1
|
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
|
2
|
Salehi R, Wyse BA, Asare-Werehene M, Esfandiarinezhad F, Abedini A, Pan B, Urata Y, Gutsol A, Vinas JL, Jahangiri S, Xue K, Xue Y, Burns KD, Vanderhyden B, Li J, Osuga Y, Burger D, Tan SL, Librach CL, Tsang BK. Androgen-induced exosomal miR-379-5p release determines granulosa cell fate: cellular mechanism involved in polycystic ovaries. J Ovarian Res 2023; 16:74. [PMID: 37046285 PMCID: PMC10091561 DOI: 10.1186/s13048-023-01141-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is a complex multi-factorial syndrome associated with androgen excess and anovulatory infertility. In the current study, we investigated the role of dihydrotestosterone-induced exosomal miR-379-5p release in determining the destiny of the developing follicles. Our hypothesis was that androgen regulates granulosa cell miR-379-5p content by facilitating its exosomal release in a follicular-stage dependent manner, a process which determines granulosa cell fate. Compared to human non-PCOS subjects, individuals with PCOS exhibit higher follicular fluid free testosterone levels, lower exosomal miR-379-5p content and granulosa cell proliferation. Androgenized rats exhibited lower granulosa cell miR-379-5p but higher phosphoinositide-dependent kinase-1 (PDK1; a miR-379-5p target) content and proliferation. Androgen reduced granulosa cell miR-379-5p content by increasing its exosomal release in preantral follicles, but not in antral follicles in vitro. Studies with an exosomal release inhibitor confirmed that androgen-induced exosomal miR-379-5p release decreased granulosa cell miR-379-5p content and proliferation. Ovarian overexpression of miR-379-5p suppressed granulosa cell proliferation, and basal and androgen-induced preantral follicle growth in vivo. These findings suggest that increased exosomal miR-379-5p release in granulosa cells is a proliferative response to androgenic stimulation specific for the preantral stage of follicle development and that dysregulation of this response at the antral stage is associated with follicular growth arrest, as observed in human PCOS.
Collapse
Affiliation(s)
- Reza Salehi
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Departments of Obstetrics and Gynecology, and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- CReATe Fertility Centre, Toronto, ON, Canada
| | | | - Meshach Asare-Werehene
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Departments of Obstetrics and Gynecology, and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Fereshteh Esfandiarinezhad
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Departments of Obstetrics and Gynecology, and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Atefeh Abedini
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Bo Pan
- Department of Animal BioScience, University of Guelph, Guelph, ON, Canada
| | - Yoko Urata
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Obstetrics and Gynecology, University of Tokyo, Tokyo, Japan
| | - Alex Gutsol
- Division of Nephrology, Department of Medicine, Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
| | - Jose L Vinas
- Division of Nephrology, Department of Medicine, Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
| | | | - Kai Xue
- Department of Gynecology, The Affiliated Obstetrics and Gynecology Hospital, Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Yunping Xue
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Kevin D Burns
- Division of Nephrology, Department of Medicine, Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
| | - Barbara Vanderhyden
- Departments of Obstetrics and Gynecology, and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Julang Li
- Department of Animal BioScience, University of Guelph, Guelph, ON, Canada
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, University of Tokyo, Tokyo, Japan
| | - Dylan Burger
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Nephrology, Department of Medicine, Kidney Research Centre, University of Ottawa, Ottawa, ON, Canada
| | - Seang-Lin Tan
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
- Originelle Fertility Clinic and Women's Health Centre, Ottawa, ON, Canada
| | - Clifford L Librach
- CReATe Fertility Centre, Toronto, ON, Canada
- Departments of Obstetrics and Gynaecology, Physiology, Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Benjamin K Tsang
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- Departments of Obstetrics and Gynecology, and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
3
|
Xu X, Qiu Y, Chen S, Wang S, Yang R, Liu B, Li Y, Deng J, Su Y, Lin Z, Gu J, Li S, Huang L, Zhou Y. Different roles of the insulin-like growth factor (IGF) axis in non-small cell lung cancer. Curr Pharm Des 2022; 28:2052-2064. [DOI: 10.2174/1381612828666220608122934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/29/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Non-small cell lung cancer (NSCLC) remains one of the deadliest malignant diseases, with high incidence and mortality worldwide. The insulin-like growth factor (IGF) axis, consisting of IGF-1, IGF-2, related receptors (IGF-1R, -2R), and high-affinity binding proteins (IGFBP 1–6), is associated with promoting fetal development, tissue growth, and metabolism. Emerging studies have also identified the role of the IGF axis in NSCLC, including cancer growth, invasion, and metastasis. Upregulation of IGE-1 and IGF-2, overexpression of IGF-1R, and dysregulation of downstream signaling molecules involved in the PI-3K/Akt and MAPK pathways jointly increase the risk of cancer growth and migration in NSCLC. At the genetic level, some noncoding RNAs could influence the proliferation and differentiation of tumor cells through the IGF signaling pathway. The resistance to some promising drugs might be partially attributed to the IGF axis. Therapeutic strategies targeting the IGF axis have been evaluated, and some have shown promising efficacy. In this review, we summarize the biological roles of the IGF axis in NSCLC, including the expression and prognostic significance of the related components, noncoding RNA regulation, involvement in drug resistance, and therapeutic application. This review offers comprehensive understanding of NSCLC and provides insightful ideas for future research.
Collapse
Affiliation(s)
- Xiongye Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanli Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Simin Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuaishuai Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ruifu Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Baomo Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yufei Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiating Deng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Su
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziying Lin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jincui Gu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shaoli Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lixia Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanbin Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Bhatt S, Singh P, Sharma A, Rai A, Dohare R, Sankhwar S, Sharma A, Syed MA. Deciphering Key Genes and miRNAs Associated With Hepatocellular Carcinoma via Network-Based Approach. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:843-853. [PMID: 32795971 DOI: 10.1109/tcbb.2020.3016781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hepatocellular carcinoma (HCC)is a common type of liver cancer and has a high mortality world-widely. The diagnosis, prognoses, and therapeutics are very poor due to the unclear molecular mechanism of progression of the disease. To unveil the molecular mechanism of progression of HCC, we extract a large sample of mRNA expression levels from the GEO database where a total of 167 samples were used for study, and out of them, 115 samples were from HCC tumor tissue. This study aims to investigate the module of differentially expressed genes (DEGs)which are co-expressed only in HCC sample data but not in normal tissue samples. Thereafter, we identified the highly significant module of significant co-expressed genes and formed a PPI network for these genes. There were only six genes (namely, MSH3, DMC1, ALPP, IL10, ZNF223, and HSD17B7)obtained after analysis of the PPI network. Out of six only MSH3, DMC1, HSD17B7, and IL10 were found enriched in GO Term & Pathway enrichment analysis and these candidate genes were mainly involved in cellular process, metabolic and catalytic activity, which promote the development & progression of HCC. Lastly, the composite 3-node FFL reveals the driver miRNAs and TFs associated with our key genes.
Collapse
|
5
|
Mesenchymal stem cell (MSC)-derived exosomes as novel vehicles for delivery of miRNAs in cancer therapy. Cancer Gene Ther 2022; 29:1105-1116. [PMID: 35082400 DOI: 10.1038/s41417-022-00427-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/11/2021] [Accepted: 01/14/2022] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are known as promising sources for cancer therapy and can be utilized as vehicles in cancer gene therapy. MSC-derived exosomes are central mediators in the therapeutic functions of MSCs, known as the novel cell-free alternatives to MSC-based cell therapy. MSC-derived exosomes show advantages including higher safety as well as more stability and convenience for storage, transport and administration compared to MSCs transplant therapy. Unmodified MSC-derived exosomes can promote or inhibit tumors while modified MSC-derived exosomes are involved in the suppression of cancer development and progression via the delivery of several therapeutics molecules including chemotherapeutic drugs, miRNAs, anti-miRNAs, specific siRNAs, and suicide gene mRNAs. In most malignancies, dysregulation of miRNAs not only occurs as a consequence of cancer progression but also is directly involved during tumor initiation and development due to their roles as oncogenes (oncomiRs) or tumor suppressors (TS-miRNAs). MiRNA restoration is usually achieved by overexpression of TS-miRNAs using synthetic miRNA mimics and viral vectors or even downregulation of oncomiRs using anti-miRNAs. Similar to other therapeutic molecules, the efficacy of miRNAs restoration in cancer therapy depends on the effectiveness of the delivery system. In the present review, we first provided an overview of the properties and potentials of MSCs in cancer therapy as well as the application of MSC-derived exosomes in cancer therapy. Finally, we specifically focused on harnessing the MSC-derived exosomes for the aim of miRNA delivery in cancer therapy.
Collapse
|
6
|
Cao D, Cao X, Jiang Y, Xu J, Zheng Y, Kang D, Xu C. Circulating exosomal microRNAs as diagnostic and prognostic biomarkers in patients with diffuse large B-cell lymphoma. Hematol Oncol 2021; 40:172-180. [PMID: 34874565 PMCID: PMC9299807 DOI: 10.1002/hon.2956] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/31/2021] [Accepted: 11/30/2021] [Indexed: 02/05/2023]
Abstract
Exosomal microRNAs (miRNAs) are potential biomarkers for a variety of tumors, but have not yet been studied in diffuse large B-cell lymphoma (DLBCL). Here, we investigated the use of exosomal miRNAs in DLBCL diagnosis and prognosis. A total of 256 individuals, including 133 DLBCL patients, 94 healthy controls (HCs), and 29 non-DLBCL concurrent controls (CCs), were enrolled. Exosomal miRNAs were profiled in the screening stage using microarray analysis, and miRNA candidates were confirmed in training, testing, and external testing stages using qRT-PCR. Follow-up information on the DLBCL patients was collected, and miRNAs were used to develop diagnostic and prognostic models for these patients. Five exosomal miRNAs (miR-379-5p, miR-135a-3p, miR-4476, miR-483-3p, and miR-451a) were differentially expressed between DLBCL patients and HCs with areas under the receiver operating characteristic curve (AUC) of 0.86, 0.90, and 0.86 for the training, testing, and external testing stages, respectively. Four exosomal miRNAs (miR-379-5p, miR-135a-3p, miR-4476, and miR-451a) were differentially expressed between patients with DLBCL and CCs, with an AUC of 0.78. One miRNA (miR-451a) was significantly associated with both progression-free survival (PFS) and overall survival (OS) of DLBCL patients, R analysis indicated the combination of miR-451a with international prognostic index was a better predictor of PFS and OS for these patients. Our study suggests that subsets of circulating exosomal miRNAs can be useful noninvasive biomarkers for the diagnosis of DLBCL and that the use of circulating exosomal miRNAs improves the identification of patients with newly diagnosed DLBCL with poor outcomes.
Collapse
Affiliation(s)
- Di Cao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Cao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Jiang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China.,Department of Hematology, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Juan Xu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Deying Kang
- Department of Evidence-Based Medicine and Clinical Epidemiology, Sichuan University, Chengdu, China
| | - Caigang Xu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Ghafouri-Fard S, Shaterabadi D, Abak A, Shoorei H, Bahroudi Z, Taheri M, Mousavinejad SA. An update on the role of miR-379 in human disorders. Biomed Pharmacother 2021; 139:111553. [PMID: 33845370 DOI: 10.1016/j.biopha.2021.111553] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/23/2021] [Accepted: 03/27/2021] [Indexed: 12/24/2022] Open
Abstract
miR-379 is a miRNA transcribed from the MIR379 locus on 14q32.31. This miRNA is located in an evolutionarily conserved miRNA cluster in an imprinted region that contains DLK1 and DIO3 genes. The mouse homolog of this miRNA has been shown to be under-expressed in response to glucocorticoid receptor deficiency. Moreover, miR-379 has a tumor-suppressive role in a wide variety of tissues including the brain, breast, lung, and liver. In addition to restraining cell proliferation and migration, miR-379 can suppress the epithelial-mesenchymal transition process. Abnormal expression of this miRNA implies the pathogenesis of Duchene muscular dystrophy, spinal cord injury, diabetic nephropathy, acute myocardial infarction, and premature ovarian failure. This review aims to the summarization of the role of miR-379 in neoplastic and non-neoplastic conditions.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Donya Shaterabadi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afete Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Bahroudi
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Ali Mousavinejad
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Chen K, Zhang B, Sun Z. MicroRNA 379 Regulates Klotho Deficiency-Induced Cardiomyocyte Apoptosis Via Repression of Smurf1. Hypertension 2021; 78:342-352. [PMID: 34120450 DOI: 10.1161/hypertensionaha.120.16888] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kai Chen
- From the Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., B.Z., Z.S.).,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (K.C., Z.S.)
| | - Bo Zhang
- From the Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., B.Z., Z.S.)
| | - Zhongjie Sun
- From the Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., B.Z., Z.S.).,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (K.C., Z.S.)
| |
Collapse
|
9
|
Liu YR, Wang PY, Xie N, Xie SY. MicroRNAs as Therapeutic Targets for Anticancer Drugs in Lung Cancer Therapy. Anticancer Agents Med Chem 2021; 20:1883-1894. [PMID: 32538735 DOI: 10.2174/1871520620666200615133011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are short, non-coding RNA molecules that regulate gene expression by translational repression or deregulation of messenger RNAs. Accumulating evidence suggests that miRNAs play various roles in the development and progression of lung cancers. Although their precise roles in targeted cancer therapy are currently unclear, miRNAs have been shown to affect the sensitivity of tumors to anticancer drugs. A large number of recent studies have demonstrated that some anticancer drugs exerted antitumor activities by affecting the expression of miRNAs and their targeted genes. These studies have elucidated the specific biological mechanism of drugs in tumor suppression, which provides a new idea or basis for their clinical application. In this review, we summarized the therapeutic mechanisms of drugs in lung cancer therapy through their effects on miRNAs and their targeted genes, which highlights the roles of miRNAs as targets in lung cancer therapy.
Collapse
Affiliation(s)
- Yuan-Rong Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Binzhou Medical University, YanTai, ShanDong, 264003, China
| | - Ping-Yu Wang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Binzhou Medical University, YanTai, ShanDong, 264003, China
| | - Ning Xie
- Department of Chest Surgery, YanTaiShan Hospital, YanTai, 264000, ShanDong, China
| | - Shu-Yang Xie
- Department of Biochemistry and Molecular Biology, Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Binzhou Medical University, YanTai, ShanDong, 264003, China
| |
Collapse
|
10
|
Ghafouri-Fard S, Abak A, Mohaqiq M, Shoorei H, Taheri M. The Interplay Between Non-coding RNAs and Insulin-Like Growth Factor Signaling in the Pathogenesis of Neoplasia. Front Cell Dev Biol 2021; 9:634512. [PMID: 33768092 PMCID: PMC7985092 DOI: 10.3389/fcell.2021.634512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
The insulin-like growth factors (IGFs) are polypeptides with similar sequences with insulin. These factors regulate cell growth, development, maturation, and aging via different processes including the interplay with MAPK, Akt, and PI3K. IGF signaling participates in the pathogenesis of neoplasia, insulin resistance, diabetes mellitus, polycystic ovarian syndrome, cerebral ischemic injury, fatty liver disease, and several other conditions. Recent investigations have demonstrated the interplay between non-coding RNAs and IGF signaling. This interplay has fundamental roles in the development of the mentioned disorders. We designed the current study to search the available data about the role of IGF-associated non-coding RNAs in the evolution of neoplasia and other conditions. As novel therapeutic strategies have been designed for modification of IGF signaling, identification of the impact of non-coding RNAs in this pathway is necessary for the prediction of response to these modalities.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Mohaqiq
- School of Advancement, Centennial College, Ashtonbee Campus, Toronto, ON, Canada
- Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC, United States
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Biranjd University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Wu C, Han R, Yang S, Jiang Y, Shu Z, Liu J, Ji S, Yan W, Liu B. A case–control study of microRNA polymorphisms in gastric cancer screening by SNP chip combined with time of flight mass spectrometry. Biomark Med 2020; 14:1563-1572. [DOI: 10.2217/bmm-2020-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aims: To explore new SNP sites of miRNAs associated with gastric cancer, thereby providing valuable biomarkers to diagnose and screen gastric cancer. Materials & methods: A 1:1 case–control study was carried out. Microarrays were used to screen the SNP loci of miRNAs in the genomes of matched pairs of patients, 96 with gastric cancer and 96 healthy controls. For validation, mass spectrometry was used to classify miRNA SNP loci in 622 pairs of subjects. Results: rs7143252 was linked to a higher occurrence of gastric cancer. Conclusion: These results suggest that rs7143252 could be used as a specific biomarker to diagnose and screen gastric cancer.
Collapse
Affiliation(s)
- Chuancheng Wu
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
- Fujian Provincial Key Laboratory of Environment Factors & Cancer, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
| | - Renjie Han
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
- Fujian Provincial Key Laboratory of Environment Factors & Cancer, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
- Department of Occupational Health, Fuzhou Center for Disease Control & Prevention, Fuzhou, Fujian, 350004, China
| | - Shuangfeng Yang
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
- Fujian Provincial Key Laboratory of Environment Factors & Cancer, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
| | - Yu Jiang
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
- Fujian Provincial Key Laboratory of Environment Factors & Cancer, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
| | - Zhixiong Shu
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
- Fujian Provincial Key Laboratory of Environment Factors & Cancer, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
| | - Jin Liu
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
| | - Shumi Ji
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
- Fujian Provincial Key Laboratory of Environment Factors & Cancer, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
| | - Wei Yan
- Department of Science and Education, Hospital of Xianyou County, No. 910, 825 Main Street, Licheng Town, Putian, 351200, China
| | - Baoying Liu
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
- Fujian Provincial Key Laboratory of Environment Factors & Cancer, 1 Xueyuan Road, Minhou Fuzhou, 350122, China
| |
Collapse
|
12
|
MicroRNAs as regulators of ERK/MAPK pathway: A comprehensive review. Biomed Pharmacother 2020; 132:110853. [PMID: 33068932 DOI: 10.1016/j.biopha.2020.110853] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/19/2020] [Accepted: 10/04/2020] [Indexed: 02/07/2023] Open
Abstract
The ERK/MAPK cascade is one the four distinctive MAPK cascades which transmit extracellular signals to intracellular targets. This cascade has an important role in the regulation of several fundamental processes such as proliferation, differentiation and cell response to diverse extrinsic stresses. Moreover, several studies have shown participation of this cascade in the pathogenesis of cancer. Recent investigations have unraveled interaction between microRNAs (miRNAs) and ERK/MAPK cascade. These transcripts reside in both upstream and downstream of this cascade, regulating or being regulated by ERK/MAPK proteins. In the current review, we summarize the role of miRNAs in the regulation of ERK/MAPK and their contribution in the pathogenesis of human disorders with particular focus on cancers.
Collapse
|
13
|
Liu X, Chu KM. Exosomal miRNAs as circulating biomarkers for prediction of development of haematogenous metastasis after surgery for stage II/III gastric cancer. J Cell Mol Med 2020; 24:6220-6232. [PMID: 32383554 PMCID: PMC7294143 DOI: 10.1111/jcmm.15253] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 12/24/2022] Open
Abstract
Exosomes secreted by living cancer cells can regulate metastasis. Exosomal miRNAs can reflect pathological conditions of the original cancer cells. Therefore, we aim to identify exosomal miRNAs as circulating biomarkers for haematogenous metastasis of gastric cancer. Pre‐treatment serum samples of eighty‐nine patients with stage II/III gastric cancer were collected. Thirty‐four of them developed haematogenous metastasis after surgery and the other fifty‐five did not. Extraction of exosomes was validated by western blot, transmission electron microscopy and nanoparticle tracking analysis. MiRNA qPCR array was performed in three matched pairs of samples. Internal control was selected from PCR array and validated in the remaining samples. Expressions of exosomal miRNAs were evaluated in the remaining samples by RT‐qPCR, as well as in gastric cancer tissue samples and cell culture medium. Expression levels of exosomal miRNAs were analysed with clinical characteristics. The results indicated thirteen up‐regulated and six down‐regulated miRNAs were found after normalization. MiR‐379‐5p and miR‐410‐3p were significantly up‐regulated in metastatic patients (P < .01). Higher expression of exosomal miR‐379‐5p or miR‐410‐3p showed shorter progression‐free survival of the patients (P < .05). It was also found that miR‐379‐5p and miR‐410‐3p were down‐regulated in gastric cancer tissue samples, while they were significantly up‐regulated in gastric cancer cell culture medium compared with cancer cells. In conclusion, exosomal miRNAs are promising circulating biomarkers for prediction of development of haematogenous metastasis after surgery for stage II/III gastric cancer.
Collapse
Affiliation(s)
- Xin Liu
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Kent-Man Chu
- Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong
| |
Collapse
|
14
|
Ye H, Jin Q, Wang X, Li Y. MicroRNA-802 Inhibits Cell Proliferation and Induces Apoptosis in Human Laryngeal Cancer by Targeting cAMP-Regulated Phosphoprotein 19. Cancer Manag Res 2020; 12:419-430. [PMID: 32021454 PMCID: PMC6980851 DOI: 10.2147/cmar.s228429] [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: 08/23/2019] [Accepted: 12/17/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND/AIMS miR-802 plays a key role in cancer progression and development. The purpose of this work is to investigate the functional role of miR-802 in laryngeal cancer and to elucidate the function of miR-802 and cAMP-regulated phosphoprotein 19 (ARPP19) on laryngeal cancer. METHODS RT-qPCR was applied to study the expression level of ARPP19 and miR-802 in the laryngeal carcinoma cell lines and tissues. CCK-8, colony formation, flow cytometry (FACS) assay were used to study the effect of ARPP19 and miR-802 on apoptosis, proliferation, and cell cycle of laryngeal carcinoma cells. Target gene prediction and luciferase reporter gene assay were applied to identify target gene of miR-802. The transcriptional mRNA and protein expression levels of ARPP19 were measured by RT-qPCR or Western blotting. RESULTS miR-802 was down-regulated in laryngeal carcinoma cell lines and tissues. Laryngeal cancer cells transfected by miR-802 mimic were significantly inhibited in the terms of cell colony formation and proliferation. Furthermore, miR-802 can inhibit the expression level of ARPP19 by directly targeting the 3' untranslated region (3'-UTR) of ARPP19. Overexpression of the ARPP19 gene can reverse the suppressive effect of miR-802 on laryngeal cancer cells. CONCLUSION miR-802 can exert tumor suppressor effects in laryngeal carcinoma by targeting ARPP19, indicating that miR-802 protein may play a role of potential therapeutic target for clinical laryngeal cancer.
Collapse
Affiliation(s)
- Huafu Ye
- E.N.T. Department, Taizhou Municipal Hospital, Taizhou City, Zhejiang Province318000, People’s Republic of China
| | - Qiaozhi Jin
- E.N.T. Department, Taizhou Municipal Hospital, Taizhou City, Zhejiang Province318000, People’s Republic of China
| | - Xiaoqiong Wang
- E.N.T. Department, Taizhou Municipal Hospital, Taizhou City, Zhejiang Province318000, People’s Republic of China
| | - Yong Li
- E.N.T. Department, Taizhou Municipal Hospital, Taizhou City, Zhejiang Province318000, People’s Republic of China
| |
Collapse
|
15
|
Gao P, Wang Z, Hu Z, Jiao X, Yao Y. Circular RNA circ_0074027 indicates a poor prognosis for NSCLC patients and modulates cell proliferation, apoptosis, and invasion via miR-185-3p mediated BRD4/MADD activation. J Cell Biochem 2019; 121:2632-2642. [PMID: 31680319 DOI: 10.1002/jcb.29484] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/10/2019] [Indexed: 01/05/2023]
Abstract
Circular RNAs play an imperative role in cancer development and metastasis by regulating oncogenic and tumor-suppressive pathways. However, the role and mechanism of circ_0074027 in non-small-cell lung cancer (NSCLC) have not been elucidated. The expression levels of circ_0074027 were detected by qRT-PCR. The link between circ_0074027 expression and clinicopathologic parameters was analyzed by Fisher's exact test. The prognostic role of circ_0074027 was investigated by Kaplan-Meier and Cox regression analysis. Cell counting kit-8 and flow cytometric assays were utilized to evaluate NSCLC cell proliferation and apoptosis, respectively. Wound scratch and Transwell tests were applied to detect cell migratory and invasive capacities. The interaction potential of circ_0074027 and miR-185-3p was analyzed by the circBank database, and verified by dual-luciferase reporter assay. The downstream gene of miR-185-3p was also investigated. Circ_0074027 was elevated in NSCLC specimens and cell lines. Overexpressed circ_0074027 was related to more advanced TNM stages, poorer differentiation grade, and worse overall survival. Upregulated circ_0074027 increased the proliferation of H1299 cells by inhibiting cell apoptosis. Cell migration and invasion were enhanced after circ_0074027 overexpression. Silenced circ_0074027 caused the opposite effects in the A549 cell line. For mechanism investigation, circ_0074027 directly sponges miR-185-3p to enhance bromodomain-containing protein 4 (BRD4) and MAPK-activating death domain-containing protein (MADD) expression levels at the posttranscriptional level. Furthermore, we found the oncogenic function of circ_0074027 is attributed to its modulation of BRD4 and MADD. Collectively, upregulated circ_0074027 in NSCLC accelerates cell progression via miR-185-3p/BRD4/MADD pathway as a competing endogenous RNA.
Collapse
Affiliation(s)
- Ping Gao
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhidong Wang
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhanliang Hu
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuan Jiao
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Yao
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Ding L, Feng Z, Bai Y. Clustering analysis of microRNA and mRNA expression data from TCGA using maximum edge-weighted matching algorithms. BMC Med Genomics 2019; 12:117. [PMID: 31382962 PMCID: PMC6683425 DOI: 10.1186/s12920-019-0562-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/26/2019] [Indexed: 12/21/2022] Open
Abstract
Background microRNA (miRNA) is a short RNA (~ 22 nt) that regulates gene expression at the posttranscriptional level. Aberration of miRNA expressions could affect their targeting mRNAs involved in cancer-related signaling pathways. We conduct clustering analysis of miRNA and mRNA using expression data from the Cancer Genome Atlas (TCGA). We combine the Hungarian algorithm and blossom algorithm in graph theory. Data analysis is done using programming language R and Python. Methods We first quantify edge-weights of the miRNA-mRNA pairs by combining their expression correlation coefficient in tumor (T_CC) and correlation coefficient in normal (N_CC). We thereby introduce a bipartite graph partition procedure to identify cluster candidates. Specifically, we propose six weight formulas to quantify the change of miRNA-mRNA expression T_CC relative to N_CC, and apply the traditional hierarchical clustering to subjectively evaluate the different weight formulas of miRNA-mRNA pairs. Among these six different weight formulas, we choose the optimal one, which we define as the integrated mean value weights, to represent the connections between miRNA and mRNAs. Then the Hungarian algorithm and the blossom algorithm are employed on the miRNA-mRNA bipartite graph to passively determine the clusters. The combination of Hungarian and the blossom algorithms is dubbed maximum weighted merger method (MWMM). Results MWMM identifies clusters of different sizes that meet the mathematical criterion that internal connections inside a cluster are relatively denser than external connections outside the cluster and biological criterion that the intra-cluster Gene Ontology (GO) term similarities are larger than the inter-cluster GO term similarities. MWMM is developed using breast invasive carcinoma (BRCA) as training data set, but can also applies to other cancer type data sets. MWMM shows advantage in GO term similarity in most cancer types, when compared to other algorithms. Conclusions miRNAs and mRNAs that are likely to be affected by common underlying causal factors in cancer can be clustered by MWMM approach and potentially be used as candidate biomarkers for different cancer types and provide clues for targets of precision medicine in cancer treatment. Electronic supplementary material The online version of this article (10.1186/s12920-019-0562-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lizhong Ding
- Department of Biology, Indiana State University, Terre Haute, IN, 47809, USA
| | - Zheyun Feng
- Department of Mathematics and Computer Science, Indiana State University, Terre Haute, IN, 47809, USA
| | - Yongsheng Bai
- Department of Biology, Indiana State University, Terre Haute, IN, 47809, USA. .,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
17
|
Yang H, Liu JX, Shang HX, Lin S, Zhao JY, Lin JM. Qingjie Fuzheng granules inhibit colorectal cancer cell growth by the PI3K/AKT and ERK pathways. World J Gastrointest Oncol 2019; 11:377-392. [PMID: 31139308 PMCID: PMC6522764 DOI: 10.4251/wjgo.v11.i5.377] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/17/2018] [Accepted: 01/03/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Qingjie Fuzheng granules (QFGs) are part of a traditional Chinese medicine formula, which has been widely used and found to be clinically effective with few side effects in various cancer treatments, including colorectal cancer (CRC). However, the precise mechanisms and molecular signaling pathways involved in the activity of QFGs' anticancer effect have not been reported in the literature. In this study, we hypothesized that QFGs can inhibit the growth of colorectal cancer cells, and that its mechanism is closely related to one or more intracellular signal transduction pathways. AIM To better evaluate the mechanism underlying the anti-cancer effect of QFGs on the CRC cell lines HCT-116 and HCT-8. METHOD First, we measured cell viability and cytotoxicity by performing MTT and lactate dehydrogenase (LDH) assays. We evaluated the role of QFGs in cell proliferation and apoptosis by assessing colony formation and analyzing Hoechst 33258 staining. Second, cell cycle and apoptosis rates were measured by fluorescence activated cell sorting, and the expression levels of survivin, cyclin D1, CDK4, p21, Bax, Bcl-2, Fas, FasL, and cleaved-caspase-3/-8/-9 were measured by performing western blots and caspase activity assays. Furthermore, inhibitors of caspase-3/-8/-9 were used to elucidate the specific apoptosis pathway induced by QFGs in cancer cells. Finally, activation of the PI3K/AKT and ERK signaling pathways was examined using the western blot assay to investigate the possible mechanism. RESULTS MTT and LDH assays revealed that after 0.5-2.0 mg/mL of QFGs treatment, cell viability was reduced by (6.90% ± 1.03%)-(59.70% ± 1.51%) (HCT-116; P < 0.05) and (5.56% ± 4.52%)-(49.44% ± 2.47%) (HCT-8; P < 0.05), and cytotoxicity was increased from 0.52 ± 0.023 to 0.77 ± 0.002 (HCT-116; P < 0.01) and from 0.56 ± 0.054 to 0.81 ± 0.044 (HCT-8; P < 0.01) compared with the non-QFGs treatment groups. Additionally, colony formation and Hoechst 33258 staining assays showed that QFGs inhibited proliferation and induced apoptosis in CRC cells. QFGs also increased the expression levels of Bax, Fas and FasL, decreased the level of Bcl-2, and stimulated the activation of caspase-3/-8/-9, which were revealed by western blot and caspase activity assays. In contrast, when adding the three caspase inhibitors, the suppression effect of QFGs on cell viability and apoptosis were markedly inhibited. Moreover, QFGs suppressed the phosphorylation levels of PI3K, AKT and ERK. CONCLUSION These results demonstrated that QFGs can inhibit CRC cell proliferation and induce apoptosis by suppressing the PI3K/AKT and ERK signaling pathways.
Collapse
Affiliation(s)
- Hong Yang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
| | - Jian-Xin Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
| | - Hai-Xia Shang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
| | - Shan Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
| | - Jin-Yan Zhao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
| | - Jiu-Mao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian Province, China
| |
Collapse
|
18
|
MicroRNA in Lung Cancer Metastasis. Cancers (Basel) 2019; 11:cancers11020265. [PMID: 30813457 PMCID: PMC6406837 DOI: 10.3390/cancers11020265] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor metastasis is a hallmark of cancer, with distant metastasis frequently developing in lung cancer, even at initial diagnosis, resulting in poor prognosis and high mortality. However, available biomarkers cannot reliably predict cancer spreading sites. The metastatic cascade involves highly complicated processes including invasion, migration, angiogenesis, and epithelial-to-mesenchymal transition that are tightly controlled by various genetic expression modalities along with interaction between cancer cells and the extracellular matrix. In particular, microRNAs (miRNAs), a group of small non-coding RNAs, can influence the transcriptional and post-transcriptional processes, with dysregulation of miRNA expression contributing to the regulation of cancer metastasis. Nevertheless, although miRNA-targeted therapy is widely studied in vitro and in vivo, this strategy currently affords limited feasibility and a few miRNA-targeted therapies for lung cancer have entered into clinical trials to date. Advances in understanding the molecular mechanism of metastasis will thus provide additional potential targets for lung cancer treatment. This review discusses the current research related to the role of miRNAs in lung cancer invasion and metastasis, with a particular focus on the different metastatic lesions and potential miRNA-targeted treatments for lung cancer with the expectation that further exploration of miRNA-targeted therapy may establish a new spectrum of lung cancer treatments.
Collapse
|
19
|
Zhao W, Zhang LN, Wang XL, Zhang J, Yu HX. Long noncoding RNA NSCLCAT1 increases non-small cell lung cancer cell invasion and migration through the Hippo signaling pathway by interacting with CDH1. FASEB J 2019; 33:1151-1166. [PMID: 30148675 DOI: 10.1096/fj.201800408r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022]
Abstract
Metastatic growth is the leading cause of cancer-related death in non-small cell lung cancer (NSCLC). Metastasis is believed to be initiated by an increase in cell motility mediated by the loss of cell-cell adhesion because of the suppression of E-cadherin [encoded by cadherin 1 ( CDH1)]. However, very little is known about the molecular mechanism of CDH1 regulation. Therefore, we hypothesized that non-small cell lung cancer-associated transcript-1 (NSCLCAT1) suppresses functional CDH1 and mediates the Hippo signaling pathway, resulting in increased cell migration and invasion, and reduced apoptosis. Initially, microarray profiling and target prediction programs were employed to identify whether NSCLCAT1 targets CDH1. Next, quantitative PCR was used to determine the expression pattern of NSCLCAT1 in 114 specimens. The biologic functions of NSCLCAT1 in NSCLC were assessed through the up-regulation and down-regulation of the levels of endogenous NSCLCAT1 with the use of NSCLCAT1 vector or small interfering RNA against NSCLCAT1 in NSCLC cells. Furthermore, the Hippo signaling pathway in NSCLC cells was blocked by applying the verteporfin treatment to have a better understanding on the pivotal role of the Hippo signaling pathway in NSCLC. Microarray expression profiles of long noncoding RNAs, GSE19804 and GSE27262), revealed that NSCLCAT1 was up-regulated in NSCLC. Among patients with NSCLC, we determined that the NSCLCAT1 was robustly induced, whereas CDH1 was suppressed. The luciferase activity determination identified CDH1 as a NSCLCAT1 target. NSCLCAT1 was found to increase cell viability, migration, and invasion and to reduce apoptosis in NSCLC cells. The results from the quantitative PCR and Western blot analysis revealed that NSCLCAT1 modulated the Hippo signaling pathway. Furthermore, the inhibition of the Hippo signaling pathway by verteporfin treatment led to the loss of the effect of NSCLCAT1 on NSCLC cells. In summary, our findings suggested that NSCLCAT1 potentially has a role in NSCLC and NSCLCAT1-mediated regulation of the Hippo signaling pathway through the transcriptional repression of CDH1; therefore, the functional suppression or inhibition of NSCLCAT1 could be used as a novel therapeutic pathway in the control of aggressive and metastatic NSCLC.-Zhao, W., Zhang, L.-N., Wang, X.-L., Zhang, J., Yu, H.-X. Long noncoding RNA NSCLCAT1 increases non-small cell lung cancer cell invasion and migration through the Hippo signaling pathway by interacting with CDH1.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Thoracic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Le-Ning Zhang
- Department of Thoracic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xiao-Long Wang
- Department of Thoracic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ji Zhang
- Department of Thoracic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Hai-Xiang Yu
- Department of Thoracic Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
20
|
Liu C, Zhang Y, She X, Fan L, Li P, Feng J, Fu H, Liu Q, Liu Q, Zhao C, Sun Y, Wu M. A cytoplasmic long noncoding RNA LINC00470 as a new AKT activator to mediate glioblastoma cell autophagy. J Hematol Oncol 2018; 11:77. [PMID: 29866190 PMCID: PMC5987392 DOI: 10.1186/s13045-018-0619-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Despite the overwhelming number of investigations on AKT, little is known about lncRNA on AKT regulation, especially in GBM cells. METHODS RNA-binding protein immunoprecipitation assay (RIP) and RNA pulldown were used to confirm the binding of LINC00470 and fused in sarcoma (FUS). Confocal imaging, co-immunoprecipitation (Co-IP) and GST pulldown assays were used to detect the interaction between FUS and AKT. EdU assay, CCK-8 assay, and intracranial xenograft assays were performed to demonstrate the effect of LINC00470 on the malignant phenotype of GBM cells. RT-qPCR and Western blotting were performed to test the effect of LINC00470 on AKT and pAKT. RESULTS In this study, we demonstrated that LINC00470 was a positive regulator for AKT activation in GBM. LINC00470 bound to FUS and AKT to form a ternary complex, anchoring FUS in the cytoplasm to increase AKT activity. Higher pAKT activated by LINC00470 inhibited ubiquitination of HK1, which affected glycolysis, and inhibited cell autophagy. Furthermore, higher LINC00470 expression was associated with GBM tumorigenesis and poor patient prognosis. CONCLUSIONS Our findings revealed a noncanonical AKT activation signaling pathway, i.e., LINC00470 directly interacts with FUS, serving as an AKT activator to promote GBM progression. LINC00470 has an important referential significance to evaluate the prognosis of patients.
Collapse
Affiliation(s)
- Changhong Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410006, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Yan Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410006, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Xiaoling She
- Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Li Fan
- Department of Biochemistry, University of California, Riverside, CA, 92521, USA
| | - Peiyao Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410006, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Jianbo Feng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410006, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Haijuan Fu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410006, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Qing Liu
- Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qiang Liu
- Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Chunhua Zhao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410006, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Yingnan Sun
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410006, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, 410078, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Minghua Wu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, 410078, Hunan, China.
- Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China.
| |
Collapse
|
21
|
Lei W, Kang W, Nan Y, Lei Z, Zhongdong L, Demin L, Lei S, Hairong H. The Downregulation of miR-200c Promotes Lactate Dehydrogenase A Expression and Non-Small Cell Lung Cancer Progression. Oncol Res 2018; 26:1015-1022. [PMID: 29321091 PMCID: PMC7844790 DOI: 10.3727/096504018x15151486241153] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
This study was aimed to investigate the function and mechanism of microRNA-200c (miR-200c) in the progression of non-small cell lung cancer (NSCLC). A total of 76 patients diagnosed as having NSCLC were enrolled in this study. The expression level of miR-200c in NSCLC tissues and cell lines was investigated using the quantitative real-time polymerase chain reaction (RT-qPCR) method. We found that the expression of miR-200c was significantly reduced in NSCLC tissues and cell lines compared with normal lung tissues and the human bronchial epithelial cell line. Overexpression of miR-200c using the miR-200c mimic significantly suppressed cell proliferation and migration of NSCLC cell lines. The results of the luciferase reporter assay identified lactate dehydrogenase A (LDHA) as a direct target of miR-200c. The expression of LDHA was shown to be suppressed in NSCLC cell lines with miR-200c mimic transfection. Furthermore, the transfection of small interfering RNA (siRNA) targeting LDHA suppressed the proliferation and migration of NSCLC cell lines. In summary, our results presented in this study suggested that miR-200c was able to inhibit the proliferation and migration of NSCLC cells by downregulating LDHA. Therefore, miR-200c may be considered as a potential candidate for the treatment of NSCLC.
Collapse
Affiliation(s)
- Wei Lei
- Department of Cardiothoracic Surgery, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, P.R. China
| | - Wang Kang
- Department of Cardiothoracic Surgery, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, P.R. China
| | - Yang Nan
- Department of Cardiothoracic Surgery, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, P.R. China
| | - Zhang Lei
- Department of Cardiothoracic Surgery, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, P.R. China
| | - Li Zhongdong
- Department of Cardiothoracic Surgery, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, P.R. China
| | - Li Demin
- Department of Cardiothoracic Surgery, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, P.R. China
| | - Sun Lei
- Department of Cardiothoracic Surgery, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, P.R. China
| | - Huang Hairong
- Department of Cardiothoracic Surgery, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, P.R. China
| |
Collapse
|