1
|
Sweef O, Zaabout E, Bakheet A, Halawa M, Gad I, Akela M, Tousson E, Abdelghany A, Furuta S. Unraveling Therapeutic Opportunities and the Diagnostic Potential of microRNAs for Human Lung Cancer. Pharmaceutics 2023; 15:2061. [PMID: 37631277 PMCID: PMC10459057 DOI: 10.3390/pharmaceutics15082061] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Lung cancer is a major public health problem and a leading cause of cancer-related deaths worldwide. Despite advances in treatment options, the five-year survival rate for lung cancer patients remains low, emphasizing the urgent need for innovative diagnostic and therapeutic strategies. MicroRNAs (miRNAs) have emerged as potential biomarkers and therapeutic targets for lung cancer due to their crucial roles in regulating cell proliferation, differentiation, and apoptosis. For example, miR-34a and miR-150, once delivered to lung cancer via liposomes or nanoparticles, can inhibit tumor growth by downregulating critical cancer promoting genes. Conversely, miR-21 and miR-155, frequently overexpressed in lung cancer, are associated with increased cell proliferation, invasion, and chemotherapy resistance. In this review, we summarize the current knowledge of the roles of miRNAs in lung carcinogenesis, especially those induced by exposure to environmental pollutants, namely, arsenic and benzopyrene, which account for up to 1/10 of lung cancer cases. We then discuss the recent advances in miRNA-based cancer therapeutics and diagnostics. Such information will provide new insights into lung cancer pathogenesis and innovative diagnostic and therapeutic modalities based on miRNAs.
Collapse
Affiliation(s)
- Osama Sweef
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109, USA
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Elsayed Zaabout
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ahmed Bakheet
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109, USA
| | - Mohamed Halawa
- Department of Pharmacology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ibrahim Gad
- Department of Statistics and Mathematics, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Mohamed Akela
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ehab Tousson
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Ashraf Abdelghany
- Biomedical Research Center of University of Granada, Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
| | - Saori Furuta
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44109, USA
| |
Collapse
|
2
|
Sprooten J, Laureano RS, Vanmeerbeek I, Govaerts J, Naulaerts S, Borras DM, Kinget L, Fucíková J, Špíšek R, Jelínková LP, Kepp O, Kroemer G, Krysko DV, Coosemans A, Vaes RD, De Ruysscher D, De Vleeschouwer S, Wauters E, Smits E, Tejpar S, Beuselinck B, Hatse S, Wildiers H, Clement PM, Vandenabeele P, Zitvogel L, Garg AD. Trial watch: chemotherapy-induced immunogenic cell death in oncology. Oncoimmunology 2023; 12:2219591. [PMID: 37284695 PMCID: PMC10240992 DOI: 10.1080/2162402x.2023.2219591] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/25/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
Immunogenic cell death (ICD) refers to an immunologically distinct process of regulated cell death that activates, rather than suppresses, innate and adaptive immune responses. Such responses culminate into T cell-driven immunity against antigens derived from dying cancer cells. The potency of ICD is dependent on the immunogenicity of dying cells as defined by the antigenicity of these cells and their ability to expose immunostimulatory molecules like damage-associated molecular patterns (DAMPs) and cytokines like type I interferons (IFNs). Moreover, it is crucial that the host's immune system can adequately detect the antigenicity and adjuvanticity of these dying cells. Over the years, several well-known chemotherapies have been validated as potent ICD inducers, including (but not limited to) anthracyclines, paclitaxels, and oxaliplatin. Such ICD-inducing chemotherapeutic drugs can serve as important combinatorial partners for anti-cancer immunotherapies against highly immuno-resistant tumors. In this Trial Watch, we describe current trends in the preclinical and clinical integration of ICD-inducing chemotherapy in the existing immuno-oncological paradigms.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Raquel S. Laureano
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Daniel M. Borras
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lisa Kinget
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Jitka Fucíková
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Radek Špíšek
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Lenka Palová Jelínková
- Department of Immunology, Charles University, 2Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio Biotech, Prague, Czech Republic
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Liguecontre le Cancer, Université de Paris, sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée Par la Liguecontre le Cancer, Université de Paris, sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Institut du Cancer Paris CARPEM, Paris, France
| | - Dmitri V. Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Insitute Ghent, Ghent University, Ghent, Belgium
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Rianne D.W. Vaes
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Steven De Vleeschouwer
- Department Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Department Neuroscience, Laboratory for Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Els Wauters
- Laboratory of Respiratory Diseases and Thoracic Surgery (Breathe), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Sabine Tejpar
- Molecular Digestive Oncology, Department of Oncology, Katholiek Universiteit Leuven, Leuven, Belgium
- Cell Death and Inflammation Unit, VIB-Ugent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Benoit Beuselinck
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Paul M. Clement
- Laboratory of Experimental Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Peter Vandenabeele
- Cell Death and Inflammation Unit, VIB-Ugent Center for Inflammation Research (IRC), Ghent, Belgium
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Laurence Zitvogel
- Tumour Immunology and Immunotherapy of Cancer, European Academy of Tumor Immunology, Gustave Roussy Cancer Center, Inserm, Villejuif, France
| | - Abhishek D. Garg
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Arab F, Aghaee Bakhtiari SH, Pasdar A, Saburi E. Evaluation of osteogenic induction potency of miR-27a-3p in adipose tissue-derived human mesenchymal stem cells (AD-hMSCs). Mol Biol Rep 2023; 50:1281-1291. [PMID: 36451000 DOI: 10.1007/s11033-022-08084-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/01/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Bone tissue as a dynamic tissue is able to repair its minor injuries, however, sometimes the repair cannot be completed by itself due to the size of lesion. In such cases, the best treatment could be bone tissue engineering. The use of stem cells in skeletal disorders to repair bone defects has created bright prospects. On the other hand, changes in the expression level of microRNAs (miRs) can lead to the commitment of mesenchymal stem cells (MSCs) to cell lineage. Many studies reported that post-transcriptional regulations by miRNAs are involved in all stages of osteoblast differentiation. METHOD After the preparing adipose tissue-derived mesenchymal stem cells, the target cells from the third passage were cultured in two groups, transfected MSCs with miR-27a-3p (DM.C + P) and control group. In different times, 7 and 14 days after culture, differentiation of these cells into osteoblast were measured using various techniques including the ALP test and calcium content test, Alizarin Red staining, Immunocytochemistry technique (ICC). Also, the relative expression of bone differentiation marker genes including Osteonectin (ON), Osteocalcin (OC), RUNX Family Transcription Factor 2 (RUNX2), Collagen type I alpha 1 (COL1) was investigated by real-time RT PCR. RESULTS In comparison with control groups, overexpression of miR-27a-3p in transfected cells resulted in a significant increase in the expression of bone markers genes (ON, OC, RUNX2, COL1), alkaline phosphatase (ALP) activity, and calcium content (p < 0.05). In addition, the results obtained from ICC technique showed that osteocalcin protein is expressed at the surface of bone cells. Furthermore, the expression of APC, as a target of miR-27a-3p, decreased in transfected cells. CONCLUSION Our data suggest that miR-27a-3p may positively regulates adipose tissue-derived mesenchymal stem cell differentiation into bone by targeting APC and activating the Wnt/b-catenin pathway.
Collapse
Affiliation(s)
- Fatemeh Arab
- Department of Medical Genetics and Molecular Medicine Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hamid Aghaee Bakhtiari
- Assistant Professor of Medical Biotechnology, Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Pasdar
- Department of Medical Genetics and Molecular Medicine Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Banerjee S, Mandal AKA. Role of epigallocatechin-3- gallate in the regulation of known and novel microRNAs in breast carcinoma cells. Front Genet 2022; 13:995046. [PMID: 36276982 PMCID: PMC9582282 DOI: 10.3389/fgene.2022.995046] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022] Open
Abstract
Breast cancer comprises 30% of all cancer cases among the world’s women population. MicroRNAs are small, endogenous, non-coding RNAs that regulate cell proliferating and apoptotic pathways by modulating expressions of related genes. Phytochemicals like epigallocatechin-3-gallate (EGCG) are known to have a chemotherapeutic effect on cancer often through the regulation of microRNAs. The aim is to find out the key known and novel miRNAs, which are controlled by EGCG in breast cancer cell line MDA-MB-231. Next-generation sequencing (NGS) revealed 1,258 known and 330 novel miRNAs from untreated and 83 μM EGCG (IC50 value of EGCG) treated cells. EGCG modulated 873 known and 47 novel miRNAs in the control vs. treated sample. The hypothesis of EGCG being a great modulator of miRNAs that significantly control important cancer-causing pathways has been established by analyzing with Kyoto Encyclopedia of Genes and Genomes (KEGG) and Protein Analysis Through Evolutionary Relationships (PANTHER) database. Validation of known and novel miRNA expression differences in untreated vs. treated cells was done using qPCR. From this study, a few notable miRNAs were distinguished that can be used as diagnostics as well as prognostic markers for breast cancer.
Collapse
|
5
|
Bernardes JGB, Fernandes MR, Rodrigues JCG, Vinagre LWMS, Pastana LF, Dobbin EAF, Medeiros JAG, Dias Junior LB, Bernardes GM, Bernardes IMM, Santos NPCD, Demachki S, Burbano RMR. Association of Androgenic Regulation and MicroRNAs in Acinar Adenocarcinoma of Prostate. Genes (Basel) 2022; 13:genes13040622. [PMID: 35456428 PMCID: PMC9030213 DOI: 10.3390/genes13040622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 12/13/2022] Open
Abstract
Background: Prostate cancer represents 3.8% of cancer deaths worldwide. For most prostate cancer cells to grow, androgens need to bind to a cellular protein called the androgen receptor (AR). This study aims to demonstrate the expression of five microRNAs (miRs) and its influence on the AR formation in patients from the northern region of Brazil. Material and Methods: Eighty-four tissue samples were investigated, including nodular prostatic hyperplasia (NPH) and acinar prostatic adenocarcinoma (CaP). Five miRs (27a-3p, 124, 130a, 488-3p, and 506) were quantified using the TaqMan® Real Time PCR method and AR was measured using Western blotting. Results: Levels of miRs 124, 130a, 488-3p, and 506 were higher in NPH samples. Conversely, in the CaP cases, higher levels of miR 27a-3p and AR were observed. Conclusion: In the future, these microRNAs may be tested as markers of CaP at the serum level. The relative expression of AR was 20% higher in patients with prostate cancer, which suggests its potential as a biomarker for prostate malignancy.
Collapse
Affiliation(s)
- Julio Guilherme Balieiro Bernardes
- Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66050-160, Brazil; (J.G.B.B.); (L.B.D.J.); (I.M.M.B.)
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
| | - Marianne Rodrigues Fernandes
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
- Hospital Ophir Loyola, Belém 66063-240, Brazil
- Correspondence:
| | - Juliana Carla Gomes Rodrigues
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
| | - Lui Wallacy Morikawa Souza Vinagre
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
| | - Lucas Favacho Pastana
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
| | - Elizabeth Ayres Fragoso Dobbin
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
| | - Jéssyca Amanda Gomes Medeiros
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
| | - Leonidas Braga Dias Junior
- Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém 66050-160, Brazil; (J.G.B.B.); (L.B.D.J.); (I.M.M.B.)
| | | | | | - Ney Pereira Carneiro Dos Santos
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
| | - Samia Demachki
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
| | - Rommel Mario Rodriguez Burbano
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.C.G.R.); (L.W.M.S.V.); (L.F.P.); (E.A.F.D.); (J.A.G.M.); (N.P.C.D.S.); (S.D.); (R.M.R.B.)
- Hospital Ophir Loyola, Belém 66063-240, Brazil
| |
Collapse
|
6
|
Lu M, Li J, Fan X, Xie F, Fan J, Xiong Y. Novel Immune-Related Ferroptosis Signature in Esophageal Cancer: An Informatics Exploration of Biological Processes Related to the TMEM161B-AS1/hsa-miR-27a-3p/GCH1 Regulatory Network. Front Genet 2022; 13:829384. [PMID: 35281840 PMCID: PMC8908453 DOI: 10.3389/fgene.2022.829384] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Considering the role of immunity and ferroptosis in the invasion, proliferation and treatment of cancer, it is of interest to construct a model of prognostic-related differential expressed immune-related ferroptosis genes (PR-DE-IRFeGs), and explore the ferroptosis-related biological processes in esophageal cancer (ESCA).Methods: Four ESCA datasets were used to identify three PR-DE-IRFeGs for constructing the prognostic model. Validation of our model was based on analyses of internal and external data sets, and comparisons with past models. With the biological-based enrichment analysis as a guide, exploration for ESCA-related biological processes was undertaken with respect to the immune microenvironment, mutations, competing endogenous RNAs (ceRNA), and copy number variation (CNV). The model’s clinical applicability was measured by nomogram and correlation analysis between risk score and gene expression, and also immune-based and chemotherapeutic sensitivity.Results: Three PR-DE-IRFeGs (DDIT3, SLC2A3, and GCH1), risk factors for prognosis of ESCA patients, were the basis for constructing the prognostic model. Validation of our model shows a meaningful capability for prognosis prediction. Furthermore, many biological functions and pathways related to immunity and ferroptosis were enriched in the high-risk group, and the role of the TMEM161B-AS1/hsa-miR-27a-3p/GCH1 network in ESCA is supported. Also, the KMT2D mutation is associated with our risk score and SLC2A3 expression. Overall, the prognostic model was associated with treatment sensitivity and levels of gene expression.Conclusion: A novel, prognostic model was shown to have high predictive value. Biological processes related to immune functions, KMT2D mutation, CNV and the TMEM161B-AS1/hsa-miR-27a-3p/GCH1 network were involved in ESCA progression.
Collapse
Affiliation(s)
- Min Lu
- Department of Emergency, Shangrao People’s Hospital, Shangrao Hospital Affiliated to Nanchang University, Shangrao, China
| | - Jiaqi Li
- School of Stomatology, Nanchang University, Nanchang, China
| | - Xin Fan
- School of Stomatology, Nanchang University, Nanchang, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xin Fan,
| | - Fei Xie
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jie Fan
- Shangrao Municipal Hospital, Shangrao, China
| | - Yuanping Xiong
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
7
|
miRNA-27a Transcription Activated by c-Fos Regulates Myocardial Ischemia-Reperfusion Injury by Targeting ATAD3a. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2514947. [PMID: 34413925 PMCID: PMC8369174 DOI: 10.1155/2021/2514947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022]
Abstract
MicroRNA-27a (miR-27a) has been implicated in myocardial ischemia-reperfusion injury (MIRI), but the underlying mechanism is not well understood. This study is aimed at determining the role of miR-27a in MIRI and at investigating upstream molecules that regulate miR-27a expression and its downstream target genes. miR-27a expression was significantly upregulated in myocardia exposed to ischemia/reperfusion (I/R) and cardiomyocytes exposed to hypoxia/reoxygenation (H/R). c-Fos could regulate miR-27a expression by binding to its promoter region. Moreover, overexpression of miR-27a led to a decrease in cell viability, an increase in LDH and CK-MB secretion, and an increase in apoptosis rates. In contrast, suppression of miR-27a expression resulted in the opposite effects. ATPase family AAA-domain-containing protein 3A (ATAD3a) was identified as a target of miR-27a. miR-27a regulated the translocation of apoptosis-inducing factor (AIF) from the mitochondria to the nucleus and H/R-induced apoptosis via the regulation of ATAD3a. It was found that inhibiting miR-27a in vivo by injecting a miR-27a sponge could ameliorate MIRI in an isolated rat heart model. In conclusion, our study demonstrated that c-Fos functions as an upstream regulator of miR-27a and that miR-27a regulates the translocation of AIF from the mitochondria to the nucleus by targeting ATAD3a, thereby contributing to MIRI. These findings provide new insight into the role of the c-Fos/miR-27a/ATAD3a axis in MIRI.
Collapse
|
8
|
HPV16 E6 enhances the radiosensitivity in HPV-positive human head and neck squamous cell carcinoma by regulating the miR-27a-3p/SMG1 axis. Infect Agent Cancer 2021; 16:56. [PMID: 34389030 PMCID: PMC8361787 DOI: 10.1186/s13027-021-00397-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/29/2021] [Indexed: 01/13/2023] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is the 6th most common malignant cancer type worldwide. Radiosensitivity has been shown to be significantly increased in patients with human papillomavirus (HPV)-positive HNSCC compared with HPV-negative patients. However, the clinical significance of HPV and its regulatory mechanisms in HNSCC are largely unknown. The aim of our study was to explore the regulatory mechanism of miR-27a-3p in the radiosensitivity of HPV-positive HNSCC cells. Methods E6-overexpressing and E6-knockdown HNSCC cell lines were generated and the transfection efficiencies were evaluated by quantitative real-time PCR (RT-qPCR) and western blotting. The expression of miR-27a-3p and DiGeorge syndrome critical region 8 (DGCR8) was examined by RT-qPCR after transfection with E6 overexpressing plasmid or E6 siRNA. The effects of miR-27a-3p on the radiosensitivity of HNSCC cells were explored by a colony formation and TUNEL staining assays. Bioinformatic tools and luciferase reporter assays were used to identify that SMG1 is the direct target of miR-27a-3p. Furthermore, the effect of E6 overexpression on the regulation of the miR-27a-3p/SMG1 axis was investigated. Results In our study, we found overexpression of HPV E6 upregulated the expression of DGCR8 and miR-27a-3p in HNSCC cells. We next confirmed that DGCR8 positively regulated the expression of miR-27a-3p in HNSCC cells. The luciferase reporter gene results verified that miR-27a-3p targeted the 3’UTR of SMG1 mRNA. MiR-27a-3p mimics transfection resulted in a decrease in SMG1 expression and miR-27a-3p inhibitor transfection increased SMG1 expression. Apoptotic activity of HNSCC cells was significantly increased in miR-27a-3p mimics HNSCC cells compared with control HNSCC cells. After treatment with 4 Gy irradiation, UM-SCC47 cells transfected with miR-27a-3p inhibitor or SMG1 overexpressing plasmid formed more colonies than the corresponding control cells. Furthermore, the rescue experiments demonstrated that HPV16 E6 improved the radiosensitivity of HNSCC cells by targeting miR-27a-3p/SMG1. Conclusion Our study demonstrated that HPV16 E6 activated the DGCR8/miR-27a-3p/SMG1 axis to enhance the radiosensitivity. Our findings might provide a novel therapeutic target to improve the response of HNSCC to radiotherapy.
Collapse
|
9
|
Zhang Y, Zhan B, Hu Y, Chen S, Zhang Q. Sevoflurane inhibits the apoptosis of hypoxia/reoxygenation-induced cardiomyocytes via regulating miR-27a-3p-mediated autophagy. J Pharm Pharmacol 2021; 73:1470-1479. [PMID: 34383044 DOI: 10.1093/jpp/rgab111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 07/14/2021] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Sevoflurane (Sevo) prevents hypoxia/reoxygenation (H/R)-induced cardiomyocytes apoptosis. MiR-27a-3p expression is up-regulated in Sevo-treated hippocampal neurons. OBJECTIVE This study explored whether the effect of Sevo on cardiomyocytes was mediated by miR-27a-3p. METHODS The cardiomyocytes were cultured under H/R condition or pre-treated with Sevo, and further transfected with miR-27a-3p inhibitor or treated with an autophagy inhibitor 3-methyladenine (3-MA). Then the cell morphology was observed under an optical microscope. The cell viability and apoptosis were measured by MTT and flow cytometry. Expressions of miR-27a-3p, apoptosis-related, and autophagy-related factors were determined by western blot or RT-qPCR. KEY FINDINGS Sevo improved the abnormal morphology, promoted the cell viability and the expressions of Bcl-2 and miR-27a-3p, but reduced the apoptosis and Bax and C-caspase-3 levels of H/R-induced cardiomyocytes. MiR-27a-3p inhibitor had an effect opposite to Sevo on the cardiomyocytes and further counteracted the effect of Sevo on the H/R-induced cardiomyocytes. Downregulation miR-27a-3p increased the expression of Beclin 1 and the ratio of LC3B-II to LC3B-I in H/R-induced cardiomyocytes. Furthermore, 3-MA had an opposite effect to miR-27a-3p inhibitor and further counteracted the effect of the miR-27a-3p inhibitor on H/R-induced cardiomyocytes. CONCLUSION Sevo inhibited the apoptosis of H/R-induced cardiomyocytes via regulating miR-27a-3p-mediated autophagy.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Biming Zhan
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ying Hu
- Endocrinology Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shibiao Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
Lamberti MJ, Nigro A, Casolaro V, Rumie Vittar NB, Dal Col J. Damage-Associated Molecular Patterns Modulation by microRNA: Relevance on Immunogenic Cell Death and Cancer Treatment Outcome. Cancers (Basel) 2021; 13:cancers13112566. [PMID: 34073766 PMCID: PMC8197279 DOI: 10.3390/cancers13112566] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Inside the cell, damage-associated molecular pattern molecules (DAMPs) play several physiological functions, but when they are released or translocated to the extracellular space, they gain additional immunogenic roles. Thus, DAMPs are considered key hallmarks of immunogenic cell death (ICD) in cancer, a functionally unique regulated form of stress-mediated cell death that activates the immune system response against tumor cells. Several epigenetic modulators of DAMPs have been reported. In this review, we aimed to provide an overview of the effects of microRNAs (miRNAs) on the expression of DAMPs and the putative link between miRNA, DAMPs, and cell death, focused on ICD. Overall, we propose that miRNAs, by targeting DAMPs, play critical roles in the regulation of both cell death and immune-associated mechanisms in cancer, while evidence of their potential involvement in ICD is limited. Finally, we discuss emerging data regarding the impact of miRNAs’ modulation on cancer treatment outcome. Abstract Immunogenic cell death (ICD) in cancer is a functionally unique regulated form of stress-mediated cell death that activates both the innate and adaptive immune response against tumor cells. ICD makes dying cancer cells immunogenic by improving both antigenicity and adjuvanticity. The latter relies on the spatiotemporally coordinated release or exposure of danger signals (DAMPs) that drive robust antigen-presenting cell activation. The expression of DAMPs is often constitutive in tumor cells, but it is the initiating stressor, called ICD-inducer, which finally triggers the intracellular response that determines the kinetics and intensity of their release. However, the contribution of cell-autonomous features, such as the epigenetic background, to the development of ICD has not been addressed in sufficient depth. In this context, it has been revealed that several microRNAs (miRNAs), besides acting as tumor promoters or suppressors, can control the ICD-associated exposure of some DAMPs and their basal expression in cancer. Here, we provide a general overview of the dysregulation of cancer-associated miRNAs whose targets are DAMPs, through which new molecular mediators that underlie the immunogenicity of ICD were identified. The current status of miRNA-targeted therapeutics combined with ICD inducers is discussed. A solid comprehension of these processes will provide a framework to evaluate miRNA targets for cancer immunotherapy.
Collapse
Affiliation(s)
- María Julia Lamberti
- INBIAS, CONICET-UNRC, Río Cuarto, Córdoba 5800, Argentina;
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi, 84081 Salerno, Italy; (A.N.); (V.C.)
- Correspondence: (M.J.L.); (J.D.C.)
| | - Annunziata Nigro
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi, 84081 Salerno, Italy; (A.N.); (V.C.)
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi, 84081 Salerno, Italy; (A.N.); (V.C.)
| | | | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi, 84081 Salerno, Italy; (A.N.); (V.C.)
- Correspondence: (M.J.L.); (J.D.C.)
| |
Collapse
|
11
|
Kinget L, Roussel E, Lambrechts D, Boeckx B, Vanginderhuysen L, Albersen M, Rodríguez-Antona C, Graña-Castro O, Inglada-Pérez L, Verbiest A, Zucman-Rossi J, Couchy G, Caruso S, Laenen A, Baldewijns M, Beuselinck B. MicroRNAs Possibly Involved in the Development of Bone Metastasis in Clear-Cell Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13071554. [PMID: 33800656 PMCID: PMC8036650 DOI: 10.3390/cancers13071554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Bone metastases cause substantial morbidity and implicate worse clinical outcomes for clear-cell renal cell carcinoma patients. MicroRNAs are small RNA molecules that modulate gene translation and are involved in the development of cancer and metastasis. We identified six microRNAs that are potentially specifically involved in metastasis to bone, of which two seem protective and four implicate a higher risk. This aids further understanding of the process of metastasizing to bone. Furthermore, these microRNA hold potential for biomarkers or therapeutic targets. Abstract Bone metastasis in clear-cell renal cell carcinoma (ccRCC) leads to substantial morbidity through skeletal related adverse events and implicates worse clinical outcomes. MicroRNAs (miRNA) are small non-protein coding RNA molecules with important regulatory functions in cancer development and metastasis. In this retrospective analysis we present dysregulated miRNA in ccRCC, which are associated with bone metastasis. In particular, miR-23a-3p, miR-27a-3p, miR-20a-5p, and miR-335-3p specifically correlated with the earlier appearance of bone metastasis, compared to metastasis in other organs. In contrast, miR-30b-3p and miR-139-3p were correlated with less occurrence of bone metastasis. These miRNAs are potential biomarkers and attractive targets for miRNA inhibitors or mimics, which could lead to novel therapeutic possibilities for bone targeted treatment in metastatic ccRCC.
Collapse
Affiliation(s)
- Lisa Kinget
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (E.R.); (M.A.)
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (D.L.); (B.B.)
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Bram Boeckx
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (D.L.); (B.B.)
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Loïc Vanginderhuysen
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (E.R.); (M.A.)
| | | | - Osvaldo Graña-Castro
- Centro Nacional de Investigaciones Oncológicas (CNIO), 28040 Madrid, Spain; (C.R.-A.); (O.G.-C.)
| | - Lucía Inglada-Pérez
- Department of Statistics and Operational Research, Faculty of Medicine, Complutense University, 28040 Madrid, Spain;
| | - Annelies Verbiest
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | - Gabrielle Couchy
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | | | | | - Benoit Beuselinck
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
- Correspondence: ; Tel.: +32-16-346900
| |
Collapse
|
12
|
Ibing S, Michels BE, Mosdzien M, Meyer HR, Feuerbach L, Körner C. On the impact of batch effect correction in TCGA isomiR expression data. NAR Cancer 2021; 3:zcab007. [PMID: 34316700 PMCID: PMC8210273 DOI: 10.1093/narcan/zcab007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/30/2021] [Accepted: 02/18/2021] [Indexed: 11/18/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs with diverse functions in post-transcriptional regulation of gene expression. Sequence and length variants of miRNAs are called isomiRs and can exert different functions compared to their canonical counterparts. The Cancer Genome Atlas (TCGA) provides isomiR-level expression data for patients of various cancer entities collected in a multi-center approach over several years. However, the impact of batch effects within individual cohorts has not been systematically investigated and corrected for before. Therefore, the aim of this study was to identify relevant cohort-specific batch variables and generate batch-corrected isomiR expression data for 16 TCGA cohorts. The main batch variables included sequencing platform, plate, sample purity and sequencing depth. Platform bias was related to certain length and sequence features of individual recurrently affected isomiRs. Furthermore, significant downregulation of reported tumor suppressive isomiRs in lung tumor tissue compared to normal samples was only observed after batch correction, highlighting the importance of working with corrected data. Batch-corrected datasets for all cohorts including quality control are provided as supplement. In summary, this study reveals that batch effects present in the TCGA dataset might mask biologically relevant effects and provides a valuable resource for research on isomiRs in cancer (accessible through GEO: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE164767).
Collapse
Affiliation(s)
- Susanne Ibing
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Straße 41, 69120 Heidelberg, Germany
| | - Birgitta E Michels
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Moritz Mosdzien
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Helen R Meyer
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Lars Feuerbach
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Berliner Straße 41, 69120 Heidelberg, Germany
| | - Cindy Körner
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
13
|
Wang Y, Zhou X, Han P, Lu Y, Zhong X, Yang Y, Li D, Liu D, Li Q, Pan N, Mo Y, Luo W, Li P, Zhou X, Liudmila M. Inverse correlation of miR-27a-3p and CDH5 expression serves as a diagnostic biomarker of proliferation and metastasis of clear cell renal carcinoma. Pathol Res Pract 2021; 220:153393. [PMID: 33740544 DOI: 10.1016/j.prp.2021.153393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Cadherin-5 (CDH5) is aberrantly expressed in a variety of human cancers and plays an important role in angiogenesis. The present study provides further insight into the role of miR-27a-3p in the regulation of CDH5 expression in renal clear cell carcinoma (ccRCC). METHODS Thedysregulation of CDH5 expression in ccRCC and its association with clinicopathological characteristics were analyzed using the TCGA database. A meta-analysis was performed to verify the alteration of CDH5 expression in ccRCC using the GEO database. Quantitative RT-PCR and immunohistochemical staining were applied to assess the transcriptional and protein levels of CDH5. TargetScan and Tarbase were employed to predict the miRNAs with the potential to target mRNA of CDH5. RESULTS The mRNA level of CDH5 in ccRCCwas significantly higher than in normal tissue. CDH5 mRNA expression could therefore serve as a potential diagnostic biomarker for ccRCC (AUC = 0.844). However, the reduced CDH5 transcription levels were significantly correlated with patients in the T3-4 stage, lymph node, and distant metastasis, as well as with a worse clinical outcome. We further observed that CDH5, at the protein level, was almost absent in ccRCC samples. In addition, a few databases screen showed that mir-27a-3p is a highly conserved miRNA targeting CDH5. The expression of mir-27a-3p was significantly elevated in ccRCC tissues in contrast to normal tissues. Importantly, it was positively associated with the T3-4 stage and M stage, respectively, suggesting that the expression level of mir-27a-3p could serve as a diagnostic biomarker for ccRCC (AUC = 0.775). CONCLUSION Our data suggest that thereduced translational level of CDH5 in ccRCC was related to the overexpression of mir-27a-3p. The higher mir-27a-3p and lower CDH5 expression significantly correlated with advanced clinical stages for ccRCC patients.
Collapse
Affiliation(s)
- Yifang Wang
- Life Science Institute, Guangxi Medical University, China
| | - Xiaohui Zhou
- Life Science Institute, Guangxi Medical University, China
| | - Peipei Han
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Yunliang Lu
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xuemin Zhong
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Yanping Yang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Danping Li
- Life Science Institute, Guangxi Medical University, China
| | - Deling Liu
- Life Science Institute, Guangxi Medical University, China
| | - Qiuyun Li
- Life Science Institute, Guangxi Medical University, China
| | - Nenghui Pan
- Life Science Institute, Guangxi Medical University, China
| | - Yingxi Mo
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Wenqi Luo
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Ping Li
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xiaoying Zhou
- Life Science Institute, Guangxi Medical University, China.
| | - Matskova Liudmila
- Institute of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad, Russia.
| |
Collapse
|
14
|
Chen H, Cai B, Liu K, Hua Q. miR‑27a‑3p regulates the inhibitory influence of endothelin 3 on the tumorigenesis of papillary thyroid cancer cells. Mol Med Rep 2021; 23:243. [PMID: 33537832 PMCID: PMC7893708 DOI: 10.3892/mmr.2021.11882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 01/04/2021] [Indexed: 11/27/2022] Open
Abstract
Several studies on papillary thyroid cancer (PTC) have been performed. However, the effects of endothelin 3 (EDN3) and microRNA (miR)-27a-3p on PTC cells has yet to be investigated, to the best of the authors' knowledge. The present study aimed to explore the biological functions of EDN3 and miR-27a-3p in PTC cells. Bioinformatics analysis was conducted to identify possible key genes and miRs involved in PTC progression. Western blot analysis and reverse transcription-quantitative (RT-q) PCR were employed to confirm the key genes or miRs expressed in PTC cells. Cytological methods were used to detect cell viability, proliferation, apoptosis and migration and luciferase reporter assay was performed to confirm the relationship between END3 and miR-27a-3p. After analyzing the results of gene microarray analyses and RT-qPCR, EDN3 with low expression was identified as the key gene associated with PTC progression. It was also found that EDN3 overexpression in PTC cells impaired cell viability, proliferation and migration but promoted cell apoptosis. In addition, the findings revealed that miR-27a-3p could relieve the inhibitory influence of EDN3 on PTC cells by binding to EDN3 mRNA 3′ untranslated region (UTR), thereby suppressing EDN3 expression. Overall, the results of the present study demonstrated that by binding to EDN3 mRNA 3′UTR, miR-27a-3p could attenuate the inhibitory function of EDN3 in the tumorigenesis of PTC cells.
Collapse
Affiliation(s)
- Hongxin Chen
- Department of Otorhinolaryngology‑Head and Neck Surgery, Wuhan Puren Hospital, Wuhan, Hubei 430081, P.R. China
| | - Binlin Cai
- Department of Otorhinolaryngology‑Head and Neck Surgery, Wuhan Puren Hospital, Wuhan, Hubei 430081, P.R. China
| | - Kun Liu
- Department of Otorhinolaryngology‑Head and Neck Surgery, Wuhan Puren Hospital, Wuhan, Hubei 430081, P.R. China
| | - Qingquan Hua
- Department of Otorhinolaryngology‑Head and Neck Surgery, Wuhan University Renmin Hospital, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
15
|
Guo X, Li M. LINC01089 is a tumor-suppressive lncRNA in gastric cancer and it regulates miR-27a-3p/TET1 axis. Cancer Cell Int 2020; 20:507. [PMID: 33088215 PMCID: PMC7568383 DOI: 10.1186/s12935-020-01561-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most common malignancies around the world. Recently, the role of long non-coding RNA (lncRNA) in cancer biology has become a hot research topic. This work aimed to explore the biological function and underlying mechanism of LINC01089 in GC. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to investigate the expression of LINC01089 in GC tissues and cells. The relationship between the expression level of LINC01089 and the clinicopathological parameters of GC was assessed. Cell models of LINC01089 overexpression, LINC01089 knockdown, miR-27a-3p overexpression, and miR-27a-3p inhibition were established by transfection. CCK-8 assay, BrdU assay, and Transwell assay were utilized to investigate the malignant biological behaviors of GC cell lines after transfection. Dual luciferase activity reporter assay, Pearson’s correlation analysis, and Western blot were utilized to the regulatory relationships among LINC01089, miR-27a-3p and tet methylcytosine dioxygenase 1 (TET1). Result LINC01089 down-regulation was observed in GC tissues and cell lines. Low expression level of LINC01089 in GC tissues was markedly linked to larger tumor size, higher T stage, as well as lymphatic metastasis of the patients. Functional experiments implied that LINC01089 overexpression impeded the proliferation, migration, as well as invasion of GC cells, whereas LINC01089 knockdown promoted the above malignant phenotypes. Additionally, up-regulation of miR-27a-3p was also observed in GC tissues. Functional experiments also showed that, miR-27a-3p overexpression boosted the malignant biological behaviors of GC cells; on the contrast, these phenotypes were impeded by miR-27a-3p inhibition. Moreover, LINC01089 interacted with and repressed miR-27a-3p, and miR-27a-3p antagonized the impact of LINC01089 on GC cells. Additionally, TET1 was verified as a target gene of miR-27a-3p, and could be positively regulated by LINC01089. Conclusion LINC01089 impedes the proliferation, migration, and invasion of GC cells by adsorbing miR-27a-3p and up-regulating the expression of TET1.
Collapse
Affiliation(s)
- Xufeng Guo
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430000 Hubei China
| | - Ming Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Zhangzhidong Road, Wuchang District, Wuhan, 430000 Hubei China
| |
Collapse
|
16
|
Salmani T, Ghaderian SMH, Hajiesmaeili M, Rezaeimirghaed O, Hoseini MS, Rakhshan A, Nasiri MJ, Ghaedi H, Akbarzadeh R. Hsa-miR-27a-3p and epidermal growth factor receptor expression analysis in glioblastoma FFPE samples. Asia Pac J Clin Oncol 2020; 17:e185-e190. [PMID: 33029912 DOI: 10.1111/ajco.13399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 05/23/2020] [Indexed: 12/27/2022]
Abstract
AIM Glioblastoma multiforme (GBM) is the most invasive type of glial tumors. MicroRNAs as the small, noncoding RNAs have been revealed that play an important role in tumorigenic processes. So, they may be used as potential biomarkers for detection and prognosis of cancers at the early stages. In addition, they can be applied as therapeutic targets. In the present study, the expression levels of hsa-miR-27a-3p and EGFR were investigated in GBM. METHODS Real-time RT-PCR was applied to evaluate hsa-miR-27a-3p and EGFR expressions in the formalin-fixed paraffin-embedded (FFPE) tissue samples obtained from 50 GBM and 50 normal people. RESULTS The expression level of hsa-miR-27a-3p and EGFR was significantly different between cases and controls. Positive association was found between gene expressions and immunohistochemistry markers, such as Ki67 and glial fibrillary acidic protein, except for IDH1 status. CONCLUSION We showed the association of hsa-miR-27a-3p and EGFR with GBM and it can be concluded that they have a promising potential to use as primary cancer biomarkers.
Collapse
Affiliation(s)
- Tayyebali Salmani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayyed Mohammad Hossein Ghaderian
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Hajiesmaeili
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omidvar Rezaeimirghaed
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Azadeh Rakhshan
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Nasiri
- Department of Microbiology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Akbarzadeh
- Institute of Anatomy, University of Lübeck, Lübeck, Germany.,Skin Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Li S, Han Y, Liang X, Zhao M. LINC01089 inhibits the progression of cervical cancer via inhibiting miR-27a-3p and increasing BTG2. J Gene Med 2020; 23:e3280. [PMID: 33025678 DOI: 10.1002/jgm.3280] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/27/2020] [Accepted: 09/27/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increasing evidence confirms that long non-coding RNA (lncRNA) has a vital impact on the procession of cervical cancer (CC). The present study aimed to investigate the clinical significance of LINC01089 in CC, as well as explore its biological functions and potential molecular mechanisms. METHODS A quantitative real-time polymerase chain reaction (qRT-PCR) was utilized to investigate the expression of LINC01089 and miR-27a-3p in CC cells and tissues. Analysis of the correlation between the expression level of LINC01089 and the clinical pathological parameters of CC was then conducted. The human CC cell lines HeLa and SiHa were utilized for transfection to establish a gain-of-function model and loss-of-function models. Western blotting and a qRT-PCR were performed to detect B-cell translocation gene-2 (BTG2) expression in CC cells. Cell counting kit (CCK)-8 and 5-bromo-2-deoxyuridine (BrdU) assays were performed to detect the proliferation of CC cells. The transwell method was employed to evaluate the migration and invasion of CC cells. The interactions between LINC01089 and miR-27a-3p were verified by bioinformatics, a dual luciferase reporter gene experiment and a RNA immunoprecipitation experiment, respectively. RESULTS The expression of LINC01089 in CC was markedly down-regulated. The low expression of LINC01089 in CC was closely associated with a larger tumor size and positive lymph node metastasis. Moreover, overexpression of LINC01089 impeded the proliferation and metastasis of CC cells, whereas knockdown of LINC01089 had the opposite biological functions. In terms of mechanism, LINC01089 could sponge miR-27a-3p and indirectly up-regulate BTG2 expression. CONCLUSIONS LINC01089, as a tumor suppressor, impedes the development of CC by targeting miR-27a-3p to up-regulate BTG2 expression.
Collapse
Affiliation(s)
- Shuoxi Li
- Jiamusi College of Heilongjiang University of Chinese Medicine, Jiamusi, Heilongjiang Province, China
| | - Yu Han
- Graduate school of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Xuesong Liang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital Group, Shenzhen, Guangdong Province, China
| | - Min Zhao
- Jiamusi College of Heilongjiang University of Chinese Medicine, Jiamusi, Heilongjiang Province, China
| |
Collapse
|
18
|
Liu Y, Liu X, Zhou Y, Liu T, Li J. Overexpression of miR-27a predicts poor prognosis and promotes the progression in cholangiocarcinoma. Clin Exp Med 2020; 21:121-128. [PMID: 32816152 DOI: 10.1007/s10238-020-00655-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 08/13/2020] [Indexed: 10/23/2022]
Abstract
The function of microRNA-27a (miR-27a) expression in cholangiocarcinoma (CCA) remains largely unclear; therefore, this study aimed to investigate the clinical significance and functional role of miR-27a in CCA. This study included 117 paired CCA tissues and adjacent normal tissues from CCA patients who received surgical resection. Reverse transcription-quantitative polymerase chain reaction was used to measure the expression levels of miR-27a in CCA tissues and cell lines. A Kaplan-Meier curve and Cox regression analysis were used to determine overall prognostic performance. The effects of miR-27a on cell proliferation, migration, and invasion were measured by CCK-8 and Transwell assays. The expression levels of miR-27a in patients with CCA and cell lines were higher than those in adjacent normal tissues and normal cells, respectively. Additionally, miR-27a levels were found to be associated with lymph node metastasis and TNM stages. The overall survival time of CCA patients with high miR-27a expression was poorer than that of those with low miR-27a expression. Furthermore, miR-27a overexpression promoted CCA cell proliferation, migration, and invasion, whereas knockdown of miR-27a suppressed cell proliferation, migration, and invasion. Taken together, these results suggest the potential usefulness of miR-27a in the prognosis and progression of CCA.
Collapse
Affiliation(s)
- Yunxia Liu
- Department of Internal Medicine, Fuyanshan Branch of Affiliated Hospital of Weifang Medical University, Intersection of Limin Road and Fuyuan Street, Weifang, 261053, Shandong, China.
| | - Xia Liu
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, Shandong, China
| | - Yanhua Zhou
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, Shandong, China
| | - Tingting Liu
- Department of Internal Medicine, Fuyanshan Branch of Affiliated Hospital of Weifang Medical University, Intersection of Limin Road and Fuyuan Street, Weifang, 261053, Shandong, China
| | - Jie Li
- Department of Internal Medicine, Fuyanshan Branch of Affiliated Hospital of Weifang Medical University, Intersection of Limin Road and Fuyuan Street, Weifang, 261053, Shandong, China
| |
Collapse
|
19
|
Aguilera-Rojas M, Sharbati S, Stein T, Einspanier R. Deregulation of miR-27a may contribute to canine fibroblast activation after coculture with a mast cell tumour cell line. FEBS Open Bio 2020; 10:802-816. [PMID: 32133790 PMCID: PMC7193169 DOI: 10.1002/2211-5463.12831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/27/2020] [Accepted: 03/03/2020] [Indexed: 12/26/2022] Open
Abstract
The tumour microenvironment comprises a diverse range of cells, including fibroblasts, immune cells and endothelial cells, along with extracellular matrix. In particular, fibroblasts are of significant interest as these cells are reprogrammed during tumorigenesis to become cancer‐associated fibroblasts (CAFs), which in turn support cancer cell growth. MicroRNAs (miRNAs) have been shown to be involved in this intercellular crosstalk in humans. To assess whether miRNAs are also involved in the activation of fibroblasts in dogs, we cocultured primary canine skin fibroblasts with the canine mast cell tumour cell line C2 directly or with C2‐derived exosomes, and measured differential abundance of selected miRNAs. Expression of the CAF markers alpha‐smooth muscle actin (ACTA2) and stanniocalcin 1 confirmed the activation of our fibroblasts after coculture. We show that fibroblasts displayed significant downregulation of miR‐27a and let‐7 family members. These changes correlated with significant upregulation of predicted target mRNAs. Furthermore, RNA interference knockdown of miR‐27a revealed that cyclin G1 (CCNG1) exhibited negative correlation at the mRNA and protein level, suggesting that CCNG1 is a target of miR‐27a in canine fibroblasts and involved in their activation. Importantly, miR‐27a knockdown itself resulted in fibroblast activation, as demonstrated by the formation of ACTA2 filaments. In addition, interleukin‐6 (IL‐6) was strongly induced in our fibroblasts when cocultured, indicating potential reciprocal signalling. Taken together, our findings are consistent with canine fibroblasts being reprogrammed into CAFs to further support cancer development and that downregulation of miR‐27a may play an important role in the tumour–microenvironment crosstalk.
Collapse
Affiliation(s)
- Matias Aguilera-Rojas
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Torsten Stein
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| |
Collapse
|