1
|
Smith K, Beach D, Silva R, Balazs G, Salani F, Crea F. Comprehensive analysis of differentially expressed miRNAs in hepatocellular carcinoma: Prognostic, predictive significance and pathway insights. PLoS One 2024; 19:e0296198. [PMID: 38635644 PMCID: PMC11025735 DOI: 10.1371/journal.pone.0296198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Robust prognostic and predictive factors for hepatocellular carcinoma, a leading cause of cancer-related deaths worldwide, have not yet been identified. Previous studies have identified potential HCC determinants such as genetic mutations, epigenetic alterations, and pathway dysregulation. However, the clinical significance of these molecular alterations remains elusive. MicroRNAs are major regulators of protein expression. MiRNA functions are frequently altered in cancer. In this study, we aimed to explore the prognostic value of differentially expressed miRNAs in HCC, to elucidate their associated pathways and their impact on treatment response. To this aim, bioinformatics techniques and clinical dataset analyses were employed to identify differentially expressed miRNAs in HCC compared to normal hepatic tissue. We validated known associations and identified a novel miRNA signature with potential prognostic significance. Our comprehensive analysis identified new miRNA-targeted pathways and showed that some of these protein coding genes predict HCC patients' response to the tyrosine kinase inhibitor sorafenib.
Collapse
Affiliation(s)
- Kayleigh Smith
- Cancer Research Group-School of Life Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Dan Beach
- School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Roger Silva
- Department of Medicine, Cancer Research Program Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
| | - Gyorffy Balazs
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
- Research Centre for Natural Sciences, Institute of Molecular Life Sciences, Budapest, Hungary
| | - Francesca Salani
- Cancer Research Group-School of Life Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Institute of Interdisciplinary Research “Health Science”, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Francesco Crea
- Cancer Research Group-School of Life Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
2
|
Tossetta G. Metformin Improves Ovarian Cancer Sensitivity to Paclitaxel and Platinum-Based Drugs: A Review of In Vitro Findings. Int J Mol Sci 2022; 23:12893. [PMID: 36361682 PMCID: PMC9654053 DOI: 10.3390/ijms232112893] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 07/30/2023] Open
Abstract
Ovarian cancer is one of the most dangerous gynecologic cancers worldwide, showing a high fatality rate and recurrence due to diagnosis at an advanced stage of the disease and the occurrence of chemoresistance, which weakens the therapeutic effects of the chemotherapeutic treatments. In fact, although paclitaxel and platinum-based drugs (carboplatin or cisplatin) are widely used alone or in combination to treat ovarian cancer, the occurrence of chemoresistance significantly reduces the effects of these drugs. Metformin is a hypoglycemic agent that is commonly used for the treatment of type 2 diabetes mellitus and non-alcoholic fatty liver disease. However, this drug also shows anti-tumor activity, reducing cancer risk and chemoresistance. This review analyzes the current literature regarding the role of metformin in ovarian cancer and investigates what is currently known about its effects in reducing paclitaxel and platinum resistance to restore sensitivity to these drugs.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; ; Tel.: +39-0712206270
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126 Ancona, Italy
| |
Collapse
|
3
|
Maharati A, Zanguei AS, Khalili-Tanha G, Moghbeli M. MicroRNAs as the critical regulators of tyrosine kinase inhibitors resistance in lung tumor cells. Cell Commun Signal 2022; 20:27. [PMID: 35264191 PMCID: PMC8905758 DOI: 10.1186/s12964-022-00840-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/05/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the second most common and the leading cause of cancer related deaths globally. Tyrosine Kinase Inhibitors (TKIs) are among the common therapeutic strategies in lung cancer patients, however the treatment process fails in a wide range of patients due to TKIs resistance. Given that the use of anti-cancer drugs can always have side effects on normal tissues, predicting the TKI responses can provide an efficient therapeutic strategy. Therefore, it is required to clarify the molecular mechanisms of TKIs resistance in lung cancer patients. MicroRNAs (miRNAs) are involved in regulation of various pathophysiological cellular processes. In the present review, we discussed the miRNAs that have been associated with TKIs responses in lung cancer. MiRNAs mainly exert their role on TKIs response through regulation of Tyrosine Kinase Receptors (TKRs) and down-stream signaling pathways. This review paves the way for introducing a panel of miRNAs for the prediction of TKIs responses in lung cancer patients. Video Abstract
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zanguei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Liu YG, Li J, Nie F, Jin GW. LINC00961 functions as an anti-oncogene in non-small cell lung carcinoma by regulation of miR-3127. Am J Transl Res 2022; 14:888-898. [PMID: 35273692 PMCID: PMC8902543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/15/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND This study set out to explore the regulatory relationship between LINC00961/miR-3127 axis and non-small-cell lung carcinoma (NSCLC), so as to provide a new and effective molecular target for targeted therapy of NSCLC. METHODS RNA-seq and miRNA-seq data of NSCLC and normal samples were obtained from The Cancer Genome Atlas (TCGA) database for analyzing LINC00961 and miR-3127 expression. Eighty-six pairs of clinical NSCLC tissues and adjacent normal tissues as well as NSCLC cell lines were obtained. Measurements of LINK00961 and miR-3127 levels were done using real-time-quantitative polymerase chain reaction (RT-qPCR). Furthermore, LINK00961 and miR-3127 in NSCLC cell were regulated respectively. The NSCLC cell proliferation, invasion and migration were determined with MTT assay, Transwell and wound healing assays, respectively. The levels of invasion- and apoptosis-related proteins were detected using western blots, and the connection of LINC00961 and miR-3127 was identified using dual luciferase reporter (DLR) assay. RESULTS Differential analysis results of TCGA databases identified that LINC00961 was ubiquitously expressed at low levels in NSCLC, while miR-3127 was highly expressed. Similar expression trends of LINC00961 and miR-3127 were observed in clinical NSCLC samples and cell lines. Overexpression of LINC00961 and knockdown of miR-3127 significantly reduced NCI-H1299 cell migration, invasiveness, and multiplication, decreased MMP-2, MMP-9 and Bcl-2 protein levels, and increased E-cadherin, Bax and Caspase-3 protein levels. The DLR assay confirmed that miR-3127 can be targeted by LINC00961. CONCLUSION LINC00961 functions as an anti-oncogene in NSCLC by modulating miR-3127.
Collapse
Affiliation(s)
- Yong-Gang Liu
- Department of Thoracic Oncology, Baotou Cancer HospitalBaotou 014000, Inner Mongolia, China
| | - Jia Li
- Department of Immunization Planning, Baotou Disease Control and Defense CenterBaotou 014000, Inner Mongolia, China
| | - Fang Nie
- Department of Thoracic Oncology, Baotou Cancer HospitalBaotou 014000, Inner Mongolia, China
| | - Gao-Wa Jin
- Department of Oncology, Ordos Central HospitalOrdos 017000, Inner Mongolia, China
| |
Collapse
|
5
|
Lu G, Wu Z, Shang J, Xie Z, Chen C, Zhang C. The effects of metformin on autophagy. Biomed Pharmacother 2021; 137:111286. [PMID: 33524789 DOI: 10.1016/j.biopha.2021.111286] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Metformin is the first-line option for treating newly diagnosed diabetic patients and also involved in other pharmacological actions, including antitumor effect, anti-aging effect, polycystic ovarian syndrome prevention, cardiovascular action, and neuroprotective effect, etc. However, the mechanisms of metformin actions were not fully illuminated. Recently, increasing researches showed that autophagy is a vital medium of metformin playing pharmacological actions. Nevertheless, results on the effects of metformin on autophagy were inconsistent. Apart from few clinical evidences, more data focused on kinds of no-clinical models. First, many studies showed that metformin could induce autophagy via a number of signaling pathways, including AMPK-related signaling pathways (e.g. AMPK/mTOR, AMPK/CEBPD, MiTF/TFE, AMPK/ULK1, and AMPK/miR-221), Redd1/mTOR, STAT, SIRT, Na+/H+ exchangers, MAPK/ERK, PK2/PKR/AKT/ GSK3β, and TRIB3. Secondly, some signaling pathways were involved in the process of metformin inhibiting autophagy, such as AMPK-related signaling pathways (AMPK/NF-κB and other undetermined AMPK-related signaling pathways), Hedgehog, miR-570-3p, miR-142-3p, and MiR-3127-5p. Thirdly, two types of signaling pathways including PI3K/AKT/mTOR and endoplasmic reticulum (ER) stress could bidirectionally impact the effectiveness of metformin on autophagy. Finally, multiple signal pathways were reviewed collectively in terms of affecting the effectiveness of metformin on autophagy. The pharmacological effects of metformin combining its actions on autophagy were also discussed. It would help better apply metformin to treat diseases in term of mediating autophagy.
Collapse
Affiliation(s)
- Guangli Lu
- School of Business, Henan University, Henan, Kaifeng, China
| | - Zhen Wu
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Henan, Kaifeng, China
| | - Jia Shang
- School of Kaifeng Culture and Tourism, Henan, Kaifeng, China
| | - Zhenxing Xie
- School of Basic Medicine, Henan University, Henan, Kaifeng, Jinming Avenue, 475004, China.
| | - Chaoran Chen
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Henan, Kaifeng, China.
| | - Chuning Zhang
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Henan, Kaifeng, China
| |
Collapse
|
6
|
Timely Inhibitory Circuit Formation Controlled by Abl1 Regulates Innate Olfactory Behaviors in Mouse. Cell Rep 2021; 30:187-201.e4. [PMID: 31914386 DOI: 10.1016/j.celrep.2019.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 10/16/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022] Open
Abstract
More than one-half of the interneurons in a mouse olfactory bulb (OB) develop during the first week after birth and predominantly connect to excitatory tufted cells near the superficial granule cell layer (sGCL), unlike late-born interneurons. However, the molecular mechanisms underlying the temporal specification are yet to be identified. In this study, we determined the role of Abelson tyrosine-protein kinase 1 (Abl1) in the temporal development of early-born OB interneurons. Lentiviral knockdown of Abl1 disrupts the sGCL circuit of early-born interneurons through defects in function and circuit integration, resulting in olfactory hyper-sensitivity. We show that doublecortin (Dcx) is phosphorylated by Abl1, which contributes to the stabilization of Dcx, thereby regulating microtubule dynamics. Finally, Dcx overexpression rescues Abl1 knockdown-induced anatomic or functional defects. In summary, specific signaling by Abl1-Dcx in early-born interneurons facilitates the temporal development of the sGCL circuit to regulate innate olfactory functions, such as detection and sensitivity.
Collapse
|
7
|
Participation of MicroRNAs in the Treatment of Cancer with Phytochemicals. Molecules 2020; 25:molecules25204701. [PMID: 33066509 PMCID: PMC7587345 DOI: 10.3390/molecules25204701] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a global health concern and one of the main causes of disease-related death. Even with considerable progress in investigations on cancer therapy, effective anti-cancer agents and regimens have thus far been insufficient. There has been compelling evidence that natural phytochemicals and their derivatives have potent anti-cancer activities. Plant-based anti-cancer agents, such as etoposide, irinotecan, paclitaxel, and vincristine, are currently being applied in medical treatments for patients with cancer. Further, the efficacy of plenty of phytochemicals has been evaluated to discover a promising candidate for cancer therapy. For developing more effective cancer therapy, it is required to apprehend the molecular mechanism deployed by natural compounds. MicroRNAs (miRNAs) have been realized to play a pivotal role in regulating cellular signaling pathways, affecting the efficacy of therapeutic agents in cancer. This review presents a feature of phytochemicals with anti-cancer activity, focusing mainly on the relationship between phytochemicals and miRNAs, with insights into the role of miRNAs as the mediators and the regulators of anti-cancer effects of phytochemicals.
Collapse
|
8
|
Yu Z, Wang Y, Wang B, Zhai J. Metformin Affects Paclitaxel Sensitivity of Ovarian Cancer Cells Through Autophagy Mediated by Long Noncoding RNASNHG7/miR-3127-5p Axis. Cancer Biother Radiopharm 2020; 37:792-801. [PMID: 32522016 DOI: 10.1089/cbr.2019.3390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Ovarian cancer is the public health issue worldwide. Paclitaxel is a first-line chemotherapy drug for ovarian cancer, but the paclitaxel resistance weakens the therapeutic effect. Metformin (Met) improved the paclitaxel sensitivity in a mouse model of ovarian cancer. However, the mechanism of Met on paclitaxel sensitivity is still unclear in ovarian cancer. Methods: Cell viability, apoptosis, migration, and invasion were measured by Cell Counting Kit-8 (CCK8), flow cytometry, and transwell assays severally. The expression of long noncoding RNA (lncRNA) small nucleolar RNA host gene 7 (SNHG7) and microRNA-3127-5p (miR-3127-5p) were detected by real-time quantitative polymerase chain reaction. The protein levels of poly (ADP-ribose) polymerase, microtubule-associated protein 1 light chain 3 (LC3)-I, LC3-II, and Beclin 1 were examined by western blot assay. RNA immunoprecipitation assay detected the relationship between SNHG7 and miR-3127-5p. Then, the binding correlation between SNHG7 and miR-3127-5p was predicted by starBase and verified by the dual-luciferase reporter. The effects of Met and SNHG7 on tumor growth were tested in ovarian cancer mice model. Results: Met inhibited cell viability, migration, invasion, SNHG7 level, and autophagy and promoted apoptosis in paclitaxel-resistant ovarian cancer cells. Moreover, Met partly reversed SNHG7-mediated paclitaxel sensitivity and autophagy in ovarian cancer cells. SNHG7 directly bound to miR-3127-5p. Met abolished the promoting effect of SNHG7 overexpression on tumor growth and autophagy in vivo. Conclusion: The authors' findings indicated that Met expedited paclitaxel sensitivity by regulating SNHG7/miR-3127-5p-mediated autophagy in ovarian cancer cells.
Collapse
Affiliation(s)
- Ze Yu
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Yuezhen Wang
- Department of Oncology, The Center Hospital of Zaozhuang Mining Group, Zaozhuang, China
| | - Bin Wang
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Tengzhou, China
| | - Junwei Zhai
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Tengzhou, China
| |
Collapse
|
9
|
Huang H, Huang J, Yao J, Li N, Yang Z. miR‐125a regulates HAS1 and inhibits the proliferation, invasion and metastasis by targeting STAT3 in non–small cell lung cancer cells. J Cell Biochem 2020; 121:3197-3207. [PMID: 31930562 DOI: 10.1002/jcb.29586] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Hu Huang
- Cancer Center, Daping Hospital and Research Institute of SurgeryThird Military Medical University Chongqing P.R. China
- Department of Oncologythe 161 Hospital of PLA Wuhan Hubei P.R. China
| | - Jingyu Huang
- Department of Thoracic SurgeryZhongnan Hospital of Wuhan University Wuhan Hubei P.R. China
| | - Jie Yao
- Department of Biological RepositoriesZhongnan Hospital of Wuhan University Wuhan Hubei P.R. China
| | - Na Li
- Corps of Law Enforcement and SupervisionHealth and Family Planning Commission of Wuhan Municipality Wuhan Hubei P.R. China
| | - Zhenzhou Yang
- Cancer Center, Daping Hospital and Research Institute of SurgeryThird Military Medical University Chongqing P.R. China
| |
Collapse
|
10
|
Díaz-Valdivia NI, Díaz J, Contreras P, Campos A, Rojas-Celis V, Burgos-Ravanal RA, Lobos-González L, Torres VA, Perez VI, Frei B, Leyton L, Quest AFG. The non-receptor tyrosine phosphatase type 14 blocks caveolin-1-enhanced cancer cell metastasis. Oncogene 2020; 39:3693-3709. [PMID: 32152405 PMCID: PMC7190567 DOI: 10.1038/s41388-020-1242-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 01/20/2020] [Accepted: 02/21/2020] [Indexed: 01/13/2023]
Abstract
Caveolin-1 (CAV1) enhanced migration, invasion, and metastasis of cancer cells is inhibited by co-expression of the glycoprotein E-cadherin. Although the two proteins form a multiprotein complex that includes β-catenin, it remained unclear how this would contribute to blocking the metastasis promoting function of CAV1. Here, we characterized by mass spectrometry the protein composition of CAV1 immunoprecipitates from B16F10 murine melanoma cells expressing or not E-cadherin. The novel protein tyrosine phosphatase PTPN14 was identified by mass spectrometry analysis exclusively in co-immunoprecipitates of CAV1 with E-cadherin. Interestingly, PTPN14 is implicated in controlling metastasis, but only few known PTPN14 substrates exist. We corroborated by western blotting experiments that PTPN14 and CAV1 co-inmunoprecipitated in the presence of E-cadherin in B16F10 melanoma and other cancer cells. Moreover, the CAV1(Y14F) mutant protein was shown to co-immunoprecipitate with PTPN14 even in the absence of E-cadherin, and overexpression of PTPN14 reduced CAV1 phosphorylation on tyrosine-14, as well as suppressed CAV1-enhanced cell migration, invasion and Rac-1 activation in B16F10, metastatic colon [HT29(US)] and breast cancer (MDA-MB-231) cell lines. Finally, PTPN14 overexpression in B16F10 cells reduced the ability of CAV1 to induce metastasis in vivo. In summary, we identify here CAV1 as a novel substrate for PTPN14 and show that overexpression of this phosphatase suffices to reduce CAV1-induced metastasis.
Collapse
Affiliation(s)
- Natalia I Díaz-Valdivia
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jorge Díaz
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Institute for Research in Dental Science, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Pamela Contreras
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - América Campos
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Fundación Ciencia & Vida, Santiago, Chile
| | - Victoria Rojas-Celis
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Renato A Burgos-Ravanal
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lorena Lobos-González
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Fundación Ciencia & Vida, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Science, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Viviana I Perez
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Balz Frei
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Lisette Leyton
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| | - Andrew F G Quest
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
11
|
Wang X, Meng R, Hu QM. LINC00319-Mediated miR-3127 Repression Enhances Bladder Cancer Progression Through Upregulation of RAP2A. Front Genet 2020; 11:180. [PMID: 32194636 PMCID: PMC7063470 DOI: 10.3389/fgene.2020.00180] [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: 07/06/2019] [Accepted: 02/14/2020] [Indexed: 12/22/2022] Open
Abstract
Recent studies suggested that microRNA-3127 (miR-3127) was dysregulated in multiple tumor types and has important roles in tumorigenesis and cancer progression. However, its biological roles and the mechanisms that regulate its expression in bladder cancer (BCA) remain to be determined. The expression level of miR-3127 was measured in BCA tissues and its cellular functions were examined using both in vitro and in vivo experiments. The interaction between miR-3127 and long non-coding RNA (lncRNA) LINC00319 was explored using RNA immunoprecipitation assay and luciferase reporter assays. We showed that miR-3127 expression was significantly downregulated in human BCA tissues and BCA cell lines. Lower miR-3127 levels were associated with worse survival in BCA patients. The overexpression of miR-3127 impaired BCA cell proliferation and invasion, and the knockdown of miR-3127 enhanced BCA cell proliferation and invasion in vitro. Importantly, miR-3127 was able to suppress cell growth in vivo. We demonstrated that miR-3127 repressed the proliferation and invasion of BCA cells though directly targeted the 3′-UTR of RAP2A, which served as a novel oncogene in BCA cells. The suppression of cell proliferation and invasion caused by miR-3127 overexpression could be partially abrogated by ectopic expression of RAP2A. Furthermore, high expression of LINC00319 was correlated with adverse survival in BCA patients. LINC00319 could bind directly with miR-3127 and inhibited its expression, and the tumor-promoting effects of LINC00319 could be reversed by re-expression of miR-3127 in BCA cells. Our findings indicated that lncRNA LINC00319-mediated miR-3127 repression promotes BCA progression through the upregulation of RAP2A. The re-introduction of miR-3127 or inhibition of LINC00319 might represent a promising therapeutic strategy for BCA treatment.
Collapse
Affiliation(s)
- Xiaoqing Wang
- Department of Operation Room, Shangqiu First People's Hospital of Henan, Shangqiu, China
| | - Ran Meng
- Department of Urology, Shangqiu First People's Hospital of Henan, Shangqiu, China
| | - Qing-Mei Hu
- Department of Operation Room, Shangqiu First People's Hospital of Henan, Shangqiu, China
| |
Collapse
|
12
|
Zhang Y, Chen X. lncRNA FOXD2-AS1 affects trophoblast cell proliferation, invasion and migration through targeting miRNA. ZYGOTE 2020; 28:1-8. [PMID: 31928563 DOI: 10.1017/s0967199419000807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The abnormal expression of lncRNAs and miRNAs has been found in the placentas of patients with preeclampsia (PE). Therefore, we determined the role of lncRNA FOXD2-AS1/miR-3127 in trophoblast cells. The expression of lncRNA FOXD2-AS1 was detected by qRT-PCR. The proliferation, migration and invasion ability of trophoblast cells were evaluated using CCK-8, wound healing and transwell assays. The target gene of lncRNA FOXD2-AS1 was determined by StarBase and luciferase reporter assays. Western blotting was used to analyze the expression of matrix metalloproteinase 2 (MMP2) and matrix metalloproteinase 9 (MMP9). The results showed that FOXD2-AS1 affected trophoblast cell viability in vitro, while the expression of miR-3127 was decreased. FOXD2-AS1 silencing decreased the promotion effects on trophoblast cell induced by miR-3127 inhibition. In addition, FOXD2-AS1 and miR-3127 presented the same effect on MMP2 and MMP9 levels. lncRNA FOXD2-AS1 modulated trophoblast cell proliferation, invasion and migration through downregulating miR-3127 expression. Therefore, lncRNA FOXD2-AS1 could act as a latent therapeutic marker in preeclampsia.
Collapse
Affiliation(s)
| | - Xiaoqin Chen
- Obstetrics Department, Huai'an First People's Hospital, Huaian, Jiangsu, 223300, China
| |
Collapse
|
13
|
Vishnubalaji R, Shaath H, Elango R, Alajez NM. Noncoding RNAs as potential mediators of resistance to cancer immunotherapy. Semin Cancer Biol 2019; 65:65-79. [PMID: 31733291 DOI: 10.1016/j.semcancer.2019.11.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/31/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022]
Abstract
Substantial evolution in cancer therapy has been witnessed lately, steering mainly towards immunotherapeutic approaches, replacing or in combination with classical therapies. Whereas the use of various immunotherapy approaches, such as adoptive T cell therapy, genetically-modified T cells, or immune checkpoint inhibitors, has been a triumph for cancer immunotherapy, the great challenge is the ability of the immune system to sustain long lasting anti-tumor response. Additionally, epigenetic changes in a suppressive tumor microenvironment can pertain to T cell exhaustion, limiting their functionality. Noncoding RNAs (ncRNAs) have emerged over the last years as key players in epigenetic regulation. Among those, microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) have been studied extensively for their potential role in regulating tumor immunity through direct regulation of genes involved in immune activation or suppression. In this review, we will provide an overview of contemporary approaches for cancer immunotherapy and will present the current state of knowledge implicating miRNAs and lncRNAs in regulating immune response against human cancer and their potential implications in resistance to cancer immunotherapy, with main emphasis on immune checkpoints regulation.
Collapse
Affiliation(s)
- Radhakrishnan Vishnubalaji
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Hibah Shaath
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Ramesh Elango
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Nehad M Alajez
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
| |
Collapse
|
14
|
Carrasco P, Inostroza C, Didier M, Godoy M, Holt CL, Tabak J, Loftus A. Optimizing DNA recovery and forensic typing of degraded blood and dental remains using a specialized extraction method, comprehensive qPCR sample characterization, and massively parallel sequencing. Int J Legal Med 2019; 134:79-91. [PMID: 31414202 PMCID: PMC6949324 DOI: 10.1007/s00414-019-02124-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/10/2019] [Indexed: 12/30/2022]
Abstract
Human dental remains encountered in criminal casework evidence, missing person cases, or mass disaster tragedies provide a valuable sample source for DNA typing when suitable soft tissue is unavailable. Using traditional methods, teeth samples can be challenging to process, resulting in low-quantity and/or quality nuclear DNA and insufficient profiles for comparisons. This study examines the performance of a three-part nuclear DNA analysis workflow for teeth samples based on (1) improved dental tissue recovery using the Dental Forensic Kit (DFKMR) (Universidad de los Andes) and DNA extraction with QuickExtract™ FFPE DNA Extraction Kit (Lucigen®), (2) quantification with InnoQuant® HY (InnoGenomics Technologies) for sensitive assessment of total human and male DNA quantity/quality, and (3) massively parallel sequencing for simultaneous genotyping of 231 short tandem repeat (STR) and single-nucleotide polymorphism (SNP) markers with the ForenSeq® DNA Signature Prep Kit (Verogen, Inc.). Initial evaluation of artificially degraded blood samples (n = 10) achieved highly sensitive and informative quantification results with InnoQuant® HY, enabling successful first pass genotyping with the MiSeq FGx® System. Twenty-three STR alleles (out of 85) and 70 identity informative SNP loci (out of 94) were recovered from two pg total long target DNA input (0.86 ng total short target input) and an InnoQuant degradation index (DI) of 460 (severely degraded). The three-part workflow was subsequently applied to teeth samples (dental pulp, root cement tissues; n = 13) with postmortem intervals (PMI) of the teeth ranging from 8 days to approximately 6 months. Informative SNP and STR DNA profiles were obtained, to include 78 STR alleles and 85 identity informative SNP loci typed (of 94 total SNP targets) in a 1 month, four-day PMI root cement sample with one pg total long target DNA input and a DI of 76. These data indicate successful performance of the proposed workflow from degraded DNA from teeth samples.
Collapse
Affiliation(s)
- Patricio Carrasco
- Universidad de los Andes, Mons. Álvaro del Portillo 12.455, Las Condes, Santiago Chile
| | - Carolina Inostroza
- Universidad de los Andes, Mons. Álvaro del Portillo 12.455, Las Condes, Santiago Chile
| | - Meghan Didier
- Verogen, Inc., 11111 Flintkote Avenue, San Diego, CA 92121 USA
| | - Marianela Godoy
- Universidad de los Andes, Mons. Álvaro del Portillo 12.455, Las Condes, Santiago Chile
| | - Cydne L. Holt
- Verogen, Inc., 11111 Flintkote Avenue, San Diego, CA 92121 USA
| | - Jonathan Tabak
- Verogen, Inc., 11111 Flintkote Avenue, San Diego, CA 92121 USA
| | - Andrew Loftus
- InnoGenomics Technologies, LLC, 1441 Canal Street, New Orleans, LA 70112 USA
| |
Collapse
|
15
|
Overexpression of miR-758 inhibited proliferation, migration, invasion, and promoted apoptosis of non-small cell lung cancer cells by negatively regulating HMGB. Biosci Rep 2019; 39:BSR20180855. [PMID: 30446524 PMCID: PMC6340954 DOI: 10.1042/bsr20180855] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/04/2018] [Accepted: 11/15/2018] [Indexed: 12/20/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most fatal types of cancer with significant mortality and morbidity worldwide. MicroRNAs (miRs) have been confirmed to have positive functions in NSCLC. In the present study, we try to explore the role of miR-758 in proliferation, migration, invasion, and apoptosis of NSCLC cells by regulating high-mobility group box (HMGB) 3 (HMGB3.) NSCLC and adjacent tissues were collected. Reverse transcription quantitative PCR (RT-qPCR) was employed to detect expression of miR-758 and HMGB3 in NSCLC and adjacent tissues, in BEAS-2B cells and NSCLC cell lines. The targetted relationship between miR-758 and HMGB3 was identified by dual luciferase reporter gene assay. The effects of miR-758 on proliferation, migration, invasion, cell cycle, and apoptosis of A549 cells. MiR-758 expression was lower in NSCLC tissues, which was opposite to HMGB3 expression. The results also demonstrated that miR-758 can target HMGB3. The cells transfected with miR-758 mimic had decreased HMGB3 expression, proliferation, migration, and invasion, with more arrested cells in G1 phase and increased apoptosis. Our results supported that the overexpression of miR-758 inhibits proliferation, migration, and invasion, and promotes apoptosis of NSCLC cells by negative regulating HMGB2. The present study may provide a novel target for NSCLC treatment.
Collapse
|
16
|
Tripathi R, Liu Z, Plattner R. EnABLing Tumor Growth and Progression: Recent progress in unraveling the functions of ABL kinases in solid tumor cells. CURRENT PHARMACOLOGY REPORTS 2018; 4:367-379. [PMID: 30746323 PMCID: PMC6368175 DOI: 10.1007/s40495-018-0149-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to summarize our current knowledge regarding how ABL family kinases are activated in solid tumors and impact on solid tumor development/progression, with a focus on recent advances in the field. RECENT FINDINGS Although ABL kinases are known drivers of human leukemia, emerging data also implicates the kinases in a large number of solid tumor types where they promote diverse processes such as proliferation, survival, cytoskeletal reorganization, cellular polarity, EMT (epithelial-mesenchymal-transition), metabolic reprogramming, migration, invasion and metastasis via unique signaling pathways. ABL1 and ABL2 appear to have overlapping but also unique roles in driving these processes. In some tumor types, the kinases may act to integrate pro- and anti-proliferative and -invasive signals, and also may serve as a switch during EMT/MET (mesenchymal-epithelial) transitions. CONCLUSIONS Most data indicate that targeting ABL kinases may be effective for reducing tumor growth and preventing metastasis; however, ABL kinases also may have a tumor suppressive role in some tumor types and in some cellular contexts. Understanding the functions of ABL kinases in solid tumors is critical for developing successful clinical trials aimed at targeting ABL kinases for the treatment of solid tumors.
Collapse
Affiliation(s)
- Rakshamani Tripathi
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, Kentucky 40536
| | - Zulong Liu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, Kentucky 40536
| | - Rina Plattner
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, Kentucky 40536
| |
Collapse
|
17
|
Xu L, Du B, Lu Q, Fan X, Tang K, Yang L, Liao W. miR-541 suppresses proliferation and invasion of squamous cell lung carcinoma cell lines via directly targeting high-mobility group AT-hook 2. Cancer Med 2018; 7:2581-2591. [PMID: 29659195 PMCID: PMC6010725 DOI: 10.1002/cam4.1491] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/13/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
An increasing number of studies have demonstrated that micro-ribonucleic acids (miRNAs) are important tumor suppressors during carcinogenesis. However, the function of miRNA-541 (miR-541) in malignancies, especially lung cancer, has not been widely reported. In this study, miR-541 expression was significantly decreased in squamous cell lung carcinoma (SCLC) cancerous tissue and SCLC cell lines. To analyze miR-541 function in SCLC, we overexpressed miR-541 in SCLC cell lines (SK-MES-1 and H226). According to the CCK8, wound scratch, and transwell invasion assay results, miR-541 overexpression significantly inhibited SCLC cell proliferation, migration, and invasion ability. Next, using RT-PCR, Western blotting, immunocytochemistry, and luciferase assays, HMGA2 was identified, for the first time, as a direct regulatory target of miR-541 in SK-MES-1 and H226 cells. Furthermore, upregulating HMGA2 expression significantly alleviated the suppressive effects of miR-541 on SK-MES-1 and H226 cell proliferation, migration, and invasion. In summary, our study revealed that miR-541 inhibited SCLC proliferation and invasion by directly targeting HMGA2.
Collapse
Affiliation(s)
- Li Xu
- Department of Thoracic SurgeryShanghai Pulmonary HospitalTongji University School of MedicineShanghai200433China
| | - Bin Du
- Department of Thoracic SurgeryChengdu Military General HospitalChengdu610083SichuanChina
| | - Qi‐Jue Lu
- Department of Thoracic SurgeryChanghai HospitalSecond Military Medical UniversityShanghai200438China
| | - Xiao‐Wen Fan
- Department of Thoracic SurgeryChengdu Military General HospitalChengdu610083SichuanChina
| | - Ke Tang
- Department of Thoracic SurgeryChengdu Military General HospitalChengdu610083SichuanChina
| | - Lie Yang
- Department of Thoracic SurgeryChengdu Military General HospitalChengdu610083SichuanChina
| | - Wei‐Lin Liao
- Department of Thoracic SurgeryChengdu Military General HospitalChengdu610083SichuanChina
| |
Collapse
|
18
|
Tang D, Zhao D, Wu Y, Yao R, Zhou L, Lu L, Gao W, Sun Y. The miR-3127-5p/p-STAT3 axis up-regulates PD-L1 inducing chemoresistance in non-small-cell lung cancer. J Cell Mol Med 2018; 22:3847-3856. [PMID: 29726585 PMCID: PMC6050495 DOI: 10.1111/jcmm.13657] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/27/2018] [Indexed: 01/15/2023] Open
Abstract
It is less known about miRNA3127‐5p induced up‐regulation of PD‐L1, immune escape and drug resistance caused by increased PD‐L1 in lung cancer. In this study, lentivirus was transduced into lung cancer cells, and quantitative PCR and Western blot were used to detect the expression of PD‐L1. Then immunofluorescence assay was applied to detect autophagy, finally we explored the relationship between PD‐L1 expressions and chemoresistance in patients. As a result, we found that microRNA‐3127‐5p promotes pSTAT3 to induce the expression of PD‐L1; microRNA‐3127‐5p promotes STAT3 phosphorylation through suppressing autophagy, and autophagy could retaine pSTAT3 into the nucleus in miRNA‐3127‐5p knocked cells, and immune escape induced by elevated level of PD‐L1 results in chemoresistance of lung cancer. In conclusion, microRNA‐3127‐5p induces PD‐L1 elevation through regulating pSTAT3 expression. We also demonstrate that immune escape induced by PD‐L1 can be dismissed by corresponding monoclonal antibody.
Collapse
Affiliation(s)
- Dongfang Tang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, HuaDong Hospital Affiliated with FuDan University, Shanghai, China
| | - Dandan Zhao
- Central Laboratory of Shanghai Chest Hospital Affiliated with Shanghai Jiaotong University, Shanghai, China
| | - Yun Wu
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, HuaDong Hospital Affiliated with FuDan University, Shanghai, China
| | - Ruyong Yao
- Central laboratory of the Affiliated Hospital of Qingdao University, Shanghai, China
| | - Lin Zhou
- Central Laboratory of Shanghai Chest Hospital Affiliated with Shanghai Jiaotong University, Shanghai, China
| | - Liming Lu
- Central Laboratory of Shanghai Chest Hospital Affiliated with Shanghai Jiaotong University, Shanghai, China
| | - Wen Gao
- Department of Thoracic Surgery, Shanghai Key Laboratory of Clinical Geriatric Medicine, HuaDong Hospital Affiliated with FuDan University, Shanghai, China
| | - Yifeng Sun
- Department of Thoracic Surgery, Shanghai Chest Hospital Affiliated with Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
19
|
Yang Y, Sun Y, Wu Y, Tang D, Ding X, Xu W, Su B, Gao W. Downregulation of miR-3127-5p promotes epithelial-mesenchymal transition via FZD4 regulation of Wnt/β-catenin signaling in non-small-cell lung cancer. Mol Carcinog 2018; 57:842-853. [PMID: 29566281 DOI: 10.1002/mc.22805] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 03/11/2018] [Accepted: 03/20/2018] [Indexed: 12/31/2022]
Abstract
MiR-3127-5p has been implicated as a tumor-suppressive microRNA (miRNA) in non-small-cell lung cancer (NSCLC) and its expression was associated with tumor recurrence and poor prognosis. The aim of this study was to determine whether miR-3127-5p regulates epithelial-mesenchymal transition (EMT) in NSCLC, and to investigate the underlying mechanisms. Using qRT-PCR, we examined the expression levels of miR-3127-5p in a cohort of primary NSCLC specimens with and without distant metastasis. We further performed a series of in vitro and in vivo experiments to investigate the effects and underlying mechanism of miR-3127-5p on EMT, cell migration, invasion, and adhesion in NSCLC. We found that metastatic NSCLC tissues showed markedly downregulated miR-3127-5p expression. Transforming growth factor-β1 (TGF-β1) treatment induced EMT in A549 and H1299 cells, and downregulation of miR-3127-5p could result in the similar effect. Mechanically, we demonstrated that frizzled-4 (FZD4) is a target gene and miR-3127-5p exerts its effects by regulating the Wnt/β-catenin signaling. In addition, the expression levels of FZD4 and miR-3127-5p were also negatively associated in both clinical and xenografted tumors. Overall, these findings suggest that downregulation of miR-3127-5p promotes EMT through activating the Wnt/FZD4/β-catenin signaling pathway and may represent a therapeutic target for NSCLC metastasis.
Collapse
Affiliation(s)
- Yang Yang
- Department of Thoracic Surgery, Huadong Hospital Affiliated to FuDan University, Shanghai, China.,Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yifeng Sun
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Wu
- Department of Thoracic Surgery, Huadong Hospital Affiliated to FuDan University, Shanghai, China
| | - Dongfang Tang
- Department of Thoracic Surgery, Huadong Hospital Affiliated to FuDan University, Shanghai, China
| | - Xi Ding
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen Xu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bo Su
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen Gao
- Department of Thoracic Surgery, Huadong Hospital Affiliated to FuDan University, Shanghai, China
| |
Collapse
|
20
|
Yasui T, Yanagida T, Ito S, Konakade Y, Takeshita D, Naganawa T, Nagashima K, Shimada T, Kaji N, Nakamura Y, Thiodorus IA, He Y, Rahong S, Kanai M, Yukawa H, Ochiya T, Kawai T, Baba Y. Unveiling massive numbers of cancer-related urinary-microRNA candidates via nanowires. SCIENCE ADVANCES 2017; 3:e1701133. [PMID: 29291244 PMCID: PMC5744465 DOI: 10.1126/sciadv.1701133] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 11/16/2017] [Indexed: 05/06/2023]
Abstract
Analyzing microRNAs (miRNAs) within urine extracellular vesicles (EVs) is important for realizing miRNA-based, simple, and noninvasive early disease diagnoses and timely medical checkups. However, the inherent difficulty in collecting dilute concentrations of EVs (<0.01 volume %) from urine has hindered the development of these diagnoses and medical checkups. We propose a device composed of nanowires anchored into a microfluidic substrate. This device enables EV collections at high efficiency and in situ extractions of various miRNAs of different sequences (around 1000 types) that significantly exceed the number of species being extracted by the conventional ultracentrifugation method. The mechanical stability of nanowires anchored into substrates during buffer flow and the electrostatic collection of EVs onto the nanowires are the two key mechanisms that ensure the success of the proposed device. In addition, we use our methodology to identify urinary miRNAs that could potentially serve as biomarkers for cancer not only for urologic malignancies (bladder and prostate) but also for nonurologic ones (lung, pancreas, and liver). The present device concept will provide a foundation for work toward the long-term goal of urine-based early diagnoses and medical checkups for cancer.
Collapse
Affiliation(s)
- Takao Yasui
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- ImPACT Research Center for Advanced Nanobiodevices, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Japan Science and Technology Agency, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- Corresponding author. (T. Yasui); (T. Yanagida); (T.K.); (Y.B.)
| | - Takeshi Yanagida
- Institute of Materials Chemistry and Engineering, Kyushu University, 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
- Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka-cho, Ibaraki, Osaka 567-0047, Japan
- Corresponding author. (T. Yasui); (T. Yanagida); (T.K.); (Y.B.)
| | - Satoru Ito
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Yuki Konakade
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Daiki Takeshita
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Tsuyoshi Naganawa
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Kazuki Nagashima
- Institute of Materials Chemistry and Engineering, Kyushu University, 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
| | - Taisuke Shimada
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Noritada Kaji
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- ImPACT Research Center for Advanced Nanobiodevices, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Japan Science and Technology Agency, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Yuta Nakamura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Ivan Adiyasa Thiodorus
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Yong He
- Institute of Materials Chemistry and Engineering, Kyushu University, 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
| | - Sakon Rahong
- ImPACT Research Center for Advanced Nanobiodevices, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- College of Nanotechnology, King Mongkut’s Institute of Technology Ladkrabang, Chalongkrung Road, Ladkrabang, Bangkok 10520, Thailand
| | - Masaki Kanai
- Institute of Materials Chemistry and Engineering, Kyushu University, 6-1 Kasuga-koen, Kasuga, Fukuoka 816-8580, Japan
| | - Hiroshi Yukawa
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- ImPACT Research Center for Advanced Nanobiodevices, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Tomoji Kawai
- Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka-cho, Ibaraki, Osaka 567-0047, Japan
- Corresponding author. (T. Yasui); (T. Yanagida); (T.K.); (Y.B.)
| | - Yoshinobu Baba
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- ImPACT Research Center for Advanced Nanobiodevices, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Health Research Institute, National Institute of Advanced Industrial Science and Technology, Takamatsu 761-0395, Japan
- College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan, Republic of China
- Corresponding author. (T. Yasui); (T. Yanagida); (T.K.); (Y.B.)
| |
Collapse
|
21
|
Abstract
Recent studies suggest that neuropilin-1 (NRP-1) promotes angiogenesis mainly via VEGF and its receptors. It promotes tumorigenesis via formation of the NRP-1/ VEGF (vascular endothelial growth factor)/VEGFR2 (vascular endothelial growth factor receptor 2) complex. In addition to VEGF and its receptors, NRP-1 also binds with other growth factors such as platelet-derived growth factor (PDGF) and platelet-derived growth factor receptor (PDGFR). PDGF plays important roles in cellular proliferation and, in particular, blood vessel formation. Moreover, recent studies show that NRP-1 promotes angiogenesis via the NRP-1-ABL pathway, but independent of VEGF-VEGFR2. RAD51 is a protein involved in the signaling pathways of NRP1-ABL and PDGF(R), the expression of which is positively associated with cell radioresistance and chemoresistance. NRP-1 activates the signaling pathways of ABL and PDGF(R) to upregulate RAD51, which induces resistance to radiotherapy and chemotherapy in cancer cells. Furthermore, NRP-1 activates the tumor microenvironment by binding with fibronectin and activating ABL, thereby promoting tumor growth. Inhibition of NRP-1 may overcome the limitations of individually inhibiting the VEGF-VEGFR2 pathway in cancer therapy and provide new ideas for cancer treatment. Therefore, we review the role of NRP-1 in VEGF-VEGFR2-independent tumorigenesis.
Collapse
Affiliation(s)
- Chenxi Hu
- Department of Radiation Oncology, Lianyungang First People's Hospital, No.182, Tongguan Road, Lianyungang City, 222002, Jiangsu Province, China
| | - Xiaodong Jiang
- Department of Radiation Oncology, Lianyungang First People's Hospital, No.182, Tongguan Road, Lianyungang City, 222002, Jiangsu Province, China.
| |
Collapse
|
22
|
Shumayla, Sharma S, Taneja M, Tyagi S, Singh K, Upadhyay SK. Survey of High Throughput RNA-Seq Data Reveals Potential Roles for lncRNAs during Development and Stress Response in Bread Wheat. FRONTIERS IN PLANT SCIENCE 2017; 8:1019. [PMID: 28649263 PMCID: PMC5465302 DOI: 10.3389/fpls.2017.01019] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/29/2017] [Indexed: 09/01/2023]
Abstract
Long non-coding RNAs (lncRNAs) are a family of regulatory RNAs that play essential role in the various developmental processes and stress responses. Recent advances in sequencing technology and computational methods enabled identification and characterization of lncRNAs in certain plant species, but they are less known in Triticum aestivum (bread wheat). Herein, we analyzed 52 RNA seq data (>30 billion reads) and identified 44,698 lncRNAs in T. aestivum genome, which were characterized in comparison to the coding sequences (mRNAs). Similar to the mRNAs, lncRNAs were also derived from each sub-genome and chromosome, and showed tissue developmental stage specific and differential expression, as well. The modulated expression of lncRNAs during abiotic stresses like heat, drought, and salt indicated their putative role in stress response. The co-expression of lncRNAs with vital mRNAs including various transcription factors and enzymes involved in Abscisic acid (ABA) biosynthesis, and gene ontology mapping inferred their regulatory roles in numerous biological processes. A few lncRNAs were predicted as precursor (19 lncRNAs), while some as target mimics (1,047 lncRNAs) of known miRNAs involved in various regulatory functions. The results suggested numerous functions of lncRNAs in T. aestivum, and unfolded the opportunities for functional characterization of individual lncRNA in future studies.
Collapse
Affiliation(s)
- Shumayla
- Department of Botany, Panjab UniversityChandigarh, India
| | | | - Mehak Taneja
- Department of Botany, Panjab UniversityChandigarh, India
| | - Shivi Tyagi
- Department of Botany, Panjab UniversityChandigarh, India
| | - Kashmir Singh
- Department of Biotechnology, Panjab UniversityChandigarh, India
| | | |
Collapse
|
23
|
Li J, Hu K, Gong G, Zhu D, Wang Y, Liu H, Wu X. Upregulation of MiR-205 transcriptionally suppresses SMAD4 and PTEN and contributes to human ovarian cancer progression. Sci Rep 2017; 7:41330. [PMID: 28145479 PMCID: PMC5286557 DOI: 10.1038/srep41330] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/12/2016] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) function as critical regulators of gene expression and their deregulation is associated with the development and progression of various cancers. This study aimed to investigate the biological role and mechanism of miR-205 in ovarian cancer (OC). MiR-205 was upregulated in OC tissues and cells in comparison to the controls. Meanwhile, overexpression of miR-205 was significantly associated with poor overall survival of OC patients. Functional study indicated that ectopic expression of miR-205 significantly promoted cell proliferation, migration, invasion and chemoresistance of OC cells. SMAD4 and PTEN were identified as direct targets of miR-205 using luciferase reporter assays, real-time PCR (qRT-PCR), and western blot. Most interestingly, in vivo studies indicated that miR-205 markedly promoted the growth and metastasis of tumors and the expression of miR-205 was also found to be inversely correlated with that of SMAD4 and PTEN in nude mice. Overall, we suggest that miR-205 functions as an oncogenic miRNA by directly binding to SMAD4 and PTEN, providing a novel target for the molecular treatment of ovarian cancer.
Collapse
Affiliation(s)
- Juanni Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Kuan Hu
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guanghui Gong
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ding Zhu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yixuan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hailing Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xiaoying Wu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
MicroRNA-585 acts as a tumor suppressor in non-small-cell lung cancer by targeting hSMG-1. Clin Transl Oncol 2016; 19:546-552. [PMID: 27743168 DOI: 10.1007/s12094-016-1562-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/06/2016] [Indexed: 10/20/2022]
Abstract
PURPOSE To investigate the role of miR-585 in the development and progression of non-small-cell lung cancer (NSCLC). METHODS The expression levels of miR-585 in NSCLC cell lines and clinical samples were measured by quantitative PCR. NSCLC cells, A549 and H1299, were stably transfected with lentiviral vectors of miR-585 mimics or negative control. The effects of miR-585 on cell proliferation were detected both in vitro and in vivo. Cell migration and invasion were evaluated using wound healing assay and Transwell assay. Furthermore, luciferase reporter assay was used to identify the direct regulation of hSMG-1 by miR-585. RESULTS Our results showed that miR-585 was downregulated in NSCLC cell lines and tumor tissues. Ectopic expression of miR-585 inhibited the ability of cell proliferation, migration, and invasion in vitro. In addition, miR-585 also decreased the growth rate of xenografted tumor in nude mice. Mechanically, miR-585 directly targeted the 3'-untranslated region (UTR) of hSMG-1 gene, which likely resulted in a dysfunction of mRNA surveillance and nonsense-mediated mRNA decay. CONCLUSION Taken together, miR-585 probably has an inhibitory effect on tumor growth and is a prognostic biomarker of NSCLC.
Collapse
|
25
|
Lu YJ, Liu RY, Hu K, Wang Y. MiR-541-3p reverses cancer progression by directly targeting TGIF2 in non-small cell lung cancer. Tumour Biol 2016; 37:12685-12695. [PMID: 27448300 DOI: 10.1007/s13277-016-5241-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022] Open
Abstract
Lung cancer remains a leading cause of cancer-associated mortality worldwide, and non-small lung cancer (NSCLC) is responsible for over 80 % of lung cancer-related deaths. Identifying novel molecular biomarker that can inhibit the progression of lung cancer will facilitate the development of new treatment strategies. Herein, we demonstrated that miR-541-3p is a tumor-suppressor microRNA (miRNA) in NSCLC progression. We found that expression of miR-541-3p was decreased obviously in NSCLC tissues and plasma. Down-regulation of miR-541-3p was associated with TNM stage and postoperative survival. Overexpression of miR-541-3p inhibited the growth and metastasis of NSCLC cells. The TGIF2 was a direct target of miR-541-3p and promoted the growth and metastasis of NSCLC cells. Further study showed that TGIF2 could reverse the inhibitory effect of miR-541-3p on growth and metastasis of NSCLC cells. Taken together, our data highlight the pivotal role of miR-541-3p in the progression of NSCLC. Thus, miR-541-3p may be a potential prognostic marker and of treatment relevance for NSCLC progression intervention.
Collapse
Affiliation(s)
- You-Jin Lu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Rong-Yu Liu
- Department of Geriatric Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Kun Hu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ying Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| |
Collapse
|
26
|
Koufaris C. Human and primate-specific microRNAs in cancer: Evolution, and significance in comparison with more distantly-related research models. Bioessays 2016; 38:286-94. [DOI: 10.1002/bies.201500135] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Costas Koufaris
- Department of Cytogenetics and Genomics; Cyprus institute of Neurology and Genetics; Nicosia Cyprus
| |
Collapse
|
27
|
MiR-145 acts as a metastasis suppressor by targeting metadherin in lung cancer. Med Oncol 2014; 32:344. [PMID: 25428378 DOI: 10.1007/s12032-014-0344-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/09/2014] [Indexed: 01/10/2023]
Abstract
MiR-145 has been reported to be downregulated in multiple tumors. It acts as a tumor suppressor in lung cancer. In this study, we investigated the potential effects of miR-145 on invasion and metastasis and the molecular mechanism in non-small cell lung cancer. MiR-145 was downregulated in the NSCLC specimens and significantly correlated with advanced clinical stage and lymph node metastasis. In addition, AEG-1/MTDH was a direct target of miR-145, and the expression of AEG-1/MTDH was inversely correlated with miR-145 expression in NSCLC tissues. Ectopic expression of miR-145 suppressed cell invasion and metastasis in NSCLC cells. AEG-1/MTDH overexpression partially reversed the suppressive effect of miR-145. These findings provide novel insights with potential therapeutic applications for the treatment of NSCLC.
Collapse
|