1
|
Chen Y, Chen S, Zhang J, Hu X, Li N, Liu Z, Huang L, Yu J, Zhang Y, Lin X, Xu Z, Fang Y, Chen Z, Guo Y, Chen B. Electroacupuncture pre-treatment exerts a protective effect on LPS-induced cardiomyopathy in mice through the delivery of miR-381 via exosomes. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167208. [PMID: 38701956 DOI: 10.1016/j.bbadis.2024.167208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
OBJECTIVE This study aims to investigate the cardiac protective effects and molecular mechanisms of electroacupuncture (EA) pre-treatment in lipopolysaccharide (LPS)-Induced Cardiomyopathy. METHODS AND RESULTS Pre-treatment with EA was performed 30 min before intraperitoneal injection of LPS. Cardiac function changes in mice of the EA + LPS group were observed using electrocardiography, echocardiography, and enzyme linked immunosorbent assay (ELISA) and compared with the LPS group. The results demonstrated that EA pre-treatment significantly improved the survival rate of septic mice, alleviated the severity of endotoxemia, and exhibited notable cardiac protective effects. These effects were characterized by a reduction in ST-segment elevation on electrocardiography, an increase in ejection fraction (EF) and fraction shortening (FS) on echocardiography and a decrease in the expression of serum cardiac troponin I (cTn-I) levels. Serum exosomes obtained after EA pre-treatment were extracted and administered to septic mice, revealing significant cardiac protective effects of EA-derived exosomes. Furthermore, the antagonism of circulating exosomes in mice markedly suppressed the cardiac protective effects conferred by EA pre-treatment. Analysis of serum exosomes using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed a significant upregulation of miR-381 expression after EA pre-treatment. Inhibition or overexpression of miR-381 through serotype 9 adeno-associated virus (AAV9)-mediated gene delivery demonstrated that overexpression of miR-381 exerted a cardiac protective effect, while inhibition of miR-381 significantly attenuated the cardiac protective effects conferred by EA pre-treatment. CONCLUSIONS Our research findings have revealed a novel endogenous cardiac protection mechanism, wherein circulating exosomes derived from EA pre-treatment mitigate LPS-induced cardiac dysfunction via miR-381.
Collapse
Affiliation(s)
- Yong Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300102, China
| | - Shuangli Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingyu Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiyou Hu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ningcen Li
- Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 500515, China
| | - Zhen Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lihong Huang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300102, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300102, China
| | - Xiaowei Lin
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zelin Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Bo Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
2
|
Le MT, Nguyen HT, Nguyen XH, Do XH, Mai BT, Ngoc Nguyen HT, Trang Than UT, Nguyen TH. Regulation and therapeutic potentials of microRNAs to non-small cell lung cancer. Heliyon 2023; 9:e22080. [PMID: 38058618 PMCID: PMC10696070 DOI: 10.1016/j.heliyon.2023.e22080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, accounting for 80%-85% of total cases and leading to millions of deaths worldwide. Drug resistance is the primary cause of treatment failure in NSCLC, which urges scientists to develop advanced approaches for NSCLC treatment. Among novel approaches, the miRNA-based method has emerged as a potential approach as it allows researchers to modulate target gene expression. Subsequently, cell behaviors are altered, which leads to the death and the depletion of cancer cells. It has been reported that miRNAs possess the capacity to regulate multiple genes that are involved in various signaling pathways, including the phosphoinositide 3-kinase, receptor tyrosine kinase/rat sarcoma virus/mitogen-activated protein kinase, wingless/integrated, retinoblastoma, p53, transforming growth factor β, and nuclear factor-kappa B pathways. Dysregulation of these signaling pathways in NSCLC results in abnormal cell proliferation, tissue invasion, and drug resistance while inhibiting apoptosis. Thus, understanding the roles of miRNAs in regulating these signaling pathways may enable the development of novel NSCLC treatment therapies. However, a comprehensive review of potential miRNAs in NSCLC treatment has been lacking. Therefore, this review aims to fill the gap by summarizing the up-to-date information on miRNAs regarding their targets, impact on cancer-associated pathways, and prospective outcomes in treating NSCLC. We also discuss current technologies for delivering miRNAs to the target cells, including virus-based, non-viral, and emerging extracellular vesicle-based delivery systems. This knowledge will support future studies to develop an innovative miRNA-based therapy and select a suitable carrier to treat NSCLC effectively.
Collapse
Affiliation(s)
- Mai Thi Le
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, 100000, Viet Nam
| | - Huyen-Thu Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Xuan-Hung Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- College of Health Sciences, Vin University, Hanoi, 100000, Viet Nam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Xuan-Hai Do
- Department of Gastroenterology, 108 Military Central Hospital, Hanoi, Viet Nam
| | - Binh Thanh Mai
- Department of Practical and Experimental Surgery, Vietnam Military Medical University, 160 Phung Hung Street, Phuc La, Ha Dong, Hanoi, Viet Nam
| | - Ha Thi Ngoc Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Uyen Thi Trang Than
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Thanh-Hong Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| |
Collapse
|
3
|
Li S, Wang Z, Geng R, Zhang W, Wan H, Kang X, Guo S. TMEM16A ion channel: A novel target for cancer treatment. Life Sci 2023; 331:122034. [PMID: 37611692 DOI: 10.1016/j.lfs.2023.122034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Cancer draws attention owing to the high morbidity and mortality. It is urgent to develop safe and effective cancer therapeutics. The calcium-activated chloride channel TMEM16A is widely distributed in various tissues and regulates physiological functions. TMEM16A is abnormally expressed in several cancers and associate with tumorigenesis, metastasis, and prognosis. Knockdown or inhibition of TMEM16A in cancer cells significantly inhibits cancer development. Therefore, TMEM16A is considered as a biomarker and therapeutic target for some cancers. This work reviews the cancers associated with TMEM16A. Then, the molecular mechanism of TMEM16A overexpression in cancer was analyzed, and the possible signal transduction mechanism of TMEM16A regulating cancer development was summarized. Finally, TMEM16A inhibitors with anticancer effect and their anticancer mechanism were concluded. We hope to provide new ideas for pharmacological studies on TMEM16A in cancer.
Collapse
Affiliation(s)
- Shuting Li
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Zhichen Wang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Ruili Geng
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Weiwei Zhang
- School of Basic Medical Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Haifu Wan
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China
| | - Xianjiang Kang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China.
| | - Shuai Guo
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China.
| |
Collapse
|
4
|
Wu D, Tian S, Zhu W. Modulating multidrug resistance to drug-based antitumor therapies through NF-κB signaling pathway: mechanisms and perspectives. Expert Opin Ther Targets 2023; 27:503-515. [PMID: 37314372 DOI: 10.1080/14728222.2023.2225767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/12/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Despite the advances made in cancer treatment in the past decades, therapeutic efficacy is still quite challenging, partially due to the emergence of multidrug resistance (MDR). It is crucial to decipher the underlying mechanisms of resistance in order to develop new therapeutic strategies for cancer patients. Previous studies have shown that activation of nuclear factor-κB (NF-κB) plays key roles in various cellular processes including proliferation, anti-apoptosis, metastasis, invasion, and chemoresistance. AREAS COVERED In this review, we conduct an integrated analysis of the evidence suggesting the vital roles of the NF-κB signaling pathway in MDR during chemotherapy, immunotherapy, endocrine, and targeted therapy. A literature search was performed on NF-κB and drug resistance in PubMed up to February 2023. EXPERT OPINION This review summarizes that the NF-κB signaling pathway exhibits a crucial role in enhancing drug resistance in chemotherapy, immunotherapy, endocrine, and targeted therapy. The application of combination therapy with existing antineoplastic drugs and a safe NF-κB inhibitor could become a promising strategy in cancer treatment. A better understanding of the pathway and mechanisms of drug resistance may help exploit safer and more effective NF-κB-targeting agents for clinical use in the future.
Collapse
Affiliation(s)
- Dapeng Wu
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Sai Tian
- Department of Pediatric Clinic, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Wenjing Zhu
- Clinical Research Center, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
- Department of Respiratory and Critical Care Medicine, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| |
Collapse
|
5
|
miR-381 Inhibits Proliferation and Invasion of Non-Small-Cell Cancer Cells by Targeting USP39. DISEASE MARKERS 2022; 2022:2195393. [PMID: 36046375 PMCID: PMC9420621 DOI: 10.1155/2022/2195393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/29/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022]
Abstract
It is known that miR-381 plays a therapeutic role in a variety of cancers, but the regulatory mechanism of miR-381 in the treatment of lung cancer remains unclear. This study is aimed at exploring the expression level and mechanism of miR-381 in lung cancer. In this experiment, quantitative real-time PCR (qRT-PCR), western blot, and other methods were used to detect the expression of miR-381 and ubiquitin-specific protease 39 (USP39) in lung cancer tissues. The target genes of miR-381 were predicted by bioinformatics techniques, and the targeting relationship between miR-381 and USP39 was verified by the dual-luciferase reporting method. The expression levels of miR-381 and USP39 were adjusted to verify the effect of miR-381 on the expression of USP39 gene. The effect of miR-381 expression on proliferation of lung cancer cells was verified by cell proliferation and invasion experiments. miR-381 was downregulated in non-small-cell lung cancer tissues and cell lines, while USP39 was upregulated. The dual-luciferase reporter gene assay showed that miR-381 and USP39 had targeted binding sites. After transfection with miR-381 mimics, USP39 expression was significantly decreased, cell proliferation decreased, and apoptosis increased. After transfection with miR-381 inhibitor, USP39 expression was significantly increased, cell proliferation increased, and cell apoptosis decreased. Overexpression of USP39 significantly increased the invasion ability and cell survival curve (p < 0.05). In conclusion, overexpression of miR-381 can regulate the expression of USP39, inhibit the proliferation and invasion of cancer cells, and induce apoptosis of cancer cells. This may provide a new perspective and strategy for targeted therapy of non-small-cell lung cancer.
Collapse
|
6
|
Konoshenko M, Lansukhay Y, Krasilnikov S, Laktionov P. MicroRNAs as Predictors of Lung-Cancer Resistance and Sensitivity to Cisplatin. Int J Mol Sci 2022; 23:7594. [PMID: 35886942 PMCID: PMC9321818 DOI: 10.3390/ijms23147594] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Platinum-based chemotherapy, cisplatin (DDP) specifically, is the main strategy for treating lung cancer (LC). However, currently, there is a lack of predictive drug-resistance markers, and there is increased interest in the development of a reliable and sensitive panels of markers for DDP chemotherapy-effectiveness prediction. MicroRNAs represent a perspective pool of markers for chemotherapy effectiveness. OBJECTIVES Data on miRNAs associated with LC DDP chemotherapy response are summarized and analyzed. MATERIALS AND METHODS A comprehensive review of the data in the literature and an analysis of bioinformatics resources were performed. The gene targets of miRNAs, as well as their reciprocal relationships with miRNAs, were studied using several databases. RESULTS AND DISCUSSION The complex analysis of bioinformatics resources and the literature indicated that the expressions of 12 miRNAs have a high predictive potential for LC DDP chemotherapy responses. The obtained information was discussed from the point of view of the main mechanisms of LC chemoresistance. CONCLUSIONS An overview of the published data and bioinformatics resources, with respect to the predictive microRNA markers of chemotherapy response, is presented in this review. The selected microRNAs and gene panel have a high potential for predicting LC DDP sensitiveness or DDP resistance as well as for the development of a DDP co-therapy.
Collapse
Affiliation(s)
- Maria Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Yuriy Lansukhay
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Sergey Krasilnikov
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Pavel Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| |
Collapse
|
7
|
Paul S, Ruiz-Manriquez LM, Ambriz-Gonzalez H, Medina-Gomez D, Valenzuela-Coronado E, Moreno-Gomez P, Pathak S, Chakraborty S, Srivastava A. Impact of smoking-induced dysregulated human miRNAs in chronic disease development and their potential use in prognostic and therapeutic purposes. J Biochem Mol Toxicol 2022; 36:e23134. [PMID: 35695328 DOI: 10.1002/jbt.23134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 04/20/2022] [Accepted: 05/29/2022] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are evolutionary conserved small noncoding RNA molecules with a significant ability to regulate gene expression at the posttranscriptional level either through translation repression or messenger RNA degradation. miRNAs are differentially expressed in various pathophysiological conditions, affecting the course of the disease by modulating several critical target genes. As the persistence of irreversible molecular changes caused by cigarette smoking is central to the pathogenesis of various chronic diseases, several studies have shown its direct correlation with the dysregulation of different miRNAs, affecting numerous essential biological processes. This review provides an insight into the current status of smoking-induced miRNAs dysregulation in chronic diseases such as COPD, atherosclerosis, pulmonary hypertension, and different cancers and explores the diagnostic/prognostic potential of miRNA-based biomarkers and their efficacy as therapeutic targets.
Collapse
Affiliation(s)
- Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, San Pablo, Queretaro, Mexico
| | - Luis M Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, San Pablo, Queretaro, Mexico
| | - Hector Ambriz-Gonzalez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, San Pablo, Queretaro, Mexico
| | - Daniel Medina-Gomez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, San Pablo, Queretaro, Mexico
| | - Estefania Valenzuela-Coronado
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, San Pablo, Queretaro, Mexico
| | - Paloma Moreno-Gomez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, San Pablo, Queretaro, Mexico
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, India
| | - Samik Chakraborty
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aashish Srivastava
- Section of Bioinformatics, Clinical Laboratory, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
8
|
Loren P, Saavedra N, Saavedra K, De Godoy Torso N, Visacri MB, Moriel P, Salazar LA. Contribution of MicroRNAs in Chemoresistance to Cisplatin in the Top Five Deadliest Cancer: An Updated Review. Front Pharmacol 2022; 13:831099. [PMID: 35444536 PMCID: PMC9015654 DOI: 10.3389/fphar.2022.831099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/16/2022] [Indexed: 12/02/2022] Open
Abstract
Cisplatin (DDP) is a well-known anticancer drug used for the treatment of numerous human cancers in solid organs, including bladder, breast, cervical, head and neck squamous cell, ovarian, among others. Its most important mode of action is the DNA-platinum adducts formation, inducing DNA damage response, silencing or activating several genes to induce apoptosis; these mechanisms result in genetics and epigenetics modifications. The ability of DDP to induce tumor cell death is often challenged by the presence of anti-apoptotic regulators, leading to chemoresistance, wherein many patients who have or will develop DDP-resistance. Cancer cells resist the apoptotic effect of chemotherapy, being a problem that severely restricts the successful results of treatment for many human cancers. In the last 30 years, researchers have discovered there are several types of RNAs, and among the most important are non-coding RNAs (ncRNAs), a class of RNAs that are not involved in protein production, but they are implicated in gene expression regulation, and representing the 98% of the human genome non-translated. Some ncRNAs of great interest are long ncRNAs, circular RNAs, and microRNAs (miRs). Accumulating studies reveal that aberrant miRs expression can affect the development of chemotherapy drug resistance, by modulating the expression of relevant target proteins. Thus, identifying molecular mechanisms underlying chemoresistance development is fundamental for setting strategies to improve the prognosis of patients with different types of cancer. Therefore, this review aimed to identify and summarize miRs that modulate chemoresistance in DDP-resistant in the top five deadliest cancer, both in vitro and in vivo human models.
Collapse
Affiliation(s)
- Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco, Chile
| | - Nicolás Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco, Chile
| | - Kathleen Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco, Chile
| | | | | | - Patricia Moriel
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Luis A Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
9
|
Sha H, Gan Y, Xu F, Zhu Y, Zou R, Peng W, Wu Z, Ma R, Wu J, Feng J. MicroRNA-381 in human cancer: Its involvement in tumour biology and clinical applications potential. J Cell Mol Med 2022; 26:977-989. [PMID: 35014178 PMCID: PMC8831973 DOI: 10.1111/jcmm.17161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 11/14/2021] [Accepted: 12/16/2021] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) are small non‐coding RNAs that regulate gene expression at the post‐transcriptional level. MiRNAs are involved in the development and progression of a wide range of cancers. Among such cancer‐associated miRNAs, miR‐381 has been a major focus of research. The expression pattern and role of miR‐381 vary among different cancer types. MiR‐381 modulates various cellular behaviours in cancer, including proliferation, apoptosis, cell cycle progression, migration and invasion. MiR‐381 is also involved in angiogenesis and lymphangiogenesis, as well as in the resistance to chemotherapy and radiotherapy. MiR‐381 itself is regulated by several factors, such as long noncoding RNAs, circular RNAs and cytokines. Aberrant expression of miR‐381 in blood samples indicates that it can be used as a diagnostic marker in cancer. Tissue miR‐381 expression may serve as a prognostic factor for the clinicopathological characteristics of cancers and survival of patients. Metformin and icaritin regulate miR‐381 expression and present anticancer properties. This review comprehensively summarizes the effect of miR‐381 on tumour biological behaviours, as well as the clinical application potential of miR‐381 for the treatment of cancer.
Collapse
Affiliation(s)
- Huanhuan Sha
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yujie Gan
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Xu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yue Zhu
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Renrui Zou
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiwei Peng
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhiya Wu
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rong Ma
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianzhong Wu
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jifeng Feng
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
miR-874-3p mitigates cisplatin resistance through modulating NF-κB/inhibitor of apoptosis protein signaling pathway in epithelial ovarian cancer cells. Mol Cell Biochem 2021; 477:307-317. [PMID: 34716858 DOI: 10.1007/s11010-021-04271-6] [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] [Received: 08/04/2021] [Accepted: 09/29/2021] [Indexed: 10/19/2022]
Abstract
The resistance to cisplatin, the most common platinum chemotherapy drug, may confine the efficacy of treatment in epithelial ovarian cancer patients. Aberrant expression of inhibitor of apoptosis proteins set the stage for resistance to cisplatin in EOC; besides, chemosensitivity in EOC can be chalked up to dysregulation of specific miRNAs. Herein, we investigated whether there is a potential correlation between miR-874-3p and the X-chromosome-linked inhibitor of apoptosis, a member of the IAP protein family in cisplatin-resistant EOC cells. The lower expression of miR-874-3p was found in SKOV3-DDP cells; it was also in association with cisplatin-resistance in EOC cells. XIAP was found to contribute to developing platinum resistance and is an authentic target for miR-874-3p in SKOV3-DDP cells. Consistently, restoration of miR-874-3p expression reversed cisplatin resistance in such cells by modulating XIAP and NF-κB/Survivin signaling pathway. Besides, siRNA knock down of XIAP in SKOV3-DDP cells had an anti-migratory effect like those with miR-874 overexpression. Importantly, the enforced expression of XIAP rescued SKOV3-DDP cells from the cytotoxic effects of miR-874-3p. Finally, miR-874-3p sensitized EOC cells to cisplatin-induced apoptosis, at least in part, through targeting XIAP. The cytotoxic effects of miR-874-3p can be attributed to the targeting XIAP in cisplatin-resistant EOC cells. We believe that the combination of cisplatin with miR-874-3p may make a potential strategy to reverse cisplatin resistance.
Collapse
|
11
|
Wu JY, Weng YS, Chiou YC, Hsu FT, Chiang IT. Induction of Apoptosis and Inhibition of EGFR/NF-κB Signaling Are Associated With Regorafenib-sensitized Non-small Cell Lung Cancer to Cisplatin. In Vivo 2021; 35:2569-2576. [PMID: 34410944 DOI: 10.21873/invivo.12539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/04/2021] [Accepted: 06/15/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM The combination of regorafenib with cisplatin/pemetrexed has indicated controllable safety and encouraging antitumor activity in non-small cell lung cancer (NSCLC) patients. However, the anti-NSCLC effects and action mechanisms of regorafenib combined with cisplatin is ambiguous. The major goal of the study was to study the inhibitory effects and action mechanisms of regorafenib combined with cisplatin in NSCLC cells. MATERIALS AND METHODS Cell viability, flow cytometry, immunofluorescence staining, western blotting, migration, and invasion assays were employed to verify the anti-NSCLC effects and mechanisms of regorafenib in combination with cisplatin. RESULTS Cisplatin-induced epidermal growth factor receptor (EGFR)/nuclear factor κB (NF-κB) signaling was effectively inhibited by regorafenib treatment. Regorafenib, erlotinib (EGFR inhibitor) and QNZ (NF-κB inhibitor) may all enhance the cytotoxicity effect of cisplatin. The invasion ability was effectively decreased by combination treatment. Caspase-dependent and -independent apoptosis was activated by cisplatin combined with regorafenib. CONCLUSION Apoptosis induction and EGFR/NF-κB inactivation correlate with regorafenib-enhanced anti-NSCLC efficacy of cisplatin. This study provides evidence of the therapeutic efficacy of regorafenib in combination with cisplatin on NSCLC.
Collapse
Affiliation(s)
- Jeng-Yuan Wu
- Department of Thoracic Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan, R.O.C.,School of Medicine, Tzu Chi University, Hualien, Taiwan, R.O.C
| | - Yueh-Shan Weng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Yi-Chou Chiou
- Chest Medicine Department, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - I-Tsang Chiang
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.; .,Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.,Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C
| |
Collapse
|
12
|
Yu YZ, Mu Q, Ren Q, Xie LJ, Wang QT, Wang CP. miR-381-3p suppresses breast cancer progression by inhibition of epithelial-mesenchymal transition. World J Surg Oncol 2021; 19:230. [PMID: 34362391 PMCID: PMC8348871 DOI: 10.1186/s12957-021-02344-w] [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: 04/13/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Background Accumulating evidence indicates that miRNAs are involved in multiple cellular functions and participate in various cancer development and progression, including breast cancer. Methods We aimed to investigate the role of miR-381-3p in breast cancer. The expression level of miR-381-3p and EMT transcription factors was examined by quantitative real-time PCR (qRT-PCR). The effects of miR-381-3p on breast cancer proliferation and invasion were determined by Cell Counting Kit-8 (CCK-8), colony formation, and transwell assays. The regulation of miR-381-3p on its targets was determined by dual-luciferase analysis, qRT-PCR, and western blot. Results We found that the expression of miR-381-3p was significantly decreased in breast cancer tissues and cell lines. Overexpression of miR-381-3p inhibited breast cancer proliferation and invasion, whereas knockdown of miR-381-3p promoted cell proliferation and invasion in MDA-MB-231 and SKBR3 cells. Mechanistically, overexpression of miR-381-3p inhibited breast cancer epithelial–mesenchymal transition (EMT). Both Sox4 and Twist1 were confirmed as targets of miR-381-3p. Moreover, transforming growth factor-β (TGF-β) could reverse the effects of miR-381-3p on breast cancer progression. Conclusions Our observation suggests that miR-381-3p inhibits breast cancer progression and EMT by regulating the TGF-β signaling via targeting Sox4 and Twist1.
Collapse
Affiliation(s)
- Yong-Zheng Yu
- The First Department of Breast Surgery, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, 266042, China
| | - Qiang Mu
- The First Department of Breast Surgery, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, 266042, China
| | - Qian Ren
- The First Department of Breast Surgery, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, 266042, China
| | - Li-Juan Xie
- Department of Ophthalmology, Qingdao Women and Children's Hospital, Qingdao University, Qingdao, 266034, China
| | - Qi-Tang Wang
- The First Department of Breast Surgery, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, 266042, China
| | - Cui-Ping Wang
- The First Department of Breast Surgery, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, 266042, China.
| |
Collapse
|
13
|
Martínez-Rivera V, Cárdenas-Monroy CA, Millan-Catalan O, González-Corona J, Huerta-Pacheco NS, Martínez-Gutiérrez A, Villavicencio-Queijeiro A, Pedraza-Lara C, Hidalgo-Miranda A, Bravo-Gómez ME, Pérez-Plasencia C, Guardado-Estrada M. Dysregulation of miR-381-3p and miR-23b-3p in skeletal muscle could be a possible estimator of early post-mortem interval in rats. PeerJ 2021; 9:e11102. [PMID: 33986977 PMCID: PMC8086579 DOI: 10.7717/peerj.11102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/22/2021] [Indexed: 01/01/2023] Open
Abstract
Background The post-mortem interval (PMI) is the time elapsed since the dead of an individual until the body is found, which is relevant for forensic purposes. The miRNAs regulate the expression of some genes; and due to their small size, they can better support degradation, which makes them suitable for forensic analysis. In the present work, we evaluated the gene expression of miR-381-3p, miR-23b-3p, and miR-144-3p in skeletal muscle in a murine model at the early PMI. Methods We designed a rat model to evaluate the early PMI under controlled conditions. This model consisted in 25 rats divided into five groups of rats, that correspond to the 0, 3, 6, 12 and 24 hours of PMI. The 0 h-PMI was considered as the control group. Muscle samples were taken from each rat to analyze the expression of miR-381-3p, miR-23b-3p, and miR-144-3p by quantitative RT-PCR. The gene expression of each miRNA was expressed as Fold Change (FC) and compared among groups. To find the targets of these miRNAs and the pathways where they participate, we performed an in-silico analysis. From the gene targets of miR-381-3p identified in the silico analysis, the EPC1 gene was selected for gene expression analysis by quantitative RT-PCR in these samples. Also, to evaluate if miR-381-3p could predict the early PMI, a mixed effects model was calculated using its gene expression. Results An upregulation of miR-381-3p was found at 24 h-PMI compared with the control group of 0 h-PMI and (FC = 1.02 vs. FC = 1.96; p = 0.0079). This was the opposite for miR-23b-3p, which had a down-regulation at 24 h-PMI compared to 0 h-PMI (FC = 1.22 vs. FC = 0.13; p = 0.0079). Moreover, the gene expression of miR-381-3p increased throughout the first 24 h of PMI, contrary to miR-23b-3p. The targets of these two miRNAs, participate in biological pathways related to hypoxia, apoptosis, and RNA metabolism. The gene expression of EPC1 was found downregulated at 3 and 12 h of PMI, whereas it remained unchanged at 6 h and 24 h of PMI. Using a multivariate analysis, it was possible to predict the FC of miR-381-3p of all but 6 h-PMI analyzed PMIs. Discussion The present results suggest that miR-23b-3p and miR-381-3p participate at the early PMI, probably regulating the expression of some genes related to the autolysis process as EPC1 gene. Although the miR-381-3p gene expression is a potential estimator of PMI, further studies will be required to obtain better estimates.
Collapse
Affiliation(s)
- Vanessa Martínez-Rivera
- Laboratorio de Genética, Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Christian A Cárdenas-Monroy
- Laboratorio de Genética, Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Oliver Millan-Catalan
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituo Nacional de Cancerologia, Ciudad de México, México.,Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Jessica González-Corona
- Laboratorio de Genética, Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - N Sofia Huerta-Pacheco
- Cátedras CONACYT-Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Antonio Martínez-Gutiérrez
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituo Nacional de Cancerologia, Ciudad de México, México
| | - Alexa Villavicencio-Queijeiro
- Laboratorio de Genética, Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Carlos Pedraza-Lara
- Laboratorio de Entomología, Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Nacional de Medicina Genomica, Ciudad de México, México
| | - María Elena Bravo-Gómez
- Laboratorio de Toxicología, Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Carlos Pérez-Plasencia
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituo Nacional de Cancerologia, Ciudad de México, México.,Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mariano Guardado-Estrada
- Laboratorio de Genética, Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
14
|
Safi A, Bastami M, Delghir S, Ilkhani K, Seif F, Alivand MR. miRNAs Modulate the Dichotomy of Cisplatin Resistance or Sensitivity in Breast Cancer: An Update of Therapeutic Implications. Anticancer Agents Med Chem 2021; 21:1069-1081. [PMID: 32885760 DOI: 10.2174/1871520620666200903145939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/02/2020] [Accepted: 08/08/2020] [Indexed: 11/22/2022]
Abstract
Cisplatin has a broad-spectrum antitumor activity and is widely used for the treatment of various malignant tumors. However, acquired or intrinsic resistance of cisplatin is a major problem for patients during the therapy. Recently, it has been reported Cancer Stem Cell (CSC)-derived drug resistance is a great challenge of tumor development and recurrence; therefore, the sensitivity of Breast Cancer Stem Cells (BCSCs) to cisplatin is of particular importance. Increasing evidence has shown that there is a relationship between cisplatin resistance/sensitivity genes and related miRNAs. It is known that dysregulation of relevant miRNAs plays a critical role in regulating target genes of cisplatin resistance/sensitivity in various pathways such as cellular uptake/efflux, Epithelial-Mesenchymal Transition (EMT), hypoxia, and apoptosis. Furthermore, the efficacy of the current chemotherapeutic drugs, including cisplatin, for providing personalized medicine, can be improved by controlling the expression of miRNAs. Thus, potential targeting of miRNAs can lead to miRNA-based therapies, which will help overcome drug resistance and develop more effective personalized anti-cancer and cotreatment strategies in breast cancer. In this review, we summarized the general understandings of miRNAregulated biological processes in breast cancer, particularly focused on the role of miRNA in cisplatin resistance/ sensitivity.
Collapse
Affiliation(s)
- Asma Safi
- Clinical Research Development Unit, Shohada Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Bastami
- Clinical Research Development Unit, Shohada Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Delghir
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khandan Ilkhani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Seif
- Department of Immunology & Allergy, Academic Center for Education, Culture, and Research, Tehran, Iran
| | - Mohammad R Alivand
- Clinical Research Development Unit, Shohada Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Liu Y, Huang R, Xie D, Lin X, Zheng L. ZNF674-AS1 antagonizes miR-423-3p to induce G0/G1 cell cycle arrest in non-small cell lung cancer cells. Cell Mol Biol Lett 2021; 26:6. [PMID: 33618674 PMCID: PMC7901084 DOI: 10.1186/s11658-021-00247-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND ZNF674-AS1, a recently characterized long noncoding RNA, shows prognostic significance in hepatocellular carcinoma and glioma. However, the expression and function of ZNF674-AS1 in non-small cell lung cancer (NSCLC) are unclear. METHODS In this work, we investigated the expression of ZNF674-AS1 in 83 pairs of NSCLC specimens and adjacent noncancerous lung tissues. The clinical significance of ZNF674-AS1 in NSCLC was analyzed. The role of ZNF674-AS1 in NSCLC growth and cell cycle progression was explored. RESULTS Our data show that ZNF674-AS1 expression is decreased in NSCLC compared to normal tissues. ZNF674-AS1 downregulation is significantly correlated with advanced TNM stage and decreased overall survival of NSCLC patients. Overexpression of ZNF674-AS1 inhibits NSCLC cell proliferation, colony formation, and tumorigenesis, which is accompanied by a G0/G1 cell cycle arrest. Conversely, knockdown of ZNF674-AS1 enhances the proliferation and colony formation of NSCLC cells. Biochemically, ZNF674-AS1 overexpression increases the expression of p21 through downregulation of miR-423-3p. Knockdown of p21 or overexpression of miR-423-3p blocks ZNF674-AS1-mediated growth suppression and G0/G1 cell cycle arrest. In addition, ZNF674-AS1 expression is negatively correlated with miR-423-3p in NSCLC specimens. CONCLUSIONS ZNF674-AS1 suppresses NSCLC growth by downregulating miR-423-3p and inducing p21. This work suggests the therapeutic potential of ZNF674-AS1 in the treatment of NSCLC.
Collapse
Affiliation(s)
- Yu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Risheng Huang
- Department of Thoracic Surgery, Wenzhou Central Hospital, Wenzhou, China.
| | - Deyao Xie
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoming Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liangcheng Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Moghadam ER, Owrang M, Hashemi F, Makvandi P, Goharrizi MASB, Najafi M, Khan H. Lung cancer cells and their sensitivity/resistance to cisplatin chemotherapy: Role of microRNAs and upstream mediators. Cell Signal 2021; 78:109871. [PMID: 33279671 DOI: 10.1016/j.cellsig.2020.109871] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (CP) is a well-known chemotherapeutic agent with excellent clinical effects. The anti-tumor activity of CP has been demonstrated in different cancers such as breast, cervical, reproductive, lung, brain, and prostate cancers. However, resistance of cancer cells to CP chemotherapy has led to its failure in eradication of cancer cells, and subsequent death of patients with cancer. Fortunately, much effort has been put to identify molecular pathways and mechanisms involved in CP resistance/sensitivity. It seems that microRNAs (miRs) are promising candidates in mediating CP resistance/sensitivity, since they participate in different biological aspects of cells such as proliferation, migration, angiogenesis, and differentiation. In this review, we focus on miRs and their regulation in CP chemotherapy of lung cancer, as the most malignant tumor worldwide. Oncogenic miRs trigger CP resistance in lung cancer cells via targeting various pathways such as Wnt/β-catenin, Rab6, CASP2, PTEN, and Apaf-1. In contrast, onco-suppressor miRs inhibit oncogene pathways such as STAT3 to suppress CP resistance. These topics are discussed to determine the role of miRs in CP resistance/sensitivity. We also describe the upstream modulators of miRs such as lncRNAs, circRNAs, NF-κB, SOX2 and TRIM65 and their association with CP resistance/sensitivity in lung cancer cells. Finally, the effect of anti-tumor plant-derived natural compounds on miR expression during CP sensitivity of lung cancer cells is discussed.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Owrang
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fardin Hashemi
- Student Research Committee, Department of Physiotherapy, Faculty of Rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | | | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
17
|
Torsin LI, Petrescu GED, Sabo AA, Chen B, Brehar FM, Dragomir MP, Calin GA. Editing and Chemical Modifications on Non-Coding RNAs in Cancer: A New Tale with Clinical Significance. Int J Mol Sci 2021; 22:ijms22020581. [PMID: 33430133 PMCID: PMC7827606 DOI: 10.3390/ijms22020581] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Currently, for seemingly every type of cancer, dysregulated levels of non-coding RNAs (ncRNAs) are reported and non-coding transcripts are expected to be the next class of diagnostic and therapeutic tools in oncology. Recently, alterations to the ncRNAs transcriptome have emerged as a novel hallmark of cancer. Historically, ncRNAs were characterized mainly as regulators and little attention was paid to the mechanisms that regulate them. The role of modifications, which can control the function of ncRNAs post-transcriptionally, only recently began to emerge. Typically, these modifications can be divided into reversible (i.e., chemical modifications: m5C, hm5C, m6A, m1A, and pseudouridine) and non-reversible (i.e., editing: ADAR dependent, APOBEC dependent and ADAR/APOBEC independent). The first research papers showed that levels of these modifications are altered in cancer and can be part of the tumorigenic process. Hence, the aim of this review paper is to describe the most common regulatory modifications (editing and chemical modifications) of the traditionally considered “non-functional” ncRNAs (i.e., microRNAs, long non-coding RNAs and circular RNAs) in the context of malignant disease. We consider that only by understanding this extra regulatory layer it is possible to translate the knowledge about ncRNAs and their modifications into clinical practice.
Collapse
Affiliation(s)
- Ligia I. Torsin
- Department of Anesthesiology and Critical Care, Elias Clinical Emergency Hospital, 011461 Bucharest, Romania;
| | - George E. D. Petrescu
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (G.E.D.P.); (F.M.B.)
- Department of Neurosurgery, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania
| | - Alexandru A. Sabo
- Zentrum für Kinder, Jugend und Frauenmedizin, Pediatrics 2 (General and Special Pediatrics), Klinikum Stuttgart, Olgahospital, 70174 Stuttgart, Germany;
| | - Baoqing Chen
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China;
- Guangdong Esophageal Cancer Research Institute, Guangzhou 510060, China
| | - Felix M. Brehar
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (G.E.D.P.); (F.M.B.)
- Department of Neurosurgery, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania
| | - Mihnea P. Dragomir
- Institute of Pathology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence: or (M.P.D.); (G.A.C.); Tel.: +40-254-219-493 (M.P.D.); +1-713-792-5461 (G.A.C.)
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Correspondence: or (M.P.D.); (G.A.C.); Tel.: +40-254-219-493 (M.P.D.); +1-713-792-5461 (G.A.C.)
| |
Collapse
|
18
|
Zhang J, Rao D, Ma H, Kong D, Xu X, Lu H. LncRNA SNHG15 contributes to doxorubicin resistance of osteosarcoma cells through targeting the miR-381-3p/GFRA1 axis. Open Life Sci 2020; 15:871-883. [PMID: 33817274 PMCID: PMC7874549 DOI: 10.1515/biol-2020-0086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 07/03/2020] [Accepted: 08/05/2020] [Indexed: 11/21/2022] Open
Abstract
Background Osteosarcoma is a common primary malignant bone cancer. Long noncoding RNA small nucleolar RNA host gene 15 (SNHG15) has been reported to play an oncogenic role in many cancers. Nevertheless, the role of SNHG15 in the doxorubicin (DXR) resistance of osteosarcoma cells has not been fully addressed. Methods Cell Counting Kit-8 assay was conducted to measure the half-maximal inhibitory concentration value of DXR in osteosarcoma cells. Western blotting was carried out to examine the levels of autophagy-related proteins and GDNF family receptor alpha-1 (GFRA1). Quantitative reverse transcription-polymerase chain reaction was performed to determine the levels of SNHG15, miR-381-3p, and GFRA1. The proliferation of osteosarcoma cells was measured by MTT assay. The binding sites between miR-381-3p and SNHG15 or GFRA1 were predicted by Starbase bioinformatics software, and the interaction was confirmed by dual-luciferase reporter assay. Murine xenograft model was established to validate the function of SNHG15 in vivo. Results Autophagy inhibitor 3-methyladenine sensitized DXR-resistant osteosarcoma cell lines to DXR. SNHG15 was upregulated in DXR-resistant osteosarcoma tissues and cell lines. SNHG15 knockdown inhibited the proliferation, DXR resistance, and autophagy of osteosarcoma cells. MiR-381-3p was a direct target of SNHG15, and GFRA1 bound to miR-381-3p in osteosarcoma cells. SNHG15 contributed to DXR resistance through the miR-381-3p/GFRA1 axis in vitro. SNHG15 depletion contributed to the inhibitory effect of DXR on osteosarcoma tumor growth through the miR-381-3p/GFRA1 axis in vivo. Conclusions SNHG15 enhanced the DXR resistance of osteosarcoma cells through elevating the autophagy via targeting the miR-381-3p/GFRA1 axis. Restoration of miR-381-3p expression might be an underlying therapeutic strategy to overcome the DXR resistance of osteosarcoma.
Collapse
Affiliation(s)
- Jinshan Zhang
- School of Medicine, Yangtze University, 434020, Jingzhou, China
| | - Dan Rao
- Central Hospital of Edong Medical Group, Huangshi City, Hubei Province, 435000, Huangshi, China
| | - Haibo Ma
- School of Medicine, Yangtze University, 434020, Jingzhou, China
| | - Defeng Kong
- School of Medicine, Yangtze University, 434020, Jingzhou, China
| | - Xiaoming Xu
- The Second Clinical Medical College, Yangtze University, No. 1 Renmin Road, Jingzhou City, Hubei Province, 434020, Jingzhou, China
| | - Hougen Lu
- The Second Clinical Medical College, Yangtze University, No. 1 Renmin Road, Jingzhou City, Hubei Province, 434020, Jingzhou, China
| |
Collapse
|
19
|
Zeng X, Cao Z, Luo W, Zheng L, Zhang T. MicroRNA-381-A Key Transcriptional Regulator: Its Biological Function and Clinical Application Prospects in Cancer. Front Oncol 2020; 10:535665. [PMID: 33324542 PMCID: PMC7726430 DOI: 10.3389/fonc.2020.535665] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that function by regulating messenger RNAs. Recent studies have shown that miRNAs play important roles in multiple processes of cancer development. MiR-381 is one of the most important miRNAs in cancer progression. MiR-381 is downregulated in some cancers and upregulated in other cancers, including glioma, epithelial sarcoma, and osteosarcoma. MiR-381 regulates epithelial-mesenchymal transition (EMT), chemotherapeutic resistance, radioresistance, and immune responses. Thus, miR-381 participates in tumor initiation, progression, and metastasis. Moreover, miR-381 functions in various oncogenic pathways, including the Wnt/β-catenin, AKT, and p53 pathways. Clinical studies have shown that miR-381 could be considered a biomarker or a novel prognostic factor. Here, we summarize the present studies on the role of miR-381 in cancer development, including its biogenesis and various affected signaling pathways, and its clinical application prospects. MiR-381 expression is associated with tumor stage and survival time, making miR-381 a novel prognostic factor.
Collapse
Affiliation(s)
- Xue Zeng
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
20
|
Lampis A, Hahne JC, Hedayat S, Valeri N. MicroRNAs as mediators of drug resistance mechanisms. Curr Opin Pharmacol 2020; 54:44-50. [PMID: 32898724 DOI: 10.1016/j.coph.2020.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/24/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023]
Abstract
MicroRNAs are small RNA transcripts involved in fine-tuning of several cellular mechanisms and pathways crucial for maintaining cells' homeostasis like apoptosis, differentiation, inflammation and cell-cycle regulation. They act by regulation of gene expression at post-transcriptional level through fine-tuning of target proteins expression. Expression of microRNAs is cell-type specific and since their discovery they have been proven to be deregulated in various disorders including cancer. Several lines of evidence are emerging that link microRNAs to drug resistance mechanisms in tumours given their important role in modulating oncogenic and tumour suppressive mechanisms. This review will focus on latest knowledge of the roles and mechanisms of microRNAs as mediators to drug resistance and the implications for future therapies.
Collapse
Affiliation(s)
- Andrea Lampis
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| | - Jens C Hahne
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Somaieh Hedayat
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Nicola Valeri
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK; Department of Medicine, The Royal Marsden Hospital, London and Sutton, UK
| |
Collapse
|
21
|
Wang S, Li MY, Liu Y, Vlantis AC, Chan JY, Xue L, Hu BG, Yang S, Chen MX, Zhou S, Guo W, Zeng X, Qiu S, van Hasselt CA, Tong MC, Chen GG. The role of microRNA in cisplatin resistance or sensitivity. Expert Opin Ther Targets 2020; 24:885-897. [PMID: 32559147 DOI: 10.1080/14728222.2020.1785431] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Cisplatin is a chemotherapy drug that has been used to treat a number of cancers for decades, and is still one of the most commonly used anti-cancer agents. However, some patients do not respond to cisplatin while other patients who were originally sensitive to cisplatin eventually develop chemoresistance, leading to treatment failure or/and tumor recurrence. AREAS COVERED Different mechanisms contribute to cisplatin resistance or sensitivity, involving multiple pathways or/and processes such as DNA repair, DNA damage response, drug transport, and apoptosis. Among the various mechanisms, it appears that microRNAs play an important role in determining the resistance or sensitivity. In this article, we analyzed and summarized recent findings in this area, with the aim that these data can aid further research and understanding, leading to the eventual reduction of cisplatin resistance. EXPERT COMMENTARY microRNAs can positively or negatively regulate cisplatin resistance by acting on molecules or/and pathways related to apoptosis, autophagy, hypoxia, cancer stem cells, NF-κB, and Notch1. It appears that the modulation of relevant microRNAs can effectively re-sensitize cancer cells to cisplatin regimen in certain types of cancers including breast, colorectal, gastric, liver, lung, ovarian, prostate, testicular, and thyroid cancers.
Collapse
Affiliation(s)
- Shanshan Wang
- School of Life Sciences and Biopharmaceutics, Guangdong, Pharmaceutical University , Guangzhou, China.,Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China
| | - Ming-Yue Li
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong, China
| | - Yi Liu
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong, China
| | - Alexander C Vlantis
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - Jason Yk Chan
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - Lingbin Xue
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China
| | - Bao-Guang Hu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Binzhou Medical University , Binzhou, Shenzhen, China
| | - Shucai Yang
- Department of Clinical Laboratory, Pingshan District People's Hospital of Shenzhen , Shenzhen, Guangdong, China
| | - Mo-Xian Chen
- Division of Gastroenterology, Shenzhen Children's Hospital , Shenzhen, China
| | - Shaoming Zhou
- Division of Gastroenterology, Shenzhen Children's Hospital , Shenzhen, China
| | - Wei Guo
- Shenzhen Ritzcon Biological Technology Co., LTD , Shenzhen, Guangdong, China
| | - Xianhai Zeng
- DShenzhen Key Laboratory of ENT, Institute of ENT & Longgang ENT Hospital , Shandong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - Shuqi Qiu
- DShenzhen Key Laboratory of ENT, Institute of ENT & Longgang ENT Hospital , Shandong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - C Andrew van Hasselt
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - Michael Cf Tong
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - George G Chen
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| |
Collapse
|
22
|
Prates J, Moreli JB, Gimenes AD, Biselli JM, Pires D'Avila SCG, Sandri S, Farsky SHP, Rodrigues-Lisoni FC, Oliani SM. Cisplatin treatment modulates Annexin A1 and inhibitor of differentiation to DNA 1 expression in cervical cancer cells. Biomed Pharmacother 2020; 129:110331. [PMID: 32768930 DOI: 10.1016/j.biopha.2020.110331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/19/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023] Open
Abstract
Cisplatin (Cis) is a choice chemotherapy approach to cervical cancer by inducing DNA adducts and subsequent apoptosis. We have investigated the effects of Cis on Annexin A1 (ANXA1) and inhibitor of DNA binding 1 (ID1) proteins expression to elucidate further mechanisms of Cis actions. Human cervical tissue samples from twenty-four patients, with Cervical Intraepithelial Neoplasia (CIN, stage I, II and III), were evaluated to quantified ANXA1 and ID1 expressions. In vitro, human epidermoid carcinoma of the cervix (SiHa cell line) were treated with Annexin A1 peptide (ANXA12-26), Cis or Cis + ANXA12-26 to evaluate cell proliferation and migration, cytotoxicity of treatments as well as ANXA1 and ID1 modulations by mRNA and protein expression. Our findings showed expression of ID1 and ANXA1 proteins in tissue samples from Cervical Intraepithelial Neoplasia (CIN) patients, with intense immunological identification of ID1 in the CIN III stage. In SiHa cells, treatments with Cis alone or Cis + ANXA12-26, increase mRNA expressions of the ANXA1 and reduced the ID1. In agreement, Cis + ANXA12-26 enhanced ANXA1 protein expression and Cis or Cis + ANXA12-26 abolished ID1 protein expression. Cell proliferation was reduced after treatment with ANXA12-26 peptide and more significant after Cis or Cis + ANXA12-26 treatments. These two last treatments reduced cell viability, by inducing late apoptosis, and impaired cell migration. Together, our data highlight endogenous ANXA1 is involved in Cis therapy for cervical cancer.
Collapse
Affiliation(s)
- Janesly Prates
- São Paulo State University (Unesp), Institute of Biosciences, Humanities and Exact Sciences (Ibilce), Campus São José do Rio Preto, SP, Brazil
| | - Jusciéle Brogin Moreli
- Universidade Federal de São Paulo - UNIFESP, Post-Graduation in Structural and Functional Biology, SP, Brazil; Faceres School of Medicine, São José do Rio Preto, SP, Brazil
| | - Alexandre Dantas Gimenes
- Universidade Federal de São Paulo - UNIFESP, Post-Graduation in Structural and Functional Biology, SP, Brazil
| | - Joice Matos Biselli
- São Paulo State University (Unesp), Institute of Biosciences, Humanities and Exact Sciences (Ibilce), Campus São José do Rio Preto, SP, Brazil
| | | | - Silvana Sandri
- São Paulo University (USP), Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, São Paulo, Brazil
| | - Sandra Helena Poliselli Farsky
- São Paulo University (USP), Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, São Paulo, Brazil
| | - Flávia Cristina Rodrigues-Lisoni
- São Paulo State University (Unesp), Institute of Biosciences, Humanities and Exact Sciences (Ibilce), Campus São José do Rio Preto, SP, Brazil; São Paulo State University (Unesp), Ilha Solteira School of Engineering (FEIS), Campus Ilha Solteira, SP, Brazil
| | - Sonia Maria Oliani
- São Paulo State University (Unesp), Institute of Biosciences, Humanities and Exact Sciences (Ibilce), Campus São José do Rio Preto, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Post-Graduation in Structural and Functional Biology, SP, Brazil.
| |
Collapse
|
23
|
Ma HF, Lv GX, Zhang DH. miR-381 Mediates the Development of Head and Neck Squamous Cell Carcinoma via Targeting STC2. Onco Targets Ther 2020; 13:4485-4493. [PMID: 32547079 PMCID: PMC7247612 DOI: 10.2147/ott.s246289] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Objective miR-381 is implicated in the occurrence and development of various cancers, yet its role in head and neck squamous cell carcinoma (HNSCC) remains largely unknown. This study sought to research the direct target of miR-381 in HNSCC and investigate their roles in cancer progression. Methods miRNA and mRNA expression files of HNSCC were accessed from TCGA database and then processed for differential analysis. Bioinformatics databases were employed to predict the target mRNAs of the potential miRNA. qRT-PCR was conducted to determine the expression levels of the target miRNA and mRNA. Then, a series of in vitro experiments like CCK-8, colony formation assay, wound healing assay and transwell assay were performed to detect cell proliferation, migration and invasion. Dual-luciferase reporter gene assay was carried out for the further validation of the targeted relationship between the miRNA and mRNA. Results miR-381 was observed to be greatly down-regulated in HNSCC cells, and its overexpression could inhibit cell proliferation, migration and invasion. Besides, dual-luciferase reporter gene assay confirmed that STC2 was a direct target of miR-381, and their expression levels were reversely correlated. Moreover, rescue experiments demonstrated that overexpressing STC2 could rescue the inhibitory effect of miR-381 overexpression on cell proliferation, migration and invasion. Also, we verified that miR-381/STC2 exerted its function on HNSCC proliferation by mediating the FAK/PI3K/Akt/mTOR signaling pathway. Conclusion miR-381 suppresses cell proliferation, migration and invasion in HNSCC through targeting STC2, and participates in HNSCC development probably via the FAK/PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hai-Feng Ma
- Department of Radiotherapy, Zhejiang Dongyang People's Hospital, Dongyang 322100, Zhejiang Province, People's Republic of China
| | - Guo-Xiao Lv
- Department of Radiotherapy, Zhejiang Dongyang People's Hospital, Dongyang 322100, Zhejiang Province, People's Republic of China
| | - Da-Hai Zhang
- Department of Radiotherapy, Zhejiang Dongyang People's Hospital, Dongyang 322100, Zhejiang Province, People's Republic of China
| |
Collapse
|
24
|
Non-Coding RNAs in Lung Tumor Initiation and Progression. Int J Mol Sci 2020; 21:ijms21082774. [PMID: 32316322 PMCID: PMC7215285 DOI: 10.3390/ijms21082774] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is one of the deadliest forms of cancer affecting society today. Non-coding RNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), through the transcriptional, post-transcriptional, and epigenetic changes they impose, have been found to be dysregulated to affect lung cancer tumorigenesis and metastasis. This review will briefly summarize hallmarks involved in lung cancer initiation and progression. For initiation, these hallmarks include tumor initiating cells, immortalization, activation of oncogenes and inactivation of tumor suppressors. Hallmarks involved in lung cancer progression include metastasis and drug tolerance and resistance. The targeting of these hallmarks with non-coding RNAs can affect vital metabolic and cell signaling pathways, which as a result can potentially have a role in cancerous and pathological processes. By further understanding non-coding RNAs, researchers can work towards diagnoses and treatments to improve early detection and clinical response.
Collapse
|
25
|
Mi H, Wang X, Wang F, Li L, Zhu M, Wang N, Xiong Y, Gu Y. SNHG15 Contributes To Cisplatin Resistance In Breast Cancer Through Sponging miR-381. Onco Targets Ther 2020; 13:657-666. [PMID: 32021307 PMCID: PMC6983676 DOI: 10.2147/ott.s223321] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022] Open
Abstract
Background Increasing evidence implies the participation of long non-coding RNAs (lncRNAs) in chemoresistance to cancer treatment. Their role and molecular mechanisms in breast cancer chemoresistance, nevertheless, are yet not considerably elucidated. In this work, we research the function of small nucleolar RNA host gene 15 (SNHG15) in cisplatin (DDP) resistance of breast cancer and uncover the underlying molecular mechanism. Methods SNHG15 and miR-381 expression levels were detected using Quantitative real-time PCR (qRT-PCR) analysis. The functional roles of SNHG15 and miR-381 in breast cancer were determined using MTT assay and flow cytometry analysis. The effect of SNHG15 on miR-381 expression was determined using Luciferase reporter assay, RNA immunoprecipitation (RIP) assay and qRT-PCR analysis. Results SNHG15 was found to be up-regulated in cisplatin resistant breast cancer tissues and cell lines. Breast cancer patients with high SNHG15 expression had a poor prognosis. SNHG15 silencing enhanced cisplatin sensitivity of MCF-7/DDP and MDA-MB-231/DDP cells. Additionally, SNHG15 could function as a miR-381 sponge. miR-381 overexpression could overcome cisplatin resistance. miR-381 knockdown countered SNHG15 knockdown-mediated enhancement of cisplatin sensitivity in MCF-7/DDP and MDA-MB-231/DDP cells. Besides, SNHG15 knockdown facilitated cisplatin sensitivity of cisplatin resistant breast cancer cells in vivo. Conclusion In summary, SNHG15 knockdown overcame cisplatin resistance of breast cancer by sponging miR-381, providing a novel therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Hailong Mi
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 475000, People's Republic of China
| | - Xiaochun Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 475000, People's Republic of China
| | - Fang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 475000, People's Republic of China
| | - Lin Li
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 475000, People's Republic of China
| | - Mingzhi Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 475000, People's Republic of China
| | - Nan Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 475000, People's Republic of China
| | - Youyi Xiong
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 475000, People's Republic of China
| | - Yuanting Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 475000, People's Republic of China
| |
Collapse
|
26
|
Jin D, Guo J, Wu Y, Chen W, Du J, Yang L, Wang X, Gong K, Dai J, Miao S, Li X, Su G. Metformin-repressed miR-381-YAP-snail axis activity disrupts NSCLC growth and metastasis. J Exp Clin Cancer Res 2020; 39:6. [PMID: 31906986 PMCID: PMC6945774 DOI: 10.1186/s13046-019-1503-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/12/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Recent evidence indicates that metformin inhibits mammalian cancer growth and metastasis through the regulation of microRNAs. Metformin regulates miR-381 stability, which plays a vital role in tumor progression. Moreover, increased YAP expression and activity induce non-small cell lung cancer (NSCLC) tumor growth and metastasis. However, the molecular mechanism underpinning how metformin-induced upregulation of miR-381 directly targets YAP or its interactions with the epithelial-mesenchymal transition (EMT) marker protein Snail in NSCLC is still unknown. METHODS Levels of RNA and protein were analyzed using qPCR, western blotting and immunofluorescence staining. Cellular proliferation was detected using a CCK8 assay. Cell migration and invasion were analyzed using wound healing and transwell assays. Promoter activity and transcription were investigated using the luciferase reporter assay. Chromatin immunoprecipitation was used to detect the binding of YAP to the promoter of Snail. The interaction between miR-381 and the 3'UTR of YAP mRNA was analyzed using the MS2 expression system and co-immunoprecipitation with biotin. RESULTS We observed that miR-381 expression is negatively correlated with YAP expression and plays an opposite role to YAP in the regulation of cellular proliferation, invasion, migration, and EMT of NSCLC cells. The miR-381 function as a tumor suppressor was significantly downregulated in lung cancer tissue specimens and cell lines, which decreased the expression of its direct target YAP. In addition, metformin decreased cell growth, migration, invasion, and EMT via up-regulation of miR-381. Moreover, YAP, which functions as a co-transcription factor, enhanced NSCLC progression and metastasis by upregulation of Snail. Snail knockdown downregulated the mesenchymal marker vimentin and upregulated the epithelial marker E-cadherin in lung cancer cells. Furthermore, miR-381, YAP, and Snail constitute the miR-381-YAP-Snail signal axis, which is repressed by metformin, and enhances cancer cell invasiveness by directly regulating EMT. CONCLUSIONS Metformin-induced repression of miR-381-YAP-Snail axis activity disrupts NSCLC growth and metastasis. Thus, we believe that the miR-381-YAP-Snail signal axis may be a suitable diagnostic marker and a potential therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Dan Jin
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Jiwei Guo
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China.
| | - Yan Wu
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Weiwei Chen
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Jing Du
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Lijuan Yang
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Xiaohong Wang
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Kaikai Gong
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Juanjuan Dai
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Shuang Miao
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Xuelin Li
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Guoming Su
- Department of Nursing, Binzhou Polytechnic University, Binzhou, 256603, People's Republic of China
| |
Collapse
|
27
|
Zhao Z, Bo Z, Gong W, Guo Y. Inhibitor of Differentiation 1 (Id1) in Cancer and Cancer Therapy. Int J Med Sci 2020; 17:995-1005. [PMID: 32410828 PMCID: PMC7211148 DOI: 10.7150/ijms.42805] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The inhibitor of DNA binding (Id) proteins are regulators of cell cycle and cell differentiation. Of all Id family proteins, Id1 is mostly linked to tumorigenesis, cellular senescence as well as cell proliferation and survival. Id1 is a stem cell-like gene more than a classical oncogene. Id1 is overexpressed in numerous types of cancers and exerts its promotion effect to these tumors through different pathways. Briefly, Id1 was found significantly correlated with EMT-related proteins, K-Ras signaling, EGFR signaling, BMP signaling, PI3K/Akt signaling, WNT and SHH signaling, c-Myc signaling, STAT3 signaling, RK1/2 MAPK/Egr1 pathway and TGF-β pathway, etc. Id1 has potent effect on facilitating tumorous angiogenesis and metastasis. Moreover, high expression of Id1 plays a facilitating role in the development of drug resistance, including chemoresistance, radiation resistance and resistance to drugs targeting angiogenesis. However, controversial results were also obtained. Overall, Id1 represent a promising target of anti-tumor therapeutics based on its potent promotion effect to cancer. Numerous drugs were found exerting their anti-tumor function through Id1-related signaling pathways, such as fucoidan, berberine, tetramethylpyrazine, crizotinib, cannabidiol and vinblastine.
Collapse
Affiliation(s)
- Zhengxiao Zhao
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Zhiyuan Bo
- The Second Department of Biliary Tract Surgery, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China
| | - Weiyi Gong
- The Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai 200040, PR China
| | - Yong Guo
- Department of Oncology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
28
|
Dou D, Ge X, Wang X, Xu X, Zhang Z, Seng J, Cao Z, Gu Y, Han M. EZH2 Contributes To Cisplatin Resistance In Breast Cancer By Epigenetically Suppressing miR-381 Expression. Onco Targets Ther 2019; 12:9627-9637. [PMID: 32009798 PMCID: PMC6859472 DOI: 10.2147/ott.s214104] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Emerging evidence reveals the vital role of enhancer of zeste homolog 2 (EZH2) in cancer chemoresistance. However, its function and molecular mechanisms in breast cancer chemoresistance remain largely unknown. METHODS Gene expression was evaluated using quantitative real-time PCR (qRT-PCR) and Western blot analysis. The functional roles of EZH2 and miR-381 in breast cancer were explored using cell MTT assay and flow cytometry analysis. The effect of EZH2 on miR-381 expression in transcriptional level was determined using Chromatin immunoprecipitation (ChIP) assay and Luciferase reporter assay. RESULTS In this study, we found that EZH2 was up-regulated in CDDP-resistant breast cancer tissues and cell lines. Breast cancer patients with high EZH2 expression had a poor prognosis. EZH2 silencing improved the sensitivity of MCF-7/CDDP and MDA-MB-231/CDDP cells towards CDDP. Moreover, EZH2 could epigenetically silence miR-381. miR-381 overexpression could overcome CDDP resistance in CDDP-resistant breast cancer cells. miR-381 knockdown weakened the inductive effect of EZH2 silencing on CDDP sensitivity of MCF-7/CDDP and MDA-MB-231/CDDP cells. Furthermore, EZH2 knockdown facilitated CDDP sensitivity of CDDP-resistant breast cancer cells in vivo. CONCLUSIONS Collectively, EZH2 depletion overcame CDDP resistance of breast cancer through epigenetically silencing miR-381, providing a novel therapeutic target for breast cancer chemoresistance.
Collapse
Affiliation(s)
- Dongwei Dou
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| | - Xin Ge
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| | - Xinxing Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| | - Xiaodong Xu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| | - Zhe Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| | - Jingjing Seng
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| | - Zhang Cao
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| | - Yuanting Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| | - Mingli Han
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou475000, People’s Republic of China
| |
Collapse
|
29
|
miR‑381 functions as a tumor suppressor by targeting ETS1 in pancreatic cancer. Int J Mol Med 2019; 44:593-607. [PMID: 31173154 PMCID: PMC6605709 DOI: 10.3892/ijmm.2019.4206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/15/2019] [Indexed: 12/19/2022] Open
Abstract
Increasing evidence shows that microRNA (miR)-381 is involved in the carcinogenesis and biologic progression of various types of cancer in humans. However, its potential biologic role and mechanism in pancreatic cancer remain to be elucidated. In the present study, the expression and functional role of miR-381 in pancreatic cancer were investigated. It was found that miR-381 was significantly downregulated in pancreatic cancer tissues and cell lines. The biological functions of miR-381 were examined by measuring cell proliferation, migration, invasion and apoptosis in vitro and in vivo. The miR-381 target gene and signaling pathway were identified by luciferase activity assay and western blot assay. In vitro experiments confirmed that the enforced expression of miR-381 markedly suppressed cell proliferation, migration and invasion, and induced apoptosis in pancreatic cancer cells. By contrast, silencing the expression of miR-381 had the opposite effect. In addition, miR-381 inhibited xenograft tumor growth in vivo. Furthermore, ETS1 was identified as a direct target of miR-381, and western blot analysis showed that miR-381 negatively modulated the expression of ETS1. It was also demonstrated that miR-381 serves a key role in pancreatic cancer cells through regulating the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway. In conclusion, the data obtained suggested that miR-381 mediated cell proliferation, migration and invasion by targeting ETS1, partly through PI3K/AKT/mTOR signaling pathway. These results provide novel insights into understanding the potential effects and molecular mechanism of miR-381 on pancreatic cancer. miR-381 may serve as a novel potential marker for pancreatic cancer treatment in the future.
Collapse
|
30
|
Yi D, Xu L, Wang R, Lu X, Sang J. miR-381 overcomes cisplatin resistance in breast cancer by targeting MDR1. Cell Biol Int 2019; 43:12-21. [PMID: 30444043 DOI: 10.1002/cbin.11071] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/11/2018] [Indexed: 11/08/2022]
Abstract
Increasing evidence suggests the involvement of microRNA-381 (miR-381) in chemoresistance of cancer treatment. However, its function and molecular mechanisms in breast cancer chemoresistance are still not well elucidated. In the present study, we aimed to investigate the functional role of miR-381 in cisplatin (DDP) resistance of breast cancer and discover the underlying molecular mechanism. The expression levels of miR-381 and MDR1 were detected by quantitative real-time PCR (qRT-PCR) and Western blot analysis in breast cancer tissues and cell lines. The DDP sensitivity and cell apoptosis of breast cancer cells were determined by MTT assay and flow cytometric analysis, respectively. The relationship between miR-381 and MDR1 was explored by target prediction and luciferase reporter analysis. miR-381 was decreased in DDP-resistant breast cancer tissues and cell lines. Low miR-381 expression was correlated with poor prognosis of breast cancer patients. miR-381 overexpression improved DDP sensitivity of MCF-7/DDP and MDA-MB-231/DDP cells. Conversely, miR-381 inhibition lowered the response of MCF-7 and MDA-MB-231 to DPP. Moreover, miR-381 could directly suppress multidrug resistance 1 (MDR1) expression. MDR1 knockdown could overcome DDP resistance in MCF-7/DDP and MDA-MB-231/DDP cells, while MDR1 overexpression led to DDP resistance in MCF-7 and MDA-MB-231 cells. Notably, MDR1 overexpression counteracted the inductive effect of miR-381 mimics on DDP sensitivity of MCF-7/DDP and MDA-MB-231/DDP cells. On the contrary, miR-381 inhibition-mediated DDP resistance in MCF-7 and MDA-MB-231 cells was reversed by MDR1 knockdown. In summary, miR-381 could overcome DDP resistance of breast cancer by directly targeting MDR1, providing a novel therapeutic target for breast cancer chemoresistance.
Collapse
Affiliation(s)
- Dandan Yi
- Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, 210008, China
| | - Lei Xu
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu Province, 210008, China
| | - Ru Wang
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu Province, 210008, China
| | - Xingyi Lu
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu Province, 210008, China
| | - Jianfeng Sang
- Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, 210008, China
| |
Collapse
|
31
|
Wang X, Shi J, Chen Y, Wang C, Shi H, Xie X. Retracted Article: Long noncoding RNA ANRIL knockdown increases sensitivity of non-small cell lung cancer to cisplatin by regulating the miR-656-3p/SOX4 axis. RSC Adv 2019; 9:38735-38744. [PMID: 35540191 PMCID: PMC9075935 DOI: 10.1039/c9ra06993c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/19/2019] [Indexed: 12/25/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are implicated in the development of chemoresistance in many cancers. However, the effect and mechanism of lncRNA antisense noncoding RNA in the INK4 locus (ANRIL) on cisplatin (CDDP) resistance in non-small cell lung cancer (NSCLC) remain unclear. The levels of ANRIL, microRNA (miR)-656-3p and sex-determining region Y-related high-mobility group box 4 (SOX4) in NSCLC tissues and cells were detected by quantitative real-time polymerase chain reaction or western blotting. Cell viability, apoptosis, migration and epithelial-to-mesenchymal transition (EMT) were assessed by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT assay), flow cytometry, trans-well assays and western blotting, respectively. The xenograft model was established using CDDP-resistant NSCLC cells. The target association between miR-656-3p and ANRIL or SOX4 was validated by luciferase reporter assay and RNA immunoprecipitation. ANRIL expression was increased in CDDP-resistant NSCLC tissues and cells. Knockdown of ANRIL decreased cell viability, migration and EMT but induced apoptosis in CDDP-resistant NSCLC cells. Moreover, silencing of ANRIL reduced xenograft tumor growth in vivo. miR-656-3p was targeted by ANRIL and its exhaustion attenuated the suppressive role of ANRIL knockdown in CDDP resistance in NSCLC cells. SOX4 acted as a target of miR-656-3p and was positively regulated by ANRIL. Collectively, interference of ANRIL repressed CDDP resistance through promoting apoptosis and inhibiting cell viability, migration and EMT by up-regulating miR-656-3p and down-regulating SOX4, indicating a new target to improve the chemotherapeutic efficacy in NSCLC. Long noncoding RNAs (lncRNAs) are implicated in the development of chemoresistance in many cancers.![]()
Collapse
Affiliation(s)
- Xianfang Wang
- Department of Laboratory
- People's Hospital of Rizhao
- Rizhao 276800
- China
| | - Jun Shi
- Department of Laboratory
- People's Hospital of Rizhao
- Rizhao 276800
- China
| | - Ying Chen
- Department of Laboratory
- People's Hospital of Rizhao
- Rizhao 276800
- China
| | - Caihong Wang
- Department of Pharmacy
- Rizhao Maternal and Child Health Care Hospital
- China
| | - Huifang Shi
- Department of Laboratory
- People's Hospital of Rizhao
- Rizhao 276800
- China
| | - Xuefang Xie
- Department of Laboratory
- People's Hospital of Rizhao
- Rizhao 276800
- China
| |
Collapse
|
32
|
Liu C, Tian X, Zhang J, Jiang L. Long Non-coding RNA DLEU1 Promotes Proliferation and Invasion by Interacting With miR-381 and Enhancing HOXA13 Expression in Cervical Cancer. Front Genet 2018; 9:629. [PMID: 30581456 PMCID: PMC6292861 DOI: 10.3389/fgene.2018.00629] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 11/23/2018] [Indexed: 11/30/2022] Open
Abstract
Although growing evidence has demonstrated that the long non-coding RNA DLEU1 is involved in the progression of various cancers, its functional role and underlying mechanisms have not been explored in cervical cancer (CC). In this study, we found that DLEU1 was up-regulated in both CC tissues and CC cell lines, and overexpression of DLEU1 was significantly correlated with shorter patient survival. Knockdown of DLEU1 suppressed CC cell proliferation and invasion, whereas overexpression of DLEU1 promoted the proliferation and invasion of CC cells. Bioinformatics analysis was used to elucidate the potential correlation between DLEU1 and miR-381. Moreover, qRT-PCR analysis, luciferase reporter assay and RNA immunoprecipitation assay confirmed that DLEU1 inhibited the expression of miR-381, and revealed a direct interaction between DLEU1 and miR-381. In addition, we demonstrated that miR-381 directly targeted HOXA13 in CC cells. The restoration of HOXA13 expression reversed DLEU1 knockdown or miR-381 overexpression-mediated suppression of cell proliferation and invasion. These results suggested that DLEU1 can promote CC cell proliferation and invasion via the miR-381/HOXA13 axis.
Collapse
Affiliation(s)
- Chang Liu
- Intensive Care Unit, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xing Tian
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Zhang
- Department Gynecologic Tumor, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Lifeng Jiang
- Department of Chinese and Western Medicine, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
33
|
Mi H, Wang X, Wang F, Li L, Zhu M, Wang N, Xiong Y, Gu Y. miR-381 induces sensitivity of breast cancer cells to doxorubicin by inactivation of MAPK signaling via FYN. Eur J Pharmacol 2018; 839:66-75. [PMID: 30266665 DOI: 10.1016/j.ejphar.2018.09.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/10/2018] [Accepted: 09/25/2018] [Indexed: 12/30/2022]
Abstract
The emergence of drug resistance is still a daunting challenge for the effective therapy of cancer patients. miRNAs have been elucidated as an important regulator in chemoresistance of anti-cancer drugs. miR-381 is found to exert tumor-suppressive effect in breast cancer. However, its role in modulating the sensitivity of doxorubicin (DOX) remains unknown. In this study, we found that miR-381 expression was down-regulated in DOX-resistant breast cancer cells. miR-381 overexpression increased DOX sensitivity and enhanced DOX-induced apoptosis in breast cancer cells. Moreover, miR-381 could directly target FYN to suppress its expression. Additionally, FYN knockdown displayed similar effect on DOX sensitivity as miR-381 up-regulation. Furthermore, FYN overexpression partly reversed miR-381-induced sensitivity to DOX. Finally, enforced expression of miR-381 also improved DOX sensitivity of breast cancer cells in vivo. In summary, miR-381 inactivated MAPK signaling by down-regulating FYN, thereby promoting the chemosensitization of breast cancer cells to DOX. Therefore, miR-381/FYN/MAPK pathway may be applied as a novel target to overcome DOX resistance in breast cancer patients.
Collapse
Affiliation(s)
- Hailong Mi
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xiaochun Wang
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding 071030, China
| | - Fang Wang
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Lin Li
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Mingzhi Zhu
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Nan Wang
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Youyi Xiong
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yuanting Gu
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
34
|
Wu M, Fan B, Guo Q, Li Y, Chen R, Lv N, Diao Y, Luo Y. Knockdown of SETDB1 inhibits breast cancer progression by miR-381-3p-related regulation. Biol Res 2018; 51:39. [PMID: 30309377 PMCID: PMC6180515 DOI: 10.1186/s40659-018-0189-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND SET domain bifurcated 1 (SETDB1) has been widely considered as an oncogene playing a critical role in many human cancers, including breast cancer. Nevertheless, the molecular mechanism by which SETDB1 regulates breast cancer tumorigenesis is still unknown. METHODS qRT-PCR assay or western blot analysis was performed to assess the expression level of SETDB1 mRNA or protein, respectively. siSETDB1, pCMV6-XL5-SETDB1, miR-381-3p mimic, or miR-381-3p inhibitor was transfected into cells to regulate the expression of SETDB1 or miR-381-3p. MiRNA directly interacted with SETDB1 was verified by luciferase reporter assay and RNA immunoprecipitation. CCK-8 assay, colony formation assay, flow cytometric analysis, and transwell assay were used to detect the abilities of cell proliferation, cell cycle progression and migration, respectively. Animal model of xenograft tumor was used to observe the regulatory effect of SETDB1 on tumor growth in vivo. RESULTS We verified that SETDB1 mRNA level was upregulated in breast cancer tissues and cell lines, and SETDB1 depletion led to a suppression of cell proliferation, cell cycle progression and migration in vitro, as well as tumor growth in vivo. SETDB1 was verified to be a target of miR-381-3p. Moreover, miR-381-3p overexpression suppressed cell proliferation, cell cycle progression and migration, whereas SETDB1 abated miR-381-3p-mediated regulatory function on breast cancer cells. CONCLUSIONS This study revealed that SETDB1 knockdown might suppress breast cancer progression at least partly by miR-381-3p-related regulation, providing a novel prospect in breast cancer therapy.
Collapse
Affiliation(s)
- Milu Wu
- grid.459333.bDepartment of Oncology, The Affiliated Hospital of Qinghai University, No. 29, Tongren Rd, Xining, 810001 Qinghai China
| | - Baohua Fan
- grid.459333.bDepartment of Oncology, The Affiliated Hospital of Qinghai University, No. 29, Tongren Rd, Xining, 810001 Qinghai China
| | - Qijing Guo
- grid.459333.bDepartment of Oncology, The Affiliated Hospital of Qinghai University, No. 29, Tongren Rd, Xining, 810001 Qinghai China
| | - Yan Li
- grid.459333.bDepartment of Oncology, The Affiliated Hospital of Qinghai University, No. 29, Tongren Rd, Xining, 810001 Qinghai China
| | - Rong Chen
- grid.459333.bDepartment of Oncology, The Affiliated Hospital of Qinghai University, No. 29, Tongren Rd, Xining, 810001 Qinghai China
| | - Nannan Lv
- grid.459333.bDepartment of Oncology, The Affiliated Hospital of Qinghai University, No. 29, Tongren Rd, Xining, 810001 Qinghai China
| | - Yinzhuo Diao
- grid.459333.bDepartment of Oncology, The Affiliated Hospital of Qinghai University, No. 29, Tongren Rd, Xining, 810001 Qinghai China
| | - Yushuang Luo
- grid.459333.bDepartment of Oncology, The Affiliated Hospital of Qinghai University, No. 29, Tongren Rd, Xining, 810001 Qinghai China
| |
Collapse
|
35
|
Yan T, Zhu S, Zhang J, Lu G, Lv C, Wei Y, Luo M. MicroRNA‑944 targets vascular endothelial growth factor to inhibit cell proliferation and invasion in osteosarcoma. Mol Med Rep 2018; 18:5221-5228. [PMID: 30280196 DOI: 10.3892/mmr.2018.9524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/12/2018] [Indexed: 11/05/2022] Open
Affiliation(s)
- Tingzhen Yan
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Shiyong Zhu
- Department of Spine Surgery, Lanling County People's Hospital of Shandong Province, Linyi, Shandong 277700, P.R. China
| | - Jing Zhang
- Department of Pathology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Gongbiao Lu
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Chaoliang Lv
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Yanchun Wei
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Minghua Luo
- Department of Orthopedics, The Second People's Hospital of Jingdezhen, Jingdezhen, Jiangxi 333000, P.R. China
| |
Collapse
|