechanisms by which VPA modulates the EMT process in a cell type-specific manner.
Collapse
Affiliation(s)
- Mehrnaz Jahani
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran. .,Child Growth and Development Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan, Iran.
| |
Collapse
|
6
|
He Y, Fan Z, He L, Zhang C, Ping F, Deng M, Liu S, Wang Y, Cheng B, Xia J. Metformin Combined with 4SC-202 Inhibited the Migration and Invasion of OSCC via STAT3/TWIST1. Onco Targets Ther 2020; 13:11019-11029. [PMID: 33149616 PMCID: PMC7605634 DOI: 10.2147/ott.s268851] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/11/2020] [Indexed: 01/27/2023] Open
Abstract
Background Oral squamous cell carcinoma (OSCC), the most common epithelial malignant neoplasm in the head and neck, characterizes with local infiltration and metastasis of lymph nodes. The five-year survival rate of OSCC remains low despite the advances in clinical methods. Thus, it is necessary to develop a new effective therapeutic scheme for OSCC. Our previous results showed that metformin and 4SC-202 synergistically promoted the intrinsic apoptosis of OSCC in vitro and in vivo, but the effects on invasion and migration remained unclear. Methods Human OSCC cell lines HSC6 and CAL33 were cultured with metformin (16 mM) or/and 4SC-202 (0.4 μM) for 72 h. STAT3 inhibitor S31-201 was applied at concentration of 60 μM for 48 h. Wound-healing assays and transwell assays were used to determine the invasion and migration ability of OSCC. qRT-PCR and Western blot were performed to detect mRNA levels and protein levels. Results Metformin or/and 4SC-202 suppressed the migration and invasion of OSCC cells. Importantly, the expression of TWIST1 was suppressed by metformin and 4SC-202, while the invasion and migration inhibitory effects of metformin and 4SC-202 were countered by the overexpression of TWIST1. In addition, the phosphorylation level of STAT3 decreased after the administration of metformin or/and 4SC-202. Furthermore, inhibition of STAT3 by S31-201 suppressed the expression of TWIST1 and led to a decline in migration and invasion of OSCC, while overexpression of TWIST1 attenuated these effects. Conclusion Metformin and 4SC-202 suppressed the invasion and migration of OSCC through inhibition of STAT3/TWIST1, and this scheme can serve as a novel therapeutic strategy for OSCC.
Collapse
Affiliation(s)
- Yuan He
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zhaona Fan
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Lihong He
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Chi Zhang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Fan Ping
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Miao Deng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Suyang Liu
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yanting Wang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|
7
|
Avendaño-Félix M, Aguilar-Medina M, Bermudez M, Lizárraga-Verdugo E, López-Camarillo C, Ramos-Payán R. Refocusing the Use of Psychiatric Drugs for Treatment of Gastrointestinal Cancers. Front Oncol 2020; 10:1452. [PMID: 32923398 PMCID: PMC7456997 DOI: 10.3389/fonc.2020.01452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Gastrointestinal cancers (GICs) are the most common human tumors worldwide. Treatments have limited effects, and increasing global cancer burden makes it necessary to investigate alternative strategies such as drug repurposing. Interestingly, it has been found that psychiatric drugs (PDs) are promising as a new generation of cancer chemotherapies due to their anti-neoplastic properties. This review compiles the state of the art about how PDs have been redirected for cancer therapeutics in GICs. PDs, especially anti-psychotics, anti-depressants and anti-epileptic drugs, have shown effects on cell viability, cell growth, inhibition of proliferation (cell cycle arrest), apoptosis promotion by caspases activation or cytochrome C release, production of reactive oxygen species (ROS) and nuclear fragmentation over esophageal, gastric, colorectal, liver and pancreatic cancers. Additionally, PDs can inhibit neovascularization, invasion and metastasis in a dose-dependent manner. Moreover, they can induce chemosensibilization to 5-fluorouracil and cisplatin and can act synergistically with anti-neoplastic drugs such as gemcitabine, paclitaxel and oxaliplatin. All anti-cancer activities are given by activation or inhibition of pathways such as HDAC1/PTEN/Akt, EGFR/ErbB2/ErbB3, and PI3K/Akt; PI3K-AK-mTOR, HDAC1/PTEN/Akt; Wnt/β-catenin. Further investigations and clinical trials are needed to elucidate all molecular mechanisms involved on anti-cancer activities as well as adverse effects on patients.
Collapse
Affiliation(s)
- Mariana Avendaño-Félix
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| | - Maribel Aguilar-Medina
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| | - Mercedes Bermudez
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| | - Erik Lizárraga-Verdugo
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | - Rosalío Ramos-Payán
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| |
Collapse
|
8
|
Churyukina KA, Zhuze AL, Ivanov AA, Zamulaeva IA. Effects of Dimeric Bisbenzimidazoles and Ionizing Radiation on MCF-7 Breast Cancer Stem Cells. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
9
|
A. Zamulaeva I, A. Churyukina K, N. Matchuk O, A. Ivanov A, O. Saburov V, L. Zhuze A. Dimeric bisbenzimidazoles DB(n) in combination with ionizing radiation decrease number and clonogenic activity of MCF-7 breast cancer stem cells. AIMS BIOPHYSICS 2020. [DOI: 10.3934/biophy.2020024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
10
|
Valproic acid promotes the epithelial-to-mesenchymal transition of breast cancer cells through stabilization of Snail and transcriptional upregulation of Zeb1. Eur J Pharmacol 2019; 865:172745. [PMID: 31639340 DOI: 10.1016/j.ejphar.2019.172745] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/11/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Histone deacetylases (HDACs) can regulate cancer progression and its inhibitors (HDACIs) have been widely used for cancer therapy. Valproic acid (VPA, 2-propylpentanoic acid) can inhibit the class I HDAC and suppress the malignancy of solid cancers. Our present study revealed that 1 mM VPA, which has no effect on cell proliferation, can significantly increase the migration and induce epithelial to mesenchymal transition (EMT) like properties of breast cancer cells. Further, VPA increased the expression of EMT-transcription factors (EMT-TFs) Snail and Zeb1. Knockdown of Snail and Zeb1 can attenuate VPA induced cell migration and EMT. Mechanistically, VPA increased the protein stability of Snail via suppression its phosphorylation at Ser 11. As to Zeb1, VPA can increase its promoter activity and transcription via a HDAC2 dependent manner. Over expression of HDAC2 can block VPA induced expression of Zeb1. Collectively, our data revealed that VPA can trigger the EMT of breast cancer cells via upregulation of Snail and Zeb1. It indicated that more attention should be paid to the effects of VPA on the clinical therapy of breast cancer.
Collapse
|
11
|
Wu Q, Wu Z, Bao C, Li W, He H, Sun Y, Chen Z, Zhang H, Ning Z. Cancer stem cells in esophageal squamous cell cancer. Oncol Lett 2019; 18:5022-5032. [PMID: 31612013 PMCID: PMC6781610 DOI: 10.3892/ol.2019.10900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells (CSCs) are hypothesized to govern the origin, progression, drug resistance, recurrence and metastasis of human cancer. CSCs have been identified in nearly all types of human cancer, including esophageal squamous cell cancer (ESCC). Four major methods are typically used to isolate or enrich CSCs, including: i) fluorescence-activated cell sorting or magnetic-activated cell sorting using cell-specific surface markers; ii) stem cell markers, including aldehyde dehydrogenase 1 family member A1; iii) side population cell phenotype markers; and iv) microsphere culture methods. ESCC stem cells have been identified using a number of these methods. An increasing number of stem cell signatures and pathways have been identified, which have assisted in the clarification of molecular mechanisms that regulate the stemness of ESCC stem cells. Certain viruses, such as human papillomavirus and hepatitis B virus, are also considered to be important in the formation of CSCs, and there is a crosstalk between stemness and viruses-associated genes/pathways, which may suggest a potential therapeutic strategy for the eradication of CSCs. In the present review, findings are summarized along these lines of inquiry.
Collapse
Affiliation(s)
- Qian Wu
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China.,Nurse School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhe Wu
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Cuiyu Bao
- Nurse School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Wenjing Li
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Hui He
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yanling Sun
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zimin Chen
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Hao Zhang
- Basic Medical School, Ji'nan University Medical School, Guangzhou, Guangdong 510632, P.R. China
| | - Zhifeng Ning
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
12
|
Qi G, Lu G, Yu J, Zhao Y, Wang C, Zhang H, Xia Q. Up-regulation of TIF1γ by valproic acid inhibits the epithelial mesenchymal transition in prostate carcinoma through TGF-β/Smad signaling pathway. Eur J Pharmacol 2019; 860:172551. [PMID: 31323225 DOI: 10.1016/j.ejphar.2019.172551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/07/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022]
Abstract
Valproic acid (VPA), one of the histone deacetylase inhibitors, can suppress prostate cancer (PCa) cells epithelial mesenchymal transition (EMT). Transcriptional intermediary factor 1γ (TIF1γ) which is a vital protein molecule that possesses ubiquitination enzyme activity, can mediate TGF-β induced EMT. We aimed to investigate the detailed mechanism between VPA and EMT occurrence in PCa cells to clarify the potential mechanism of TIF1γ involved. In our vitro experiments, we first investigated the effect of VPA on the expression TIF1γ. After TIF1γ was knockdown or overexpressed by related lentivirus, EMT of PCa cells were assessed. When TIF1γ knockdown or overexpress stable cell line were established, cells were treated with additional VPA, EMT index were detected and functional experiments were also conducted to confirm whether VPA inhibited EMT of PCa cells via TIF1γ. The mono-ubiquitination of Smad4 was analyzed simultaneously. In vivo, mice were facilitated with PC3 cells or TIF1γ related knockdown or overexpress virus transfected PC3 cells with or without VPA administration. Results showed that in vitro VPA can increase the expression of TIF1γ and also induce the increase expression of E-cadherin, and the decrease of N-cadherin and vimentin. Knocking down of TIF1γ can effectively block the effect of VPA on EMT and metastasis while overexpression of TIF1γ can strengthen its role. In vivo VPA also showed its anti-growth effect including tumor growth and EMT mediated by TIF1γ coincide with in vitro experiments. In conclusion, VPA inhibits the EMT in PCa cells via up-regulating the expression of TIF1γ and the mono-ubiquitination Smad4. VPA could serve as a promising agent in PCa treatment, with new strategies based on its diverse effects on posttranscriptional regulation.
Collapse
Affiliation(s)
- Guanghui Qi
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; Department of Urology, The First Hospital of Zibo City, Zibo, China
| | - Guoliang Lu
- Department of Urology, Shandong Provincial Western Hospital, Jinan, China
| | - Jianguo Yu
- Department of Urology, The First Hospital of Zibo City, Zibo, China
| | - Yanfang Zhao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Chunhui Wang
- Second Department of Gastroenterology, The First Hospital of Zibo City, Zibo, China
| | - Hongge Zhang
- Third Department of Surgery, Tengzhou Hospital of Traditional Chinese Medicine, Tengzhou, China
| | - Qinghua Xia
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.
| |
Collapse
|
13
|
Zhao Y, Zhu J, Shi B, Wang X, Lu Q, Li C, Chen H. The transcription factor LEF1 promotes tumorigenicity and activates the TGF-β signaling pathway in esophageal squamous cell carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:304. [PMID: 31296250 PMCID: PMC6625065 DOI: 10.1186/s13046-019-1296-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/25/2019] [Indexed: 12/28/2022]
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is the most difficult subtype of esophageal cancer to treat due to the paucity of effective targeted therapy. ESCC is believed to arise from cancer stem cells (CSCs) that contribute to metastasis and chemoresistance. Despite advances in diagnosis and treatment, the prognosis of ESCC patients remains poor. Methods In this study, we applied western blot, quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry, RNA-Seq analysis, luciferase reporter assay, Chip-qPCR, bioinformatics analysis, and a series of functional assays to show the potential role of LEF1 in regulating esophageal CSCs. Results We found that the overexpression of LEF1 was associated with aberrant clinicopathological characteristics and the poor prognosis of ESCC patients. In addition, the elevated expression of LEF1 and OV6 was significantly associated with aberrant clinicopathological features, and poor patient prognosis. Moreover, the overexpression of LEF1 was observed in esophageal CSCs purified by the magnetic sorting of adherent and spheroidal ESCC cells. The increased level of LEF1 in CSCs facilitated the expression of CSC markers, stem cell-like properties, resistance to chemotherapy, and tumorigenicity and increased the percentage of CSCs in ESCC samples. Conversely, the knockdown of LEF1 significantly diminished the self-renewal properties of ESCC. We showed that LEF1 played an important mechanical role in activating the TGF-β signaling pathway by directly binding to the ID1 gene promoter. A positive association between LEF1 and ID1 expression was also observed in clinical ESCC samples. Conclusion Our results indicate that the overexpression of LEF1 promotes a CSC-like phenotype in and the tumorigenicity of ESCC by activating the TGF-β signaling pathway. The inhibition of LEF1 might therefore be a novel therapeutic target to inactivate CSCs and inhibit tumor progression. Electronic supplementary material The online version of this article (10.1186/s13046-019-1296-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Ji Zhu
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Bowen Shi
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Xinyu Wang
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Qijue Lu
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Chunguang Li
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Hezhong Chen
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
14
|
Wawruszak A, Kalafut J, Okon E, Czapinski J, Halasa M, Przybyszewska A, Miziak P, Okla K, Rivero-Muller A, Stepulak A. Histone Deacetylase Inhibitors and Phenotypical Transformation of Cancer Cells. Cancers (Basel) 2019; 11:cancers11020148. [PMID: 30691229 PMCID: PMC6406474 DOI: 10.3390/cancers11020148] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylase inhibitors (HDIs) are a group of potent epigenetic drugs which have been investigated for their therapeutic potential in various clinical disorders, including hematological malignancies and solid tumors. Currently, several HDIs are already in clinical use and many more are on clinical trials. HDIs have shown efficacy to inhibit initiation and progression of cancer cells. Nevertheless, both pro-invasive and anti-invasive activities of HDIs have been reported, questioning their impact in carcinogenesis. The aim of this review is to compile and discuss the most recent findings on the effect of HDIs on the epithelial-mesenchymal transition (EMT) process in human cancers. We have summarized the impact of HDIs on epithelial (E-cadherin, β-catenin) and mesenchymal (N-cadherin, vimentin) markers, EMT activators (TWIST, SNAIL, SLUG, SMAD, ZEB), as well as morphology, migration and invasion potential of cancer cells. We further discuss the use of HDIs as monotherapy or in combination with existing or novel anti-neoplastic drugs in relation to changes in EMT.
Collapse
Affiliation(s)
- Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Joanna Kalafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Jakub Czapinski
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Trojdena 2a St., 02-091 Warsaw, Poland.
| | - Marta Halasa
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Alicja Przybyszewska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Paulina Miziak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| | - Karolina Okla
- The First Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland.
- Tumor Immunology Laboratory, Medical University of Lublin, Staszica 16 St., 20-081 Lublin, Poland.
| | - Adolfo Rivero-Muller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
- Faculty of Science and Engineering, Cell Biology, Abo Akademi University, Tykistokatu 6A, 20520 Turku, Finland.
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland.
| |
Collapse
|
15
|
Tao S, Liu M, Shen D, Zhang W, Wang T, Bai Y. TGF-β/Smads Signaling Affects Radiation Response and Prolongs Survival by Regulating DNA Repair Genes in Malignant Glioma. DNA Cell Biol 2018; 37:909-916. [PMID: 30230914 DOI: 10.1089/dna.2018.4310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
To understand the molecular mechanism underlying the causal relationship between aberrant upregulation of transforming growth factor beta (TGF-β) and radio-resistance in glioma. The mouse glioma cell GL261 was irradiated, and relative expression of TGF-β/Smad signaling genes was determined by real-time PCR and western blotting. The DNA repair response on exogenous TGF-β or LY2109761 was evaluated by quantification of diverse genes by real-time PCR and western blotting. Xenograft mice were employed for in vivo investigation to assess the response to irradiation and LY2109761 either alone or in combination. The expression of DNA repair genes was further determined in the xenograft tumor. The TGF-β/Smad signaling pathway was activated by radiation in the GL261 cell line. The exogenous complement of TGF-β significantly stimulated DNA repair response. Administration of LY2109761 suppressed DNA repair genes. Simultaneous treatment with LY2109761 abrogated the upregulation of DNA repair genes in GL261. In the xenograft tumor model, LY2109761 synergistically improved the therapeutic effect of radiation via improvement of sensitivity. Our data suggested that LY2109761 treatment re-sensitized glioma to radiation via antagonizing TGF-β/Smad-induced DNA repair.
Collapse
Affiliation(s)
- Sichen Tao
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Minli Liu
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Dawei Shen
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Wei Zhang
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Tongxin Wang
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| | - Yunan Bai
- Neurosurgery Department, Yidu Central Hospital of Weifang , Qingzhou, Shandong Province, China
| |
Collapse
|
16
|
Chen Y, Tan W, Wang C. Tumor-associated macrophage-derived cytokines enhance cancer stem-like characteristics through epithelial-mesenchymal transition. Onco Targets Ther 2018; 11:3817-3826. [PMID: 30013362 PMCID: PMC6038883 DOI: 10.2147/ott.s168317] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cells are a small population of cells with the potential for self-renewal and multi-directional differentiation and are an important source of cancer initiation, treatment resistance, and recurrence. Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells lose their epithelial phenotype and convert to mesenchymal cells. Recent studies have shown that cancer cells undergoing EMT can become stem-like cells. Many kinds of tumors are associated with chronic inflammation, which plays a role in tumor progression. Among the various immune cells mediating chronic inflammation, macrophages account for ~30%-50% of the tumor mass. Macrophages are highly infiltrative in the tumor microenvironment and secrete a series of inflammatory factors and cytokines, such as transforming growth factor (TGF)-β, IL-6, IL-10, and tumor necrosis factor (TNF)-α, which promote EMT and enhance the stemness of cancer cells. This review summarizes and discusses recent research findings on some specific mechanisms of tumor-associated macrophage-derived cytokines in EMT and cancer stemness transition, which are emerging targets of cancer treatment.
Collapse
Affiliation(s)
- Yongxu Chen
- Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, Guangdong Province, People's Republic of China, .,School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, People's Republic of China,
| | - Wei Tan
- Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, Guangdong Province, People's Republic of China,
| | - Changjun Wang
- Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, Guangdong Province, People's Republic of China, .,School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, People's Republic of China,
| |
Collapse
|
17
|
HDAC1 triggers the proliferation and migration of breast cancer cells via upregulation of interleukin-8. Biol Chem 2017; 398:1347-1356. [DOI: 10.1515/hsz-2017-0155] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/01/2017] [Indexed: 11/15/2022]
Abstract
AbstractTargeted inhibition of histone deacetylase (HDAC) is one of the potent anticancer therapy approaches. Our data showed that mRNA and protein levels of HDAC1 in breast cancer cells were greater than that in normal fibroblast 3T3 cells and normal epithelial breast MCF10A cells. The mRNA levels of HDAC1 in 75% of breast cancer tissues (18/24) were greater than that in their corresponding adjacent normal tissues. Knockdown of HDAC1 by specific siRNAs can suppress the proliferation and migration of breast cancer cells and inhibit the expression of interleukin-8 (IL-8), while not IL-6. While recombinant IL-8 (rIL-8) can attenuate the suppression effects of si-HDAC1 on the proliferation and migration of breast cancer cells. HDAC1 can positively regulate the transcription and promoter activities of IL-8. While NF-κB and MAPK, two important signals responsible for the transcription of IL-8, did not mediate HDAC1 regulated IL-8 expression. The expression and nuclear translocation of Snail were increased in HDAC1 over expressed breast cancer cells. Targeted inhibition of Snail can attenuate HDAC1 over expression induced cell proliferation and migration. Collectively, our data showed that HDAC1 can trigger the proliferation and migration of breast cancer cells via activation of Snail/IL-8 signals.
Collapse
|
18
|
Zhang C, Yu P, Zhu L, Zhao Q, Lu X, Bo S. Blockade of α7 nicotinic acetylcholine receptors inhibit nicotine-induced tumor growth and vimentin expression in non-small cell lung cancer through MEK/ERK signaling way. Oncol Rep 2017; 38:3309-3318. [PMID: 29039603 PMCID: PMC5783576 DOI: 10.3892/or.2017.6014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 09/07/2017] [Indexed: 12/22/2022] Open
Abstract
Nicotine can stimulate the progression of non-small cell lung cancer (NSCLC) through nicotinic acetylcholine receptors (nAChRs). The persistent proliferation of cancer cells is one of the key effects of nicotinic signaling. The present study aimed to clarify the mechanism of nicotine-induced proliferation in NSCLCs at the receptor subtype level. We have previously reported that there are various subtypes of nicotinic receptors expressed in NSCLC cell lines. In the present study, we demonstrated that blocking α7nAChRs agonized by nicotine could suppress the proliferation of H1299 cells in vitro and decrease H1299 tumor xenograft growth in nude mice. During this process, the expression of vimentin was also markedly attenuated, concomitant with the decreased expression of α7nAChR. These results were ascertained by knocking down the α7nAChR gene to abolish receptor functioning. Furthermore, under the stimulation of nicotine, the MEK/ERK signaling pathway was found to be inhibited when cells were treated with an antagonist of α7nAChR or an inhibitor of MEK. Collectively the results indicate that the changes in proliferation and vimentin expression of H1299 cells in response to α7nAChR stimulation are mediated by the MEK/ERK pathway. These findings demonstrate that α7nAChR plays an important role in H1299 cell proliferation, tumor growth and expression of vimentin. Therefore, blocking α7nAChRs in NSCLC may be a potential adjuvant therapy for the targeted treatment of NSCLC.
Collapse
Affiliation(s)
- Chun Zhang
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Ping Yu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Qingnan Zhao
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Xiaotong Lu
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Shuhong Bo
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
19
|
Targeting epithelial-mesenchymal plasticity in cancer: clinical and preclinical advances in therapy and monitoring. Biochem J 2017; 474:3269-3306. [PMID: 28931648 DOI: 10.1042/bcj20160782] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/01/2017] [Accepted: 08/07/2017] [Indexed: 02/07/2023]
Abstract
The concept of epithelial-mesenchymal plasticity (EMP), which describes the dynamic flux within the spectrum of phenotypic states that invasive carcinoma cells may reside, is being increasingly recognised for its role in cancer progression and therapy resistance. The myriad of events that are able to induce EMP, as well as the more recently characterised control loops, results in dynamic transitions of cancerous epithelial cells to more mesenchymal-like phenotypes through an epithelial-mesenchymal transition (EMT), as well as the reverse transition from mesenchymal phenotypes to an epithelial one. The significance of EMP, in its ability to drive local invasion, generate cancer stem cells and facilitate metastasis by the dissemination of circulating tumour cells (CTCs), highlights its importance as a targetable programme to combat cancer morbidity and mortality. The focus of this review is to consolidate the existing knowledge on the strategies currently in development to combat cancer progression via inhibition of specific facets of EMP. The prevalence of relapse due to therapy resistance and metastatic propensity that EMP endows should be considered when designing therapy regimes, and such therapies should synergise with existing chemotherapeutics to benefit efficacy. To further improve upon EMP-targeted therapies, it is imperative to devise monitoring strategies to assess the impact of such treatments on EMP-related phenomenon such as CTC burden, chemosensitivity/-resistance and micrometastasis in patients.
Collapse
|
20
|
Garg M. Epithelial plasticity and cancer stem cells: Major mechanisms of cancer pathogenesis and therapy resistance. World J Stem Cells 2017; 9:118-126. [PMID: 28928908 PMCID: PMC5583530 DOI: 10.4252/wjsc.v9.i8.118] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/22/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been linked with aggressive tumor biology and therapy resistance. It plays central role not only in the generation of cancer stem cells (CSCs) but also direct them across the multiple organ systems to promote tumor recurrence and metastasis. CSCs are reported to express stem cell genes as well as specific cell surface markers and allow aberrant differentiation of progenies. It facilitates cancer cells to leave primary tumor, acquire migratory characteristics, grow into new environment and develop radio-chemo-resistance. Based on the current information, present review discusses and summarizes the recent advancements on the molecular mechanisms that derive epithelial plasticity and its major role in generating a subset of tumor cells with stemness properties and pathophysiological spread of tumor. This paper further highlights the critical need to examine the regulation of EMT and CSC pathways in identifying the novel probable therapeutic targets. These improved therapeutic strategies based on the co-administration of inhibitors of EMT, CSCs as well as differentiated tumor cells may provide improved anti-neoplastic response with no tumor relapse.
Collapse
Affiliation(s)
- Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| |
Collapse
|
21
|
Cancer stem cells with increased metastatic potential as a therapeutic target for esophageal cancer. Semin Cancer Biol 2017; 44:60-66. [DOI: 10.1016/j.semcancer.2017.03.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/12/2017] [Accepted: 03/15/2017] [Indexed: 02/07/2023]
|
22
|
Liu S, Liang B, Jia H, Jiao Y, Pang Z, Huang Y. Evaluation of cell death pathways initiated by antitumor drugs melatonin and valproic acid in bladder cancer cells. FEBS Open Bio 2017; 7:798-810. [PMID: 28593135 PMCID: PMC5458469 DOI: 10.1002/2211-5463.12223] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023] Open
Abstract
Effective drug combinations have the potential to strengthen therapeutic efficacy and combat drug resistance. Both melatonin and valproic acid (VPA) exhibit antitumor activities in various cancer cells. The aim of this study was to evaluate the cell death pathways initiated by anticancer combinatorial effects of melatonin and VPA in bladder cancer cells. The results demonstrated that the combination of melatonin and VPA leads to significant synergistic growth inhibition of UC3 bladder cancer cells. Gene expression studies revealed that cotreatment with melatonin and VPA triggered the up-regulation of certain genes related to apoptosis (TNFRSF10A and TNFRSF10B), autophagy (BECN, ATG3 and ATG5) and necrosis (MLKL, PARP-1 and RIPK1). The combinatorial treatment increased the expression of endoplasmic reticulum (ER)-stress-related genes ATF6, IRE1, EDEM1 and ERdj4. Cotreatment with melatonin and VPA enhanced the expression of E-cadherin, and decreased the expression of N-cadherin, Fibronectin, Snail and Slug. Furthermore, the Wnt pathway and Raf/MEK/ERK pathway were activated by combinatorial treatment. However, the effects on the expression of certain genes were not further enhanced in cells following combinatorial treatment in comparison to individual treatment of melatonin or VPA. In summary, these findings provided evidence that cotreatment with melatonin and VPA exerted increased cytotoxicity by regulating cell death pathways in UC3 bladder cancer cells, but the clinical significance of combinatorial treatment still needs to be further exploited.
Collapse
Affiliation(s)
- Siwei Liu
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Bilin Liang
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Huiting Jia
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Yuhan Jiao
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Zhongqiu Pang
- College of Life and Health Sciences Northeastern University Shenyang China
| | - Yongye Huang
- College of Life and Health Sciences Northeastern University Shenyang China
| |
Collapse
|