51
|
Zhang L, Xu X, Su X. Noncoding RNAs in cancer immunity: functions, regulatory mechanisms, and clinical application. Mol Cancer 2020; 19:48. [PMID: 32122338 PMCID: PMC7050126 DOI: 10.1186/s12943-020-01154-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
It is well acknowledged that immune system is deeply involved in cancer initiation and progression, and can exert both pro-tumorigenic and anti-tumorigenic effects, depending on specific microenvironment. With the better understanding of cancer-associated immune cells, especially T cells, immunotherapy was developed and applied in multiple cancers and exhibits remarkable efficacy. However, currently only a subset of patients have responses to immunotherapy, suggesting that a boarder view of cancer immunity is required. Non-coding RNAs (ncRNAs), mainly including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are identified as critical regulators in both cancer cells and immune cells, thus show great potential to serve as new therapeutic targets to improve the response of immunotherapy. In this review, we summarize the functions and regulatory mechanisms of ncRNAs in cancer immunity, and highlight the potential of ncRNAs as novel targets for immunotherapy.
Collapse
Affiliation(s)
- Le Zhang
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, 1 Tong Dao Street, Huimin District, Hohhot, 010050, Inner Mongolia, China
| | - Xiaonan Xu
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, FL, 33612-9497, USA
| | - Xiulan Su
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, 1 Tong Dao Street, Huimin District, Hohhot, 010050, Inner Mongolia, China.
| |
Collapse
|
52
|
Role of Non-Coding RNAs in the Development of Targeted Therapy and Immunotherapy Approaches for Chronic Lymphocytic Leukemia. J Clin Med 2020; 9:jcm9020593. [PMID: 32098192 PMCID: PMC7074107 DOI: 10.3390/jcm9020593] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/15/2022] Open
Abstract
In the past decade, novel targeted therapy approaches, such as BTK inhibitors and Bcl2 blockers, and innovative treatments that regulate the immune response against cancer cells, such as monoclonal antibodies, CAR-T cell therapy, and immunomodulatory molecules, have been established to provide support for the treatment of patients. However, drug resistance development and relapse are still major challenges in CLL treatment. Several studies revealed that non-coding RNAs have a main role in the development and progression of CLL. Specifically, microRNAs (miRs) and tRNA-derived small-RNAs (tsRNAs) were shown to be outstanding biomarkers that can be used to diagnose and monitor the disease and to possibly anticipate drug resistance and relapse, thus supporting physicians in the selection of treatment regimens tailored to the patient needs. In this review, we will summarize the most recent discoveries in the field of targeted therapy and immunotherapy for CLL and discuss the role of ncRNAs in the development of novel drugs and combination regimens for CLL patients.
Collapse
|
53
|
Orso F, Quirico L, Dettori D, Coppo R, Virga F, Ferreira LC, Paoletti C, Baruffaldi D, Penna E, Taverna D. Role of miRNAs in tumor and endothelial cell interactions during tumor progression. Semin Cancer Biol 2020; 60:214-224. [DOI: 10.1016/j.semcancer.2019.07.024] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022]
|
54
|
Wang J, Yan S, Yang J, Lu H, Xu D, Wang Z. Non-coding RNAs in Rheumatoid Arthritis: From Bench to Bedside. Front Immunol 2020; 10:3129. [PMID: 32047497 PMCID: PMC6997467 DOI: 10.3389/fimmu.2019.03129] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022] Open
Abstract
Rheumatoid arthritis is a common systemic and autoimmune disease characterized by symmetrical and inflammatory destruction of distal joints. Its primary pathological characters are synovitis and vasculitis. Accumulating studies have implicated the critical role of non-coding RNAs (ncRNAs) in inflammation and autoimmune regulation, primarily including microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA). NcRNAs are significant regulators in distinct physiological and pathophysiological processes. Many validated non-coding RNAs have been identified as promising biomarkers for the diagnosis and treatment of RA. This review will shed some light on RA pathogenesis and be helpful for identifying potential ncRNA biomarkers for RA.
Collapse
Affiliation(s)
- Jinghua Wang
- Clinical Medicine College, Weifang Medical University, Weifang, China.,Department of Rheumatology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Shushan Yan
- Department of Gastrointestinal and Anal Diseases Surgery, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jinghan Yang
- Clinical Medicine College, Weifang Medical University, Weifang, China.,Department of Rheumatology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongying Lu
- Functional Laboratory, Clinical Medicine College of Weifang Medical University, Weifang, China
| | - Donghua Xu
- Clinical Medicine College, Weifang Medical University, Weifang, China.,Department of Rheumatology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zengyan Wang
- Department of Operating Room, Zhucheng People's Hospital, Zhucheng, China
| |
Collapse
|
55
|
Ding Y, Bi L, Wang J. MiR-1180 promotes cardiomyocyte cell cycle re-entry after injury through the NKIRAS2-NFκB pathway. Biochem Cell Biol 2020; 98:449-457. [PMID: 31955591 DOI: 10.1139/bcb-2019-0364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Heart failure (HF) is associated with a considerable number of symptoms and significantly impaired health for humans, including reduced quality of life and physical functioning. Previous studies have indicated that miRNAs have important roles in regulating the development of HF. MiR-1180 is involved in the proliferation, migration, invasiveness, and chemoresistance of cancer cells; however, the underlying mechanisms and role of miR-1180 in the functioning of cardiomyocytes remains unclear. In this study, we found that miR-1180 promotes cell activity and cell cycle processes by driving energy generation through NKIRAS2, which declines over time during development. The expression of miR-1180 is down-regulated in cells subjected to hypoxia-reoxygenation, and use of an miR-1180 mimic significantly reduced myocardial injury and cell apoptosis. In addition, miR-1180 regulates the NFκB pathway through NKIRAS2 in cardiomyocytes. These findings suggest that miR-1180 maybe a novel therapeutic target for use in getting cardiomyocytes to re-enter the cell cycle as well as for cardiac repair following myocardial injury.
Collapse
Affiliation(s)
- Yuhui Ding
- Department of Emergency, Qingdao Haici Medical Group, Qingdao, China 266034
| | - Liyuan Bi
- Department of Emergency, Qingdao Haici Medical Group, Qingdao, China 266034
| | - Jun Wang
- Department of Emergency, Qingdao Haici Medical Group, Qingdao, China 266034.,Department of Emergency, Qingdao Haici Medical Group, Qingdao, China 266034
| |
Collapse
|
56
|
Conti I, Varano G, Simioni C, Laface I, Milani D, Rimondi E, Neri LM. miRNAs as Influencers of Cell-Cell Communication in Tumor Microenvironment. Cells 2020; 9:cells9010220. [PMID: 31952362 PMCID: PMC7016744 DOI: 10.3390/cells9010220] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level, inducing the degradation of the target mRNA or translational repression. MiRNAs are involved in the control of a multiplicity of biological processes, and their absence or altered expression has been associated with a variety of human diseases, including cancer. Recently, extracellular miRNAs (ECmiRNAs) have been described as mediators of intercellular communication in multiple contexts, including tumor microenvironment. Cancer cells cooperate with stromal cells and elements of the extracellular matrix (ECM) to establish a comfortable niche to grow, to evade the immune system, and to expand. Within the tumor microenvironment, cells release ECmiRNAs and other factors in order to influence and hijack the physiological processes of surrounding cells, fostering tumor progression. Here, we discuss the role of miRNAs in the pathogenesis of multicomplex diseases, such as Alzheimer’s disease, obesity, and cancer, focusing on the contribution of both intracellular miRNAs, and of released ECmiRNAs in the establishment and development of cancer niche. We also review growing evidence suggesting the use of miRNAs as novel targets or potential tools for therapeutic applications.
Collapse
Affiliation(s)
- Ilaria Conti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Gabriele Varano
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Carolina Simioni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Ilaria Laface
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Daniela Milani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Erika Rimondi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Luca M. Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
- LTTA—Electron Microscopy Center, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: ; Tel.: +39-0532-455940
| |
Collapse
|
57
|
Li M, Cui X, Guan H. MicroRNAs: pivotal regulators in acute myeloid leukemia. Ann Hematol 2020; 99:399-412. [PMID: 31932900 DOI: 10.1007/s00277-019-03887-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 12/04/2019] [Indexed: 02/08/2023]
Abstract
MicroRNAs are a class of small non-coding RNAs that are 19-22 nucleotides in length and regulate a variety of biological processes at the post-transcriptional level. MicroRNA dysregulation disrupts normal biological processes, resulting in tumorigenesis. Acute myeloid leukemia is an invasive hematological malignancy characterized by the abnormal proliferation and differentiation of immature myeloid cells. Due to the low 5-year survival rate, there is an urgent need to discover novel diagnostic markers and therapeutic targets. In recent years, microRNAs have been shown to play important roles in hematological malignancies by acting as tumor suppressors and oncogenes. MicroRNAs have the potential to be a breakthrough in the diagnosis and treatment of acute myeloid leukemia. In this review, we summarize the biology of microRNAs and discuss the relationships between microRNA dysregulation and acute myeloid leukemia in the following aspects: signaling pathways, the abnormal biological behavior of acute myeloid leukemia cells, the clinical application of microRNAs and competing endogenous RNA regulatory networks.
Collapse
Affiliation(s)
- Mingyu Li
- Department of Clinical Hematology, Medical College of Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Xianglun Cui
- Department of Inspection, Medical College of Qingdao University, Qingdao, 266071, China
| | - Hongzai Guan
- Department of Clinical Hematology, Medical College of Qingdao University, 308 Ningxia Road, Qingdao, 266071, China.
| |
Collapse
|
58
|
Zhang Y, Zhang Z, Wei R, Miao X, Sun S, Liang G, Chu C, Zhao L, Zhu X, Guo Q, Wang B, Li X. IL (Interleukin)-6 Contributes to Deep Vein Thrombosis and Is Negatively Regulated by miR-338-5p. Arterioscler Thromb Vasc Biol 2019; 40:323-334. [PMID: 31852218 PMCID: PMC6975520 DOI: 10.1161/atvbaha.119.313137] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: Deep venous thrombosis (DVT), one of the most common venous thromboembolic disorders, is closely linked with pulmonary embolism and post-thrombotic syndrome, both of which have a high mortality. However, the factors that trigger DVT formation are still largely unknown. Elevated expression of IL (interleukin)-6—an important inflammatory cytokine—has been linked with DVT formation. However, the molecular mechanisms leading to the elevated IL-6 in DVT remain unclear. Here, we proposed that epigenetic modification of IL-6 at the post-transcriptional level may be a crucial trigger for IL-6 upregulation in DVT. Approach and Results: To explore the association between microRNAs and IL-6 in DVT, we performed microRNA microarray analysis and experiments both in vitro and in vivo. Microarray and quantitative real-time polymerase chain reaction results showed that IL-6 expression was increased while miR-338-5p level was decreased substantially in peripheral blood mononuclear cells of patients with DVT, and there was significant negative correlation between miR-338-5p and IL-6. Experiments in vitro showed that overexpressed miR-338-5p reduced IL-6 expression, while miR-338-5p knockdown increased IL-6 expression. Moreover, our in vivo study found that mice with anti–IL-6 antibody or agomiR-338-5p delivery resulted in decreased IL-6 expression and alleviated DVT formation, whereas antagomiR-338-5p acted inversely. Most of miR-338-5p was found located in cytoplasm by fluorescence in situ hybridization. Dual-luciferase reporter assay identified direct binding between miR-338-5p and IL-6. Conclusions: Our results suggest that decreased miR-338-5p promotes DVT formation by increasing IL-6 expression.
Collapse
Affiliation(s)
- Yunhong Zhang
- From the School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, China (Y.Z., C.C.).,Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.)
| | - Zhen Zhang
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.)
| | - Ran Wei
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.)
| | - Xiuming Miao
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China (X.M., G.L., B.W.)
| | - Shangwen Sun
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.).,Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (S.S.)
| | - Gang Liang
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China (X.M., G.L., B.W.)
| | - Chu Chu
- From the School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, China (Y.Z., C.C.).,Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.)
| | - Lin Zhao
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.)
| | - Xiaoxiao Zhu
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.)
| | - Qiang Guo
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.)
| | - Bin Wang
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China (X.M., G.L., B.W.)
| | - Xia Li
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (Y.Z., Z.Z., R.W., S.S., C.C., L.Z., X.Z., Q.G., X.L.)
| |
Collapse
|
59
|
Li L, Ouyang Y, Wang W, Hou D, Zhu Y. The landscape and prognostic value of tumor-infiltrating immune cells in gastric cancer. PeerJ 2019; 7:e7993. [PMID: 31844561 PMCID: PMC6910118 DOI: 10.7717/peerj.7993] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/04/2019] [Indexed: 12/16/2022] Open
Abstract
Background Gastric cancer (GC) is the fourth most frequently diagnosed malignancy and the second leading cause of cancer-associated mortality worldwide. The tumor microenvironment, especially tumor-infiltrating immune cells (TIICs), exhibits crucial roles both in promoting and inhibiting cancer growth. The aim of the present study was to evaluate the landscape of TIICs and develop a prognostic nomogram in GC. Materials and Methods A gene expression profile obtained from a dataset from The Cancer Genome Atlas (TCGA) was used to quantify the proportion of 22 TIICs in GC by the CIBERSORT algorithm. LASSO regression analysis and multivariate Cox regression were applied to select the best survival-related TIICs and develop an immunoscore formula. Based on the immunoscore and clinical information, a prognostic nomogram was built, and the predictive accuracy of it was evaluated by the area under the curve (AUC) of the receiver operating characteristic curve (ROC) and the calibration plot. Furthermore, the nomogram was validated by data from the International Cancer Genome Consortium (ICGC) dataset. Results In the GC samples, macrophages (25.3%), resting memory CD4 T cells (16.2%) and CD8 T cells (9.7%) were the most abundant among 22 TIICs. Seven TIICs were filtered out and used to develop an immunoscore formula. The AUC of the prognostic nomogram in the TCGA set was 0.772, similar to that in the ICGC set (0.730) and whole set (0.748), and significantly superior to that of TNM staging alone (0.591). The calibration plot demonstrated an outstanding consistency between the prediction and actual observation. Survival analysis revealed that patients with GC in the high-immunoscore group exhibited a poor clinical outcome. The result of multivariate analysis revealed that the immunoscore was an independent prognostic factor. Discussion The immunoscore could be used to reinforce the clinical outcome prediction ability of the TNM staging system and provide a convenient tool for risk assessment and treatment selection for patients with GC.
Collapse
Affiliation(s)
- Linhai Li
- Department of General Surgery, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yiming Ouyang
- Department of General Surgery, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Wenrong Wang
- Department of General Surgery, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Dezhi Hou
- Department of General Surgery, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yu Zhu
- Department of General Surgery, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
60
|
Liang LB, Zhu WJ, Chen XM, Luo FM. Plasma miR-30a-5p as an early novel noninvasive diagnostic and prognostic biomarker for lung cancer. Future Oncol 2019; 15:3711-3721. [PMID: 31664862 DOI: 10.2217/fon-2019-0393] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Aim: Circulation miRNAs have become increasingly appreciated in the diagnosis and prognosis of lung cancer. This study aims to identify and evaluate plasma miRNA-30a-5p as an early noninvasive biomarker for the diagnosis and prognosis of lung cancer. Pateints & methods: Expression levels of plasma miRNA 30a-5p were measured by quantitative real-time PCR. Receiver operating characteristic analysis and area under the curve were used to differentiate malignant from benign tumors and from healthy controls. Kaplan-Meier curves and Cox regression were used to determine survival and prognosis. Results: Our results suggest that the level of miRNA-30a-5p in plasma might be a considerable early novel noninvasive diagnostic and prognostic biomarker for lung cancer. Conclusion: Prospective studies must be performed to confirm this new early novel noninvasive diagnostic and prognostic biomarker for lung cancer.
Collapse
Affiliation(s)
- Ling-Bo Liang
- Division of General Practice & Section for Pedagogic Research on General Practice, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wen-Jun Zhu
- Department of Respiratory & Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xue-Mei Chen
- Research Core Facility, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Feng-Ming Luo
- Department of Respiratory & Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| |
Collapse
|
61
|
Rios-Colon L, Deep G, Kumar D. Emerging role of microRNA 628-5p as a novel biomarker for cancer and other diseases. Tumour Biol 2019; 41:1010428319881344. [PMID: 31608792 DOI: 10.1177/1010428319881344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs are a family of small, single-stranded RNAs that have key roles in regulating multiple signaling pathways within a cell. Studies have implicated aberrant expression of microRNAs in the development and progression of several pathologies including cancer. MicroRNAs are relatively stable and readily available in body fluids and tissues, making them desirable biomarkers for prognostic and diagnostic purposes in an array of diseases. MicroRNA 628 (5p/3p variants) is located in the 15q21.3 cancer-related region, and evidence suggests its association with various pathologies. The -5p mature variant, microRNA 628-5p, has been reported to be differentially expressed in various cancers, and its expression has been mostly associated with tumor suppression but there are few reports identifying its role in cancer progression. Several studies have also suggested its utility in diagnosis and prognosis of various cancers. Dysregulation of microRNA 628-5p has also been implicated in embryonal implantation defects, autism, immune modulation, myogenesis, cardiovascular disease, viral infection, and skeletal muscle repair. Here, we have provided a comprehensive review on available literature explaining the role of microRNA 628-5p as a potential cancer biomarker as well as briefly describe its function in other diseases and normal physiological conditions.
Collapse
Affiliation(s)
- Leslimar Rios-Colon
- Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University, Durham, NC, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical/Biotechnology Research Institute (JLC-BBRI), North Carolina Central University, Durham, NC, USA
| |
Collapse
|
62
|
Zhang Q, Zhu B, Qian J, Wang K, Zhou J. miR-942 promotes proliferation and metastasis of hepatocellular carcinoma cells by inhibiting RRM2B. Onco Targets Ther 2019; 12:8367-8378. [PMID: 31632084 PMCID: PMC6795128 DOI: 10.2147/ott.s207549] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/16/2019] [Indexed: 12/24/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death. MicroRNA-942 (miR-942) plays a critical role in promoting proliferation and metastasis of cancer cells and is associated with poor prognosis in some types of cancers. However, the prognostic value of miR-942 and its functional role in HCC remain unclear. Materials and methods Real-time PCR (RT-PCR) was used to detect the expression of miR-942 in HCC tissues and adjacent normal liver tissues. Next, the correlations between miR-942 expression and clinicopathological parameters including the survival rate were analyzed. Interaction between miR-942 and ribonucleotide reductase regulatory TP53 inducible subunit M2B (RRM2B) was determined by RT-PCR, Western blot and luciferase assay. The biological influence of miR-942 on HCC cell lines was studied using CCK-8 assay, colony formation assay and transwell assay in vitro. Western blot and RT-PCR were used to analyze the change of downstream genes after miR-942 mimics transfection. Results miR-942 was significantly up-regulated in HCC. Its high expression was associated with serum alanine transaminase level (P=0.0350), tumor size (P=0.0195), T stage (P=0.0045) and lymphatic metastasis (P=0.0013). High expression of miR-942 was associated with shorter overall survival and disease-free survival time of HCC patients. RRM2B was validated as a target gene of miR-942. miR-942 mimics markedly promoted the malignant phenotypes of Huh7 and MHCC97H cell lines, while its inhibitor had the opposite effect. miR-942 can regulate the downstream genes of RRM2B including Egr-1 and PTEN, markers of epithelial-mesenchymal transition and matrix metalloproteinases. Conclusion miR-942 may serve as a potential biomarker for HCC and its inhibitor may be a therapeutic agent for the treatment of this deadly disease.
Collapse
Affiliation(s)
- Qifan Zhang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Bili Zhu
- Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jianping Qian
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Kai Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jie Zhou
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| |
Collapse
|
63
|
Wang S, Wang W, Han X, Wang Y, Ge Y, Tan Z. Dysregulation of miR484-TUSC5 axis takes part in the progression of hepatocellular carcinoma. J Biochem 2019; 166:271-279. [PMID: 31157375 DOI: 10.1093/jb/mvz034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/25/2019] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. miR-484 is previously reported to be a crucial modulator during the process from precancerous lesion to cancer. Tumour suppressor candidate 5 (TUSC5) is a potential tumour suppressor, but its expression and function in HCC are obscure. In this study, we aimed to explore the roles of miR-484 and TUSC5 in HCC, and clarify the relationship between them. We demonstrated that miR-484 was significantly up-regulated in HCC, while TUSC5 was down-regulated. TUSC5 was validated as the target gene of miR-484 and both of them were associated with the prognosis of HCC patients. miR-484 mimics markedly promoted the malignant phenotypes while TUSC5 plasmid had the opposite effect. In conclusion, miR-484/TUSC5 is potential diagnostic biomarkers and therapy targets for HCC.
Collapse
Affiliation(s)
- Shanzong Wang
- Department of Pathology, The Third People's Hospital of Linyi, Huaxia Road No. 117, Linyi Economic and Technological Development Zone, Linyi, Shandong, China
| | - Weijuan Wang
- Department of Gynaecology, The Third People's Hospital of Linyi, Huaxia Road No. 117, Linyi Economic and Technological Development Zone, Linyi, Shandong, China
| | - Xiaoguang Han
- Department of Internal Medicine, The Third People's Hospital of Linyi, Huaxia Road No. 117, Linyi Economic and Technological Development Zone, Linyi, Shandong, China
| | - Youli Wang
- Department of Pathology, The Third People's Hospital of Linyi, Huaxia Road No. 117, Linyi Economic and Technological Development Zone, Linyi, Shandong, China
| | - Yunzhen Ge
- Department of Pathology, The Third People's Hospital of Linyi, Huaxia Road No. 117, Linyi Economic and Technological Development Zone, Linyi, Shandong, China
| | - Zhen Tan
- Department of Pathology, The Third People's Hospital of Linyi, Huaxia Road No. 117, Linyi Economic and Technological Development Zone, Linyi, Shandong, China
| |
Collapse
|
64
|
Xu D, Dong P, Xiong Y, Yue J, Ihira K, Konno Y, Kobayashi N, Todo Y, Watari H. MicroRNA-361: A Multifaceted Player Regulating Tumor Aggressiveness and Tumor Microenvironment Formation. Cancers (Basel) 2019; 11:E1130. [PMID: 31394811 PMCID: PMC6721607 DOI: 10.3390/cancers11081130] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/01/2019] [Accepted: 08/01/2019] [Indexed: 12/22/2022] Open
Abstract
MicroRNA-361-5p (miR-361) expression frequently decreases or is lost in different types of cancers, and contributes to tumor suppression by repressing the expression of its target genes implicated in tumor growth, epithelial-to-mesenchymal transition (EMT), metastasis, drug resistance, glycolysis, angiogenesis, and inflammation. Here, we review the expression pattern of miR-361 in human tumors, describe the mechanisms responsible for its dysregulation, and discuss how miR-361 modulates the aggressive properties of tumor cells and alter the tumor microenvironment by acting as a novel tumor suppressor. Furthermore, we describe its potentials as a promising diagnostic or prognostic biomarker for cancers and a promising target for therapeutic development.
Collapse
Affiliation(s)
- Daozhi Xu
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo 060-8638, Japan.
| | - Ying Xiong
- Department of Gynecology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Kei Ihira
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Yosuke Konno
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Noriko Kobayashi
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Yukiharu Todo
- Division of Gynecologic Oncology, National Hospital Organization, Hokkaido Cancer Center, Sapporo 003-0804, Japan
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo 060-8638, Japan.
| |
Collapse
|
65
|
Chen D, Lu T, Tan J, Li H, Wang Q, Wei L. Long Non-coding RNAs as Communicators and Mediators Between the Tumor Microenvironment and Cancer Cells. Front Oncol 2019; 9:739. [PMID: 31448238 PMCID: PMC6691164 DOI: 10.3389/fonc.2019.00739] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/23/2019] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are a class of more than 200 nucleotides RNA transcripts which have limited protein coding capacity. They regulate numerous biological processes in cancers through diverse molecular mechanisms. Aberrant expression of lncRNAs has been frequently associated with human cancer. Furthermore, the tumor microenvironment (TME) is composed of different cells such as cancer-associated fibroblasts (CAFs), endothelial cells and infiltrated immune cells, and all of which participate in communication with tumor cells affecting the progression of tumor. LncRNAs are directly and indirectly involved in the crosstalk between stromal cells and tumor cells and dysregulated lncRNAs expression in these cells could drive tumorigenesis. In this review, we explore the influence of aberrantly expressed lncRNAs in tumor progression, clarify the critical roles of lncRNAs in the TME, summarize findings on crosstalk between infiltrated immune cells, CAFs, endothelial cells, and tumor cells via lncRNAs, and discuss the promise of lncRNAs as tumor diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Di Chen
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tong Lu
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junying Tan
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hao Li
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qiuyue Wang
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liangzhou Wei
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
66
|
Medeiros M, Ribeiro AO, Lupi LA, Romualdo GR, Pinhal D, Chuffa LGDA, Delella FK. Mimicking the tumor microenvironment: Fibroblasts reduce miR-29b expression and increase the motility of ovarian cancer cells in a co-culture model. Biochem Biophys Res Commun 2019; 516:96-101. [PMID: 31200958 DOI: 10.1016/j.bbrc.2019.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/01/2019] [Indexed: 12/14/2022]
Abstract
Ovarian cancer (OC) is a highly prevalent gynecological malignancy worldwide. Throughout ovarian carcinogenesis, the crosstalk between cellular components of the microenvironment, including tumor cells and fibroblasts, is proposed to play critical roles in cancer progression. The dysregulation of microRNA expression is also a pronounced feature of the OC. The screening of microRNAs, mainly those involved in OC microenvironment, could have diagnostic and/or therapeutic potential for this malignancy. Thus, we assessed the influence of fibroblasts on microRNA expression and the motility of OC cells. To achieve this goal, SKOV-3 cancer cells were co-cultured with human normal fibroblasts derived from primary culture (FP-96). Cell viability, expression of tumor suppressor microRNAs and oncomiRs by RT-qPCR, cell migration by wound healing assay and analysis of MMP-2 activity by zymography were performed in SKOV-3 cells. Moreover, α-smooth muscle actin (α-SMA) expression was evaluated by Western blot in FP-96 fibroblasts. Notably, the co-culture downregulated the tumor suppressor miR-29b and increased migration of SKOV-3 cells. In addition, co-culture increased the activity of MMP-2, which is a miR-29 target, and accounted for extracellular matrix remodeling and augmented cellular motility. Concomitantly, the co-culture system induced α-SMA expression in FP-96 fibroblasts, the commonly expressed marker in cancer-associated fibroblasts (CAFs). Our findings suggest that the potential crosstalk between OC cells and fibroblasts in tumor microenvironment may play a key role in the progression of OC.
Collapse
Affiliation(s)
- Mariana Medeiros
- Sao Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu, Sao Paulo, Brazil
| | - Amanda Oliveira Ribeiro
- Sao Paulo State University (UNESP), Institute of Biosciences, Department of Genetics, Botucatu, Sao Paulo, Brazil
| | - Luiz Antônio Lupi
- Sao Paulo State University (UNESP), Institute of Biosciences, Department of Anatomy, Botucatu, Sao Paulo, Brazil
| | - Guilherme Ribeiro Romualdo
- Sao Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu, Sao Paulo, Brazil
| | - Danillo Pinhal
- Sao Paulo State University (UNESP), Institute of Biosciences, Department of Genetics, Botucatu, Sao Paulo, Brazil
| | | | - Flávia Karina Delella
- Sao Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu, Sao Paulo, Brazil.
| |
Collapse
|
67
|
Li Y, Chen F, Chu J, Wu C, Li Y, Li H, Ma H. miR-148-3p Inhibits Growth of Glioblastoma Targeting DNA Methyltransferase-1 (DNMT1). Oncol Res 2019; 27:911-921. [PMID: 30982493 PMCID: PMC7848282 DOI: 10.3727/096504019x15516966905337] [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] [Indexed: 12/16/2022] Open
Abstract
To date, miR-148-3p and DNMT1–recombinant human runt-related transcription factor 3 (RUNX3) axis have been linked to cell proliferation, migration, and invasion; however, their roles and relationships in human glioblastoma multiforme (GBM) are still not clear. Here we found that the expression of miR-148-3p in glioma tissues was decreased compared with adjacent nontumor tissues and correlated with WHO grade, tumor size, and prognosis as well as DNMT1 and RUNX3 expressions. Compared with NHA cells, the expression of miR-148-3p in U87 and U251 cells was also downregulated and accompanied with upregulation of DNMT1 and hypermethylation level of RUNX3 promoter region. miR-148-3p overexpression induced apoptosis and cell cycle arrest of U87 and U251 cells, and affected cell migration and invasion. miR-148-3p mimics effectively suppressed the expression of DNMT1 and methylation of RUNX3 promoter, finally upregulating RUNX3 expression. Mechanistically, the 3′-untranslated region (3′-UTR) of DNMT1 was a direct target of miR-148-3p. Overexpression of miR-148-3p or inhibition of DNMT1 induced the expression of E-cadherin and reduced the expressions of N-cadherin, vimentin, MMP-2, and MMP-9. In conclusion, miR-148-3p directly repressed the expression of DNMT1 and inhibited proliferation, migration, and invasion by regulating DNMT1–RUNX3 axis and the epithelial–mesenchymal transition in GBM. Our findings provide a new foundation for treatment of patients with GBM.
Collapse
Affiliation(s)
- Yongtao Li
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Fanyu Chen
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Jiancheng Chu
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Chao Wu
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Yuan Li
- School of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Heng Li
- School of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Hongxin Ma
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| |
Collapse
|