1
|
Hedayati N, Safaei Naeini M, Ale Sahebfosoul MM, Mafi A, Eshaghi Milasi Y, Rizaneh A, Nabavi N, Farahani N, Alimohammadi M, Ghezelbash B. MicroRNA dysregulation and its impact on apoptosis-related signaling pathways in myelodysplastic syndrome. Pathol Res Pract 2024; 261:155478. [PMID: 39079383 DOI: 10.1016/j.prp.2024.155478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 08/18/2024]
Abstract
Myelodysplastic syndrome (MDS) holds a unique position among blood cancers, encompassing a spectrum of blood-related disorders marked by impaired maturation of blood cell precursors, bone marrow abnormalities, genetic instability, and a higher likelihood of progressing to acute myeloid leukemia. MicroRNAs (miRNAs), short non-coding RNA molecules typically 18-24 nucleotides in length, are known to regulate gene expression and contribute to various biological processes, including cellular differentiation and programmed cell death. Additionally, miRNAs are involved in many aspects of cancer development, influencing cell growth, transformation, and apoptosis. In this study, we explore the impact of microRNAs on cellular apoptosis in MDS.
Collapse
Affiliation(s)
- Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mobina Safaei Naeini
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yaser Eshaghi Milasi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anahita Rizaneh
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada.
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Behrooz Ghezelbash
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Micheva ID, Atanasova SA. MicroRNA dysregulation in myelodysplastic syndromes: implications for diagnosis, prognosis, and therapeutic response. Front Oncol 2024; 14:1410656. [PMID: 39156702 PMCID: PMC11327013 DOI: 10.3389/fonc.2024.1410656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Myelodysplastic syndromes (MDS) are a group of malignant clonal hematological disorders with heterogeneous clinical course and risk of transformation to acute myeloid leukemia. Genetic and epigenetic dysregulation, including alterations in microRNA (miRNA) expression, plays a pivotal role in MDS pathogenesis influencing disease development and progression. MiRNAs, known for their regulatory roles in gene expression, have emerged as promising biomarkers in various malignant diseases. This review aims to explore the diagnostic and prognostic roles of miRNAs in MDS. We discuss research efforts aimed at understanding the clinical utility of miRNAs in MDS management. MiRNA dysregulation is linked to specific chromosomal abnormalities in MDS, providing insights into the molecular landscape of the disease. Circulating miRNAs in plasma offer a less invasive avenue for diagnostic and prognostic assessment, with distinct miRNA profiles identified in MDS patients. Additionally, we discuss investigations concerning the role of miRNAs as markers for treatment response to hypomethylating and immunomodulating agents, which could lead to improved treatment decision-making and monitoring. Despite significant progress, further research in larger patient cohorts is needed to fully elucidate the role of miRNAs in MDS pathogenesis and refine personalized approaches to patient care.
Collapse
Affiliation(s)
- Ilina Dimitrova Micheva
- Hematology Department, University Hospital St. Marina, Varna, Bulgaria
- Faculty of Medicine, Medical University of Varna, Varna, Bulgaria
| | - Svilena Angelova Atanasova
- Hematology Department, University Hospital St. Marina, Varna, Bulgaria
- Faculty of Medicine, Medical University of Varna, Varna, Bulgaria
| |
Collapse
|
3
|
Zheng S, Liu Y. Progress in the Study of Fra-2 in Respiratory Diseases. Int J Mol Sci 2024; 25:7143. [PMID: 39000247 PMCID: PMC11240912 DOI: 10.3390/ijms25137143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Fos-related antigen-2 (Fra-2) is a member of the activating protein-1 (AP-1) family of transcription factors. It is involved in controlling cell growth and differentiation by regulating the production of the extracellular matrix (ECM) and coordinating the balance of signals within and outside the cell. Fra-2 is not only closely related to bone development, metabolism, and immune system and eye development but also in the progression of respiratory conditions like lung tumors, asthma, pulmonary fibrosis, and chronic obstructive pulmonary disease (COPD). The increased expression and activation of Fra-2 in various lung diseases has been shown in several studies. However, the specific molecular mechanisms through which Fra-2 affects the development of respiratory diseases are not yet understood. The purpose of this research is to summarize and delineate advancements in the study of the involvement of transcription factor Fra-2 in disorders related to the respiratory system.
Collapse
Affiliation(s)
- Shuping Zheng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yun Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
4
|
Fu T, Amoah K, Chan TW, Bahn JH, Lee JH, Terrazas S, Chong R, Kosuri S, Xiao X. Massively parallel screen uncovers many rare 3' UTR variants regulating mRNA abundance of cancer driver genes. Nat Commun 2024; 15:3335. [PMID: 38637555 PMCID: PMC11026479 DOI: 10.1038/s41467-024-46795-7] [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: 05/01/2023] [Accepted: 03/06/2024] [Indexed: 04/20/2024] Open
Abstract
Understanding the function of rare non-coding variants represents a significant challenge. Using MapUTR, a screening method, we studied the function of rare 3' UTR variants affecting mRNA abundance post-transcriptionally. Among 17,301 rare gnomAD variants, an average of 24.5% were functional, with 70% in cancer-related genes, many in critical cancer pathways. This observation motivated an interrogation of 11,929 somatic mutations, uncovering 3928 (33%) functional mutations in 155 cancer driver genes. Functional MapUTR variants were enriched in microRNA- or protein-binding sites and may underlie outlier gene expression in tumors. Further, we introduce untranslated tumor mutational burden (uTMB), a metric reflecting the amount of somatic functional MapUTR variants of a tumor and show its potential in predicting patient survival. Through prime editing, we characterized three variants in cancer-relevant genes (MFN2, FOSL2, and IRAK1), demonstrating their cancer-driving potential. Our study elucidates the function of tens of thousands of non-coding variants, nominates non-coding cancer driver mutations, and demonstrates their potential contributions to cancer.
Collapse
Affiliation(s)
- Ting Fu
- Molecular, Cellular and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kofi Amoah
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tracey W Chan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jae Hoon Bahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jae-Hyung Lee
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Life and Nanopharmaceutical Sciences & Oral Microbiology, School of Dentistry, Kyung Hee University, Seoul, South Korea
| | - Sari Terrazas
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Rockie Chong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Sriram Kosuri
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinshu Xiao
- Molecular, Cellular and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
5
|
Rampioni Vinciguerra GL, Capece M, Scafetta G, Rentsch S, Vecchione A, Lovat F, Croce CM. Role of Fra-2 in cancer. Cell Death Differ 2024; 31:136-149. [PMID: 38104183 PMCID: PMC10850073 DOI: 10.1038/s41418-023-01248-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
Fos-related antigen-2 (Fra-2) is the most recently discovered member of the Fos family and, by dimerizing with Jun proteins, forms the activator protein 1 (AP-1) transcription factor. By inducing or repressing the transcription of several target genes, Fra-2 is critically involved in the modulation of cell response to a variety of extracellular stimuli, stressors and intracellular changes. In physiological conditions, Fra-2 has been found to be ubiquitously expressed in human cells, regulating differentiation and homeostasis of bone, muscle, nervous, lymphoid and other tissues. While other AP-1 members, like Jun and Fos, are well characterized, studies of Fra-2 functions in cancer are still at an early stage. Due to the lack of a trans-activating domain, which is present in other Fos proteins, it has been suggested that Fra-2 might inhibit cell transformation, eventually exerting an anti-tumor effect. In human malignancies, however, Fra-2 activity is enhanced (or induced) by dysregulation of microRNAs, oncogenes and extracellular signaling, suggesting a multifaceted role. Therefore, Fra-2 can promote or prevent transformation, proliferation, migration, epithelial-mesenchymal transition, drug resistance and metastasis formation in a tumor- and context-dependent manner. Intriguingly, recent data reports that Fra-2 is also expressed in cancer associated cells, contributing to the intricate crosstalk between neoplastic and non-neoplastic cells, that leads to the evolution and remodeling of the tumor microenvironment. In this review we summarize three decades of research on Fra-2, focusing on its oncogenic and anti-oncogenic effects in tumor progression and dissemination.
Collapse
Affiliation(s)
- Gian Luca Rampioni Vinciguerra
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Marina Capece
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Giorgia Scafetta
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Sydney Rentsch
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Francesca Lovat
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
6
|
Yu L, Zhang F, Wang Y. Circ_0005615 Regulates the Progression of Colorectal Cancer Through the miR-873-5p/FOSL2 Signaling Pathway. Biochem Genet 2023; 61:2020-2041. [PMID: 36920708 DOI: 10.1007/s10528-023-10355-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/15/2023] [Indexed: 03/16/2023]
Abstract
To determine the effects of circ_0005615 in CRC development and underneath mechanism. The expression levels of circ_0005615, microRNA-873-5p (miR-873-5p) and FOS-like antigen 2 (FOSL2) mRNA were determined by quantitative real-time polymerase chain reaction (qRT-PCR). The protein levels of exosome makers, proliferation-related makers and FOSL2 were detected by western blot or immunohistochemistry assay. Cell proliferation was evaluated by cell counting kit-8 (CCK-8) and cell colony formation assays. Cell migration and invasion were demonstrated by a transwell assay. Cell apoptosis was investigated by flow cytometry analysis. The binding relationship between miR-873-5p and circ_0005615 or FOSL2 was predicted by circular RNA interactome and targetscan online databases, respectively, and identified by dual-luciferase reporter assay. The impacts of circ_0005615 silencing on tumor formation were determined by in vivo tumor formation assay. Circ_0005615 expression was dramatically upregulated in serum exosomes of CRC patients compared with the control group. The CRC patients with a high circ_0005615 expression had a poor survival rate. Circ_0005615 and FOSL2 expressions were apparently increased, while miR-873-5p was decreased in CRC tissues or cells relative to control groups. Circ_0005615 knockdown inhibited cell proliferation, migration, and invasion, whereas promoted cell apoptosis in CRC; however, miR-873-5p inhibitor attenuated these impacts. Additionally, circ_0005615 acted as a sponge of miR-873-5p and miR-873-5p bound to FOSL2. FOSL2 overexpression restrained the effects of miR-873-5p mimic on CRC progression. Furthermore, circ_0005615 knockdown suppressed tumor growth in vivo. Circ_0005615 modulated CRC malignant progression by controlling FOSL2 expression through sponging miR-873-5p. This finding lays a foundation for the study on circRNA-mediated CRC therapy.
Collapse
Affiliation(s)
- Lihua Yu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
| | - Feifei Zhang
- Department of General Surgery, Maternity and Child Health Care of Laizhou, No. 288 Wenhua East Street, Laizhou, 261400, Shandong, People's Republic of China
| | - Yeli Wang
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Yantai, 264000, Shandong, People's Republic of China.
| |
Collapse
|
7
|
Taghehchian N, Samsami Y, Maharati A, Zangouei AS, Boroumand-Noughabi S, Moghbeli M. Molecular biology of microRNA-342 during tumor progression and invasion. Pathol Res Pract 2023; 248:154672. [PMID: 37413875 DOI: 10.1016/j.prp.2023.154672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023]
Abstract
Cancer is considered as one of the main causes of human deaths and health challenges in the world. Various factors are involved in the high death rate of cancer patients, including late diagnosis and drug resistance that result in treatment failure and tumor recurrence. Invasive diagnostic methods are one of the main reasons of late tumor detection in cancer patients. Therefore, it is necessary to investigate the molecular tumor biology to introduce efficient non-invasive markers. MicroRNAs (miRNAs) are involved in regulation of the cellular mechanisms such as cell proliferation, apoptosis, and migration. MiRNAs deregulations have been also frequently shown in different tumor types. Here, we discussed the molecular mechanisms of miR-342 during tumor growth. MiR-342 mainly functions as a tumor suppressor by the regulation of transcription factors and signaling pathways such as WNT, PI3K/AKT, NF-kB, and MAPK. Therefore, miR-342 mimics can be used as a reliable therapeutic strategy to inhibit the tumor cells growth. The present review can also pave the way to introduce the miR-342 as a non-invasive diagnostic/prognostic marker in cancer patients.
Collapse
Affiliation(s)
- Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yalda Samsami
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Boroumand-Noughabi
- Department of Hematology and Blood Bank, 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
|
8
|
Nandanpawar P, Sahoo L, Sahoo B, Murmu K, Chaudhari A, Pavan kumar A, Das P. Identification of differentially expressed genes and SNPs linked to harvest body weight of genetically improved rohu carp, Labeo rohita. Front Genet 2023; 14:1153911. [PMID: 37359361 PMCID: PMC10285081 DOI: 10.3389/fgene.2023.1153911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
In most of the aquaculture selection programs, harvest body weight has been a preferred performance trait for improvement. Molecular interplay of genes linked to higher body weight is not elucidated in major carp species. The genetically improved rohu carp with 18% average genetic gain per generation with respect to harvest body weight is a promising candidate for studying genes' underlying performance traits. In the present study, muscle transcriptome sequencing of two groups of individuals, with significant difference in breeding value, belonging to the tenth generation of rohu carp was performed using the Illumina HiSeq 2000 platform. A total of 178 million paired-end raw reads were generated to give rise to 173 million reads after quality control and trimming. The genome-guided transcriptome assembly and differential gene expression produced 11,86,119 transcripts and 451 upregulated and 181 downregulated differentially expressed genes (DEGs) between high-breeding value and low-breeding value (HB & LB) groups, respectively. Similarly, 39,158 high-quality coding SNPs were identified with the Ts/Tv ratio of 1.23. Out of a total of 17 qPCR-validated transcripts, eight were associated with cellular growth and proliferation and harbored 13 SNPs. The gene expression pattern was observed to be positively correlated with RNA-seq data for genes such as myogenic factor 6, titin isoform X11, IGF-1 like, acetyl-CoA, and thyroid receptor hormone beta. A total of 26 miRNA target interactions were also identified to be associated with significant DETs (p-value < 0.05). Genes such as Myo6, IGF-1-like, and acetyl-CoA linked to higher harvest body weight may serve as candidate genes in marker-assisted breeding and SNP array construction for genome-wide association studies and genomic selection.
Collapse
Affiliation(s)
- P. Nandanpawar
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - L. Sahoo
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - B. Sahoo
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - K. Murmu
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - A. Chaudhari
- ICAR-Central Institute of Fisheries Education, Mumbai, Maharashtra, India
| | - A. Pavan kumar
- ICAR-Central Institute of Fisheries Education, Mumbai, Maharashtra, India
| | - P. Das
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| |
Collapse
|
9
|
Słomka A, Kornek M, Cho WC. Small Extracellular Vesicles and Their Involvement in Cancer Resistance: An Up-to-Date Review. Cells 2022; 11:2913. [PMID: 36139487 PMCID: PMC9496799 DOI: 10.3390/cells11182913] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/25/2022] [Accepted: 09/15/2022] [Indexed: 12/11/2022] Open
Abstract
In recent years, tremendous progress has been made in understanding the roles of extracellular vesicles (EVs) in cancer. Thanks to advancements in molecular biology, it has been found that the fraction of EVs called exosomes or small EVs (sEVs) modulates the sensitivity of cancer cells to chemotherapeutic agents by delivering molecularly active non-coding RNAs (ncRNAs). An in-depth analysis shows that two main molecular mechanisms are involved in exosomal modified chemoresistance: (1) translational repression of anti-oncogenes by exosomal microRNAs (miRs) and (2) lack of translational repression of oncogenes by sponging of miRs through long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs). At the cellular level, these processes increase the proliferation and survival of cancer cells and improve their ability to metastasize and resist apoptosis. In addition, studies in animal models have shown enhancing tumor size under the influence of exosomal ncRNAs. Ultimately, exosomal ncRNAs are responsible for clinically significant chemotherapy failures in patients with different types of cancer. Preliminary data have also revealed that exosomal ncRNAs can overcome chemotherapeutic agent resistance, but the results are thoroughly fragmented. This review presents how exosomes modulate the response of cancer cells to chemotherapeutic agents. Understanding how exosomes interfere with chemoresistance may become a milestone in developing new therapeutic options, but more data are still required.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland
| | - Miroslaw Kornek
- Department of Internal Medicine I, University Hospital of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| |
Collapse
|
10
|
Kang SH, Kim HB, Choi JS. Upregulation of microRNA-597 in myelodysplastic syndromes induces apoptosis through FOSL2 inhibition. Eur J Haematol Suppl 2022; 109:680-685. [PMID: 36018564 DOI: 10.1111/ejh.13852] [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: 03/21/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Dysregulation of microRNAs (miRNAs) has been associated with the pathophysiology of myelodysplastic syndrome (MDS). Trisomy 8 is the most frequent chromosomal abnormalities in Korean patients with myelodysplastic syndrome (MDS). We investigated the dysregulation of miR-597-5p, located on chromosome 8, which is reported to induce cell death in numerous cancers. MATERIALS & METHODS We compared the expression profiles of miR-597-5p among 65 MDS patients and 11 controls, and analyzed the in vitro effects of miR-597 on leukemic cells using an acute myeloid leukemia (AML) cell line transfected with miR-597. RESULTS We found that miR-597-5p levels were upregulated 4.05-fold in MDS patients compared to those in controls. In vitro study results demonstrated that transfection with a miR-597 mimic induced apoptosis through downregulation of FOS like 2 (FOSL2). CONCLUSION These findings suggest that upregulation of miR-597 induces apoptosis and that miR-597 has a possible role in the pathophysiology of MDS. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Seong-Ho Kang
- Department of Laboratory Medicine, Chosun University College of Medicine, Gwangju, Republic of Korea
| | - Hong-Beum Kim
- Department of Premedical Course, Chosun University College of Medicine, Gwangju, Republic of Korea
| | - Ji Seon Choi
- Department of Laboratory Medicine, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic of Korea
| |
Collapse
|
11
|
Bhattacharya N, Indra AK, Ganguli-Indra G. Selective Ablation of BCL11A in Epidermal Keratinocytes Alters Skin Homeostasis and Accelerates Excisional Wound Healing In Vivo. Cells 2022; 11:cells11132106. [PMID: 35805190 PMCID: PMC9265695 DOI: 10.3390/cells11132106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 01/27/2023] Open
Abstract
Transcriptional regulator BCL11A plays a crucial role in coordinating a suite of developmental processes including skin morphogenesis, barrier functions and lipid metabolism. There is little or no reports so far documenting the role of BCL11A in postnatal adult skin homeostasis and in the physiological process of tissue repair and regeneration. The current study establishes for the first time the In Vivo role of epidermal BCL11A in maintaining adult epidermal homeostasis and as a negative regulator of cutaneous wound healing. Conditional ablation of Bcl11a in skin epidermal keratinocytes (Bcl11aep−/−mice) enhances the keratinocyte proliferation and differentiation program, suggesting its critical role in epidermal homeostasis of adult murine skin. Further, loss of keratinocytic BCL11A promotes rapid closure of excisional wounds both in a cell autonomous manner likely via accelerating wound re-epithelialization and in a non-cell autonomous manner by enhancing angiogenesis. The epidermis specific Bcl11a knockout mouse serves as a prototype to gain mechanistic understanding of various downstream pathways converging towards the manifestation of an accelerated healing phenotype upon its deletion.
Collapse
Affiliation(s)
- Nilika Bhattacharya
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA;
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA;
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Science Center, Oregon State University, Corvallis, OR 97331, USA
- OHSU Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
- Department of Dermatology, OHSU, Portland, OR 97239, USA
- Correspondence: (A.K.I.); (G.G.-I.)
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA;
- OHSU Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
- Correspondence: (A.K.I.); (G.G.-I.)
| |
Collapse
|
12
|
Ankill J, Aure MR, Bjørklund S, Langberg S, Kristensen VN, Vitelli V, Tekpli X, Fleischer T. Epigenetic alterations at distal enhancers are linked to proliferation in human breast cancer. NAR Cancer 2022; 4:zcac008. [PMID: 35350772 PMCID: PMC8947789 DOI: 10.1093/narcan/zcac008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/23/2022] [Accepted: 03/14/2022] [Indexed: 11/26/2022] Open
Abstract
Aberrant DNA methylation is an early event in breast carcinogenesis and plays a critical role in regulating gene expression. Here, we perform genome-wide expression-methylation Quantitative Trait Loci (emQTL) analysis through the integration of DNA methylation and gene expression to identify disease-driving pathways under epigenetic control. By grouping the emQTLs using biclustering we identify associations representing important biological processes associated with breast cancer pathogenesis including regulation of proliferation and tumor-infiltrating fibroblasts. We report genome-wide loss of enhancer methylation at binding sites of proliferation-driving transcription factors including CEBP-β, FOSL1, and FOSL2 with concomitant high expression of proliferation-related genes in aggressive breast tumors as we confirm with scRNA-seq. The identified emQTL-CpGs and genes were found connected through chromatin loops, indicating that proliferation in breast tumors is under epigenetic regulation by DNA methylation. Interestingly, the associations between enhancer methylation and proliferation-related gene expression were also observed within known subtypes of breast cancer, suggesting a common role of epigenetic regulation of proliferation. Taken together, we show that proliferation in breast cancer is linked to loss of methylation at specific enhancers and transcription factor binding and gene activation through chromatin looping.
Collapse
Affiliation(s)
- Jørgen Ankill
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Miriam Ragle Aure
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sunniva Bjørklund
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | | | - Vessela N Kristensen
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Valeria Vitelli
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Xavier Tekpli
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thomas Fleischer
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
Ji C, Hong X, Lan B, Lin Y, He Y, Chen J, Liu X, Ye W, Mo Z, She Z, Lin S. Circ_0091581 Promotes the Progression of Hepatocellular Carcinoma Through Targeting miR-591/FOSL2 Axis. Dig Dis Sci 2021; 66:3074-3085. [PMID: 33040214 DOI: 10.1007/s10620-020-06641-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Circular RNAs (circRNAs) have shown crucial regulatory roles in cancer biology. We aimed to uncover the role and underlying mechanism of circ_0091581 in hepatocellular carcinoma (HCC) progression. METHODS The abundance of circ_0091581, microRNA-591 (miR-591) and FOS like 2, AP-1 transcription factor subunit (FOSL2) was measured by quantitative real-time polymerase chain reaction. Cell viability, colony formation ability, and invasion ability were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay, and transwell invasion assay. The migration ability was analyzed by transwell migration assay and wound healing assay. Flow cytometry was used to evaluate the cell cycle and apoptosis of HCC cells. The interaction between miR-591 and circ_0091581 or FOSL2 was predicted by Circular RNA Interactome database or TargetScan database and confirmed by dual-luciferase reporter assay and RNA immune co-precipitation assay. FOSL2 protein expression was measured by Western blot assay. Xenograft tumor assay was conducted to analyze the role of circ_0091581 in HCC tumor growth in vivo. RESULTS Circ_0091581 was highly expressed in HCC tissue samples and cell lines in contrast to that in adjacent normal tissue samples and THLE-2 cell line. Circ_0091581 accelerated the viability, colony formation, metastasis, and cell cycle, while it impeded the apoptosis of HCC cells. MiR-591 bound to circ_0091581, and circ_0091581 knockdown-mediated effects in HCC cells were largely overturned by miR-591 silencing. FOSL2 was a target of miR-591, and FOSL2 overexpression largely reversed miR-591 accumulation-induced influences in HCC cells. FOSL2 protein expression was down-regulated by circ_0091581 silencing, and the addition of miR-591 inhibitor partly recovered the expression of FOSL2 in HCC cells. Circ_0091581 interference notably suppressed HCC tumor growth in vivo. CONCLUSION Circ_0091581 acted as an oncogene to enhance the viability, colony formation, metastasis and cell cycle and inhibit the apoptosis of HCC cells through targeting miR-591/FOSL2 axis.
Collapse
Affiliation(s)
- Chenggang Ji
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China
| | - Xiaocheng Hong
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China
| | - Bo Lan
- Department of Anorectal Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, Dongguan City, Guangdong Province, China
| | - Ye Lin
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangzhou City, Guangdong Province, China
| | - Yingxin He
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China
| | - Jiayang Chen
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China
| | - Xi Liu
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China
| | - Weijie Ye
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China
| | - Zhikang Mo
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China
| | - Zhanpeng She
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China
| | - Shuwen Lin
- Department of General Surgery, Binhaiwan Central Hospital of Dongguan, (also called The Fifth People's Hospital of Dongguan), The Dongguan Affiliated Hospital of Medical College of Jinan University, No.111 Humen Road, Humen Town, Dongguan City, 523905, Guangdong Province, China.
| |
Collapse
|
14
|
LncRNA GSTM3TV2 Promotes Cell Proliferation and Invasion via miR-597/FOSL2 Axis in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3445970. [PMID: 34458365 PMCID: PMC8387164 DOI: 10.1155/2021/3445970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/29/2021] [Indexed: 11/17/2022]
Abstract
Mounting evidence has recently shown that role of long noncoding RNA is critical in many human cancers. lncRNA GSTM3TV2 was first proven to play a vital role in pancreatic cancer. However, the mechanism of lncRNA GSTM3TV2 in hepatocellular carcinoma (HCC) is still uncovered. Here, we object to distinguish the expression of lncRNA GSTM3TV2 and reveal its mechanistic relationship with HCC. We observed that the expression of lncRNA GSTM3TV2 and FOSL2 were upregulated in HCC. Knockdown of lncRNA GSTM3TV2 significantly inhibited cell proliferation. Meanwhile, the migration and invasion of HCC cells were greatly decreased by the downregulated lncRNA GSTM3TV2. The luciferase reporter assays showed that lncRNA GSTM3TV2 could be directly bound to miR-597, and the level of miR-597 was also decreased in the tumor tissues. lncRNA GSTM3TV2 could stabilize FOSL2 expression, resulting in the oncogenic properties of lncRNA GSTM3TV2 in HCC. Our study indicated the oncogenic activities of lncRNA GSTM3TV2 and emphasized the role of the miR-597/FOSL2 signaling pathway.
Collapse
|
15
|
Liao S, He H, Zeng Y, Yang L, Liu Z, An Z, Zhang M. A nomogram for predicting metabolic steatohepatitis: The combination of NAMPT, RALGDS, GADD45B, FOSL2, RTP3, and RASD1. Open Med (Wars) 2021; 16:773-785. [PMID: 34041361 PMCID: PMC8130015 DOI: 10.1515/med-2021-0286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/17/2021] [Accepted: 04/17/2021] [Indexed: 02/05/2023] Open
Abstract
Objective To identify differentially expressed and clinically significant mRNAs and construct a potential prediction model for metabolic steatohepatitis (MASH). Method We downloaded four microarray datasets, GSE89632, GSE24807, GSE63067, and GSE48452, from the Gene Expression Omnibus database. The differentially expressed genes (DEGs) analysis and weighted gene co-expression network analysis were performed to screen significant genes. Finally, we constructed a nomogram of six hub genes in predicting MASH and assessed it through receiver operating characteristic (ROC) curve, calibration plot, and decision curve analysis (DCA). In addition, qRT-PCR was used for relative quantitative detection of RNA in QSG-7011 cells to further verify the expression of the selected mRNA in fatty liver cells. Results Based on common DEGs and brown and yellow modules, seven hub genes were identified, which were NAMPT, PHLDA1, RALGDS, GADD45B, FOSL2, RTP3, and RASD1. After logistic regression analysis, six hub genes were used to establish the nomogram, which were NAMPT, RALGDS, GADD45B, FOSL2, RTP3, and RASD1. The area under the ROC of the nomogram was 0.897. The DCA showed that when the threshold probability of MASH was 0–0.8, the prediction model was valuable to GSE48452. In QSG-7011 fatty liver model cells, the relative expression levels of NAMPT, GADD45B, FOSL2, RTP3, RASD1 and RALGDS were lower than the control group. Conclusion We identified seven hub genes NAMPT, PHLDA1, RALGDS, GADD45B, FOSL2, RTP3, and RASD1. The nomogram showed good performance in the prediction of MASH and it had clinical utility in distinguishing MASH from simple steatosis.
Collapse
Affiliation(s)
- Shenling Liao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - He He
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuping Zeng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lidan Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhi Liu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhenmei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Mei Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
16
|
Li M, Gai F, Chen H. MiR-30b-5p Influences Chronic Exercise Arthritic Injury by Targeting Hoxa1. Int J Sports Med 2021; 42:1199-1208. [PMID: 33930933 DOI: 10.1055/a-1342-7872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We identified the role of miR-30b-5p in chronic exercise arthritic injury. Rats with chronic exercise arthritic injury received treatment with miR-30b-5p antagomiR. H&E and Safranin O-fast green staining were performed. The levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were detected. The binding relationship between homeobox A1 (Hoxa1) and miR-30b-5p was revealed. After manipulating the expressions of miR-30b-5p and/or Hoxa1 in chondrocytes, the viability, apoptosis and migration of chondrocytes were assessed. The levels of molecules were determined by qRT-PCR or Western blot. MiR-30b-5p antagomiR ameliorated articular cartilage lesion and destruction, reduced Mankin's score and the levels of TNF-α, IL-1β, miR-30b-5p, matrix metallopeptidase 13 (MMP-13), and cleaved caspase-3, and increased relative thickness and the levels of Hoxa1, Aggrecan and type II collagen (COLII) in model rats. MiR-30b-5p up-regulation decreased Hoxa1 level, viability, migration and induced apoptosis, whereas miR-30b-5p down-regulation produced the opposite effects. MiR-30b-5p up-regulation increased the levels of MMP-13 and cleaved caspase-3, but decreased those of Aggrecan and COLII in chondrocytes. However, the action of miR-30b-5p up-regulation on chondrocytes was reversed by Hoxa1 overexpression. In conclusion, miR-30b-5p is involved in cartilage degradation in rats with chronic exercise arthritic injury and regulates chondrocyte apoptosis and migration by targeting Hoxa1.
Collapse
Affiliation(s)
- Maoxun Li
- Department of Orthopaedics, The People's Hospital of Jimo.Qingdao, Qingdao, China
| | - Fei Gai
- Department of Radiotherapy, The People's Hospital of Jimo.Qingdao, Qingdao, China
| | - Hongyu Chen
- Department of Emergency, Qingdao West Coast New Area Central Hospital, Qingdao, China
| |
Collapse
|
17
|
Han X, Zhu Y, Shen L, Zhou Y, Pang L, Zhou W, Gu H, Han K, Yang Y, Jiang C, Xie J, Zhang C, Ding L. PTIP Inhibits Cell Invasion in Esophageal Squamous Cell Carcinoma via Modulation of EphA2 Expression. Front Oncol 2021; 11:629916. [PMID: 33833989 PMCID: PMC8021923 DOI: 10.3389/fonc.2021.629916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/05/2021] [Indexed: 12/30/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly aggressive malignancy and treatment failure is largely due to metastasis and invasion. Aberrant tumor cell adhesion is often associated with tumor progression and metastasis. However, the exact details of cell adhesion in ESCC progression have yet to be determined. In our study, the clinical relevance of Pax2 transactivation domain-interacting protein (PTIP/PAXIP1) was analyzed by immunohistochemistry of ESCC tissues. We found that low expression of PTIP was associated with lymph node metastasis in ESCC, and loss-of-function approaches showed that depletion of PTIP promoted ESCC cell migration and invasion both in vitro and in vivo. Analysis integrating RNA-seq and ChIP-seq data revealed that PTIP directly regulated ephrin type-A receptor 2 (EphA2) expression in ESCC cells. Moreover, PTIP inhibited EphA2 expression by competing with Fosl2, which attenuated the invasion ability of ESCC cells. These results collectively suggest that PTIP regulates ESCC invasion through modulation of EphA2 expression and hence presents a potential therapeutic target for its treatment.
Collapse
Affiliation(s)
- Xiao Han
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yaning Zhu
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Li Shen
- Department of Hematology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yu Zhou
- Department of Medical Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Liqun Pang
- Department of General Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Wubi Zhou
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hao Gu
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Kairong Han
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yijun Yang
- Department of Obstetrics and Gynecology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Chao Jiang
- Department of Medical Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Jun Xie
- Department of Clinical Laboratory, Xuyi People's Hospital, Huai'an, China
| | - Chengwan Zhang
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
18
|
Wan X, Guan S, Hou Y, Qin Y, Zeng H, Yang L, Qiao Y, Liu S, Li Q, Jin T, Qiu Y, Liu M. FOSL2 promotes VEGF-independent angiogenesis by transcriptionnally activating Wnt5a in breast cancer-associated fibroblasts. Am J Cancer Res 2021; 11:4975-4991. [PMID: 33754039 PMCID: PMC7978317 DOI: 10.7150/thno.55074] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/24/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs), a predominant component of the tumor microenvironment, contribute to aggressive angiogenesis progression. In clinical practice, traditional anti-angiogenic therapy, mainly anti-VEGF, provides extremely limited beneficial effects to breast cancer. Here, we reveal that FOS-like 2 (FOSL2), a transcription factor in breast CAFs, plays a critical role in VEGF-independent angiogenesis in stromal fibroblasts. Methods: FOSL2 and Wnt5a expression was assessed by qRT-PCR, western blotting and immunohistochemistry in primary and immortalized CAFs and clinical samples. FOSL2- or Wnt5a-silenced CAFs and FOSL2-overexpressing NFs were established to explore their proangiogenic effects. Invasion, tubule formation, three-dimensional sprouting assays, and orthotopic xenografts were conducted as angiogenesis experiments. FZD5/NF-κB/ERK signaling activation was evaluated by western blotting after blocking VEGF/VEGFR with an anti-VEGF antibody and axitinib. Dual luciferase reporter assays and chromatin immunoprecipitation were performed to test the role of FOSL2 in regulating Wnt5a expression, and Wnt5a in the serum of the patients was measured to assess its clinical diagnostic value for breast cancer patients. Results: Enhanced FOSL2 in breast CAFs was significantly associated with angiogenesis and clinical progression in patients. The supernatant from CAFs highly expressing FOSL2 strongly promoted tube formation and sprouting of human umbilical vein endothelial cells (HUVECs) in a VEGF-independent manner and angiogenesis as well as tumor growth in vivo. Mechanistically, the enhanced FOSL2 in CAFs was regulated by estrogen/cAMP/PKA signaling. Wnt5a, a direct target of FOSL2, specifically activated FZD5/NF-κB/ERK signaling in HUVECs to promote VEGF-independent angiogenesis. In addition, a high level of Wnt5a was commonly detected in the serum of breast cancer patients and closely correlated with microvessel density in breast tumor tissues, suggesting a promising clinical value of Wnt5a for breast cancer diagnostics. Conclusion: FOSL2/Wnt5a signaling plays an essential role in breast cancer angiogenesis in a VEGF-independent manner, and targeting the FOSL2/Wnt5a signaling axis in CAFs may offer a potential option for antiangiogenesis therapy.
Collapse
|
19
|
Park AY, Seo BK, Han MR. Breast Ultrasound Microvascular Imaging and Radiogenomics. Korean J Radiol 2021; 22:677-687. [PMID: 33569931 PMCID: PMC8076833 DOI: 10.3348/kjr.2020.1166] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/13/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Microvascular ultrasound (US) techniques are advanced Doppler techniques that provide high sensitivity and spatial resolution for detailed visualization of low-flow vessels. Microvascular US imaging can be applied to breast lesion evaluation with or without US contrast agents. Microvascular US imaging without a contrast agent uses a sophisticated wall filtering system to selectively obtain low-flow Doppler signals from overlapped artifacts. Microvascular US imaging with second-generation contrast agents amplifies flow signals and makes them last longer, which facilitates hemodynamic evaluation of breast lesions. In this review article, we will introduce various microvascular US techniques, explain their clinical applications in breast cancer diagnosis and radiologic-histopathologic correlation, and provide a summary of a recent radiogenomic study using microvascular US.
Collapse
Affiliation(s)
- Ah Young Park
- Department of Radiology, Bundang CHA Medical Center, CHA University, Seongnam, Korea
| | - Bo Kyoung Seo
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea.
| | - Mi Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| |
Collapse
|
20
|
Moussa Agha D, Rouas R, Najar M, Bouhtit F, Naamane N, Fayyad-Kazan H, Bron D, Meuleman N, Lewalle P, Merimi M. Identification of Acute Myeloid Leukemia Bone Marrow Circulating MicroRNAs. Int J Mol Sci 2020; 21:7065. [PMID: 32992819 PMCID: PMC7583041 DOI: 10.3390/ijms21197065] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In addition to their roles in different biological processes, microRNAs in the tumor microenvironment appear to be potential diagnostic and prognostic biomarkers for various malignant diseases, including acute myeloid leukemia (AML). To date, no screening of circulating miRNAs has been carried out in the bone marrow compartment of AML. Accordingly, we investigated the circulating miRNA profile in AML bone marrow at diagnosis (AMLD) and first complete remission post treatment (AMLPT) in comparison to healthy donors (HD). METHODS Circulating miRNAs were isolated from AML bone marrow aspirations, and a low-density TaqMan miRNA array was performed to identify deregulated miRNAs followed by quantitative RT-PCR to validate the results. Bioinformatic analysis was conducted to evaluate the diagnostic and prognostic accuracy of the highly and significantly identified deregulated miRNA(s) as potential candidate biomarker(s). RESULTS We found several deregulated miRNAs between the AMLD vs. HD vs. AMLPT groups, which were involved in tumor progression and immune suppression pathways. We also identified significant diagnostic and prognostic signatures with the ability to predict AML patient treatment response. CONCLUSIONS This study provides a possible role of enriched circulating bone marrow miRNAs in the initiation and progression of AML and highlights new markers for prognosis and treatment monitoring.
Collapse
Affiliation(s)
- Douâa Moussa Agha
- Laboratory of Experimental Hematology, Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (D.M.A.); (R.R.); (F.B.); (H.F.-K.); (D.B.); (P.L.)
| | - Redouane Rouas
- Laboratory of Experimental Hematology, Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (D.M.A.); (R.R.); (F.B.); (H.F.-K.); (D.B.); (P.L.)
| | - Mehdi Najar
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, Montreal, QC H2X 0A9, Canada;
- Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Fatima Bouhtit
- Laboratory of Experimental Hematology, Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (D.M.A.); (R.R.); (F.B.); (H.F.-K.); (D.B.); (P.L.)
- Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Najib Naamane
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Hussein Fayyad-Kazan
- Laboratory of Experimental Hematology, Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (D.M.A.); (R.R.); (F.B.); (H.F.-K.); (D.B.); (P.L.)
| | - Dominique Bron
- Laboratory of Experimental Hematology, Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (D.M.A.); (R.R.); (F.B.); (H.F.-K.); (D.B.); (P.L.)
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Philippe Lewalle
- Laboratory of Experimental Hematology, Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (D.M.A.); (R.R.); (F.B.); (H.F.-K.); (D.B.); (P.L.)
| | - Makram Merimi
- Laboratory of Experimental Hematology, Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (D.M.A.); (R.R.); (F.B.); (H.F.-K.); (D.B.); (P.L.)
- Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| |
Collapse
|
21
|
Gao L, Jin HJ, Zhang D, Lin Q. Silencing circRNA_001937 may inhibit cutaneous squamous cell carcinoma proliferation and induce apoptosis by preventing the sponging of the miRNA‑597‑3p/FOSL2 pathway. Int J Mol Med 2020; 46:1653-1660. [PMID: 33000177 PMCID: PMC7521585 DOI: 10.3892/ijmm.2020.4723] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
Circular RNAs (circRNAs) are reported to be aberrantly expressed and perform different functions in numerous types of tumor; however, their expression levels in cutaneous squamous cell carcinoma (CSCC) remain largely unclear. Thus, the purpose of the present study was to investigate the function of circRNA_001937 in CSCC. Differential circRNA expression profiles of CSCC were analyzed using the Arraystar Human circRNAs chip and reverse transcription-quantitative PCR (RT-qPCR); and the effects of circRNA_001937 on cell behavior, in particular its regulation over the microRNA (miRNA)-597-3p/Fos-related antigen 2 (FOSL2) pathway, was investigated using a dual-luciferase reporter assay, and verified using RT-qPCR and western blotting. circRNA_001937 expression levels were significantly increased in CSCC tissues and cell lines compared with the corresponding adjacent tissues and control cells (P<0.05). The genetic silencing of circRNA_001937 with small interfering RNA significantly inhibited cell proliferation, and induced cell apoptosis (P<0.05). circRNA_001937 was observed to directly bind to miRNA-597-3p and serve as a sponge, which indirectly increased the expression levels of FOSL2, a miRNA-597-3p target gene. In conclusion, circRNA_001937 expression was increased in CSCC and silencing circRNA_001937 gene expression may inhibit CSCC progression by sponging the miRNA-597-3p/FOSL2 pathway.
Collapse
Affiliation(s)
- Ling Gao
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hong-Juan Jin
- Department of Plastic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Duo Zhang
- Department of Plastic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Quan Lin
- Department of Plastic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
22
|
Li S, Li Y, Deng B, Yan J, Wang Y. Identification of the Differentially Expressed Genes Involved in the Synergistic Neurotoxicity of an HIV Protease Inhibitor and Methamphetamine. Curr HIV Res 2020; 17:290-303. [PMID: 31550215 DOI: 10.2174/1570162x17666190924200354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/23/2019] [Accepted: 09/05/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND The abuse of psychostimulants such as methamphetamine (METH) is common in human immunodeficiency virus (HIV)-infected individuals. Acquired immunodeficiency syndrome (AIDS) patients taking METH and antiretroviral drugs could suffer severe neurologic damage and cognitive impairment. OBJECTIVE To reveal the underlying neuropathologic mechanisms of an HIV protease inhibitor (PI) combined with METH, growth-inhibition tests of dopaminergic cells and RNA sequencing were performed. METHODS A combination of METH and PI caused more growth inhibition of dopaminergic cells than METH alone or a PI alone. Furthermore, we identified differentially expressed gene (DEG) patterns in the METH vs. untreated cells (1161 genes), PI vs. untreated cells (16 genes), METH-PI vs. PI (3959 genes), and METH-PI vs. METH groups (14 genes). RESULTS The DEGs in the METH-PI co-treatment group were verified in the brains of a mouse model using quantitative polymerase chain reaction and were involved mostly in the regulatory functions of cell proliferation and inflammation. CONCLUSION Such identification of key regulatory genes could facilitate the study of their neuroprotective potential in the users of METH and PIs.
Collapse
Affiliation(s)
- Sangsang Li
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Immunology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yanfei Li
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Immunology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bingpeng Deng
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Wang
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
23
|
miR-597-5p inhibits cell growth and promotes cell apoptosis by targeting ELK1 in pancreatic cancer. Hum Cell 2020; 33:1165-1175. [PMID: 32613573 DOI: 10.1007/s13577-020-00395-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/20/2020] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer is a malignant disease with poor prognosis. Emerging evidences have showed that miR-597-5p is closely related to tumor development. However, the functional roles of miR-597-5p in pancreatic cancer remain unknown. This study aimed to investigate the expression of miR-597-5p in pancreatic cancer tissues and cells, and explored its regulatory mechanism during pancreatic cancer progression. Pancreatic cancer and adjacent tissues were obtained to detect the expression of miR-597-5p by RT-qPCR. Cell growth, apoptosis, and related protein expression were, respectively, tested by CCK-8 assay, cell formation, wound healing, Transwell assay, flow cytometry, and western blotting. Finally, the pancreatic cancer mice model was constructed. In vitro and in vivo results showed that miR-597-5p expression was down-regulated in pancreatic cancer tissues and cell lines, and increased the overall survival of pancreatic cancer patients. Moreover, miR-597-5p decreased pancreatic cancer cell viability, reduced relative wound width, suppressed colony formation and decreased invasive cell number, as well as reduced the expression of proliferating cell nuclear antigen (PCNA), Ki67, Cyclin D1, N-cad, and Bcl-2. Meanwhile, it increased pancreatic cancer cell apoptosis and the expression of E-cad, cleaved caspase 3, and Bax. The dual-luciferase reporter assay confirmed miR-597-5p could directly target e-twenty six like-1 (ELK1) oncogene. The reduction of cell growth and the induction of cell apoptosis induced by miR-597-5p were reversed by ELK1. In addition, miR-597-5p inhibited the growth of pancreatic cancer in vivo. This study demonstrated that miR-597-5p may be a novel suppressor of pancreatic cancer. It inhibits pancreatic cancer cell growth and promotes apoptosis by the down-regulation of ELK1 in vitro and in vivo.
Collapse
|
24
|
Zhang S, Yu J, Sun BF, Hou GZ, Yu ZJ, Luo H. MicroRNA-92a Targets SERTAD3 and Regulates the Growth, Invasion, and Migration of Prostate Cancer Cells via the P53 Pathway. Onco Targets Ther 2020; 13:5495-5514. [PMID: 32606766 PMCID: PMC7298502 DOI: 10.2147/ott.s249168] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/04/2020] [Indexed: 12/22/2022] Open
Abstract
Background The miR-17-92 cluster, consisting of six mature miRNAs including miR-17, miR-18a, miR-19a, miR-19b, miR-20a, and miR-92a, plays a key role in the tumorigenesis and development of various cancers. The dysregulation of the cluster correlates with the biological mechanism of tumor growth and metastasis in vivo. However, the relationship between miR-17-92 cluster and malignancy of prostate cancer remains unclear, and its regulatory mechanism is worth investigating for controlling the proliferation and invasion of prostate cancer. Materials and Methods The expressions of miR-17-92 cluster members were measured using real-time quantitative RT-PCR. WB and real-time quantitative RT-PCR were used to detect the expression of SERTAD3, p38, p21, p53 protein levels and transcription levels. Cell proliferation and apoptosis were evaluated using cell proliferation assay, EdU and Hoechst assay, colony formation experiment and flow cytometry analyses. Cell migration and invasion were determined via transwell assays. The TargetScan, miRDB, starBase databases and luciferase reporter assays were used to confirm the target gene of miR-92a. Results The relative expression of miR-92a was threefold higher in the metastatic PC-3 cells compared with the non-metastatic LNCaP cells. Down-regulation of miR-92a in PC-3 cells led to the inhibition of cell proliferation, migration, and invasion, while its overexpression in LNCaP cells resulted in the promotion of cell proliferation, migration, and invasion. The role of SERTAD3 in prostate cancer can be alleviated by miR-92a inhibitor. Conclusion SERTAD3 was the direct target gene of miR-92a in prostate cancer cells; inhibition of SERTAD3-dependent miR-92a alleviated the growth, invasion, and migration of prostate cancer cells by regulating the expression of the key genes of the p53 pathway, including p38, p53 and p21. These results suggested that targeting SERTAD3 by the induction of overexpression of miR-92a may be a treatment option in prostate cancer.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, People's Republic of China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, People's Republic of China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, People's Republic of China.,Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, People's Republic of China
| | - Bao-Fei Sun
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, People's Republic of China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, People's Republic of China
| | - Gui-Zhong Hou
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, People's Republic of China
| | - Zi-Jiang Yu
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, People's Republic of China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, People's Republic of China
| | - Heng Luo
- Department of Anatomy, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, People's Republic of China.,State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, People's Republic of China.,Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, People's Republic of China
| |
Collapse
|
25
|
Chen H, Wahafu P, Wang L, Chen X. LncRNA LINC00313 Knockdown Inhibits Tumorigenesis and Metastasis in Human Osteosarcoma by Upregulating FOSL2 through Sponging miR-342-3p. Yonsei Med J 2020; 61:359-370. [PMID: 32390359 PMCID: PMC7214116 DOI: 10.3349/ymj.2020.61.5.359] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/31/2019] [Accepted: 12/09/2019] [Indexed: 01/29/2023] Open
Abstract
PURPOSE Osteosarcoma (OS) is the most common primary bone tumor, with high morbidity in infants and adolescents. Long noncoding RNA LINC00313 has been found to modulate papillary thyroid cancer tumorigenesis and to be dysregulate in lung cancer. However, the role of LINC00313 in OS has not yet been addressed. MATERIALS AND METHODS We evaluated mRNA and protein expression using real-time quantitative PCR and Western blotting. Cell proliferation was evaluated using MTT; apoptosis and autophagy were assessed with flow cytometry, Western blotting, and/or GFP-LC3 assay. Transwell assay was conducted to measure cell migration and invasion. Potential target sites for LINC00313 and miR-342-3p were predicted with starBase v.2.0 and TargetScan Human, and verified using luciferase reporter assay, RNA immunoprecipitation, and RNA pull-down assay. In vivo, xenogeneic tumors were induced with U2OS and MG-63 cells, separately. RESULTS LINC00313 was upregulated and miR-342-3p was downregulated in OS tissues and cells. High expression of LINC00313 was associated with shorter overall survival. FOSL2 downregulation and miR-342-3p overexpression suppressed cell proliferation and migratory and invasive abilities while promoting apoptosis and autophagy, all of which were consistent with the effects of LINC00313 knockdown. miR-342-3p, sponged by LINC00313, inversely modulated FOSL2 by targeting MG-63 cells, and FOSL2 expression was positively controlled by LINC00313. LINC00313 knockdown suppressed tumor growth in vivo. CONCLUSION LINC00313 is upregulated in OS, and LINC00313 knockdown plays a vital anti-tumor role in OS cell progression through a miR-342-3p/FOSL2 axis. Our study suggests that LINC00313 may be a novel, promising biomarker for diagnosis and prognosis of OS.
Collapse
Affiliation(s)
- Hongtao Chen
- Department of Orthopedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China.
| | - Paerhati Wahafu
- Department of Orthopedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Leilei Wang
- Department of Orthopedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Xuan Chen
- Department of Orthopedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| |
Collapse
|
26
|
Li Z, Liu Y, Yan J, Zeng Q, Hu Y, Wang H, Li H, Li J, Yu Z. Circular RNA hsa_circ_0056836 functions an oncogenic gene in hepatocellular carcinoma through modulating miR-766-3p/FOSL2 axis. Aging (Albany NY) 2020; 12:2485-2497. [PMID: 32048611 PMCID: PMC7041754 DOI: 10.18632/aging.102756] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Circular RNA (circRNA) remains a tumour-related factor in various biological cells and plays regulatory roles in gene expression. It is a type of non-coding RNA, whereas the function of human circRNA in hepatocellular carcinoma (HCC) is still not clear yet. Our investigation used the HCC cell line to uncover the biological function of hsa_circ_0056836 in the development and progression of hepatocellular carcinoma. RESULTS The present study showed that hsa_circ_0056836 revealed high expressions in HCC cell lines and tissues compared with corresponding controls. Silencing of Hsa_circ_0056836 decreased cell migration, proliferation and invasion. Silencing of hsa_circ_0056836 inhibited the development of HCC in xenograft nude mice. Mechanistically, we found that hsa_circ_0056836 directly bound to miR-766-3p, thereby alleviating the targeted inhibition of Fos-related antigen 2 (FOSL2). The results of this study indicated that hsa_circ_0056836 is a novel oncogenic RNA of vast potential as a tumor biomarker. CONCLUSION In summary, the hsa_circ_0056836 / miR-766-3p / FOSL2 axis may serve as a promising strategy for HCC treatment. METHOD First, the expressions of hsa_circ_0056836 in HCC tissues and corresponding para-cancerous tissues as well as in HCC cell lines and normal hepatocytes THLE-3 were detected by RT-PCR. Subcellular localization of hsa_circ_0056836 was confirmed by FISH. To detect the association between hsa_circ_0056836 and miR-766-3p, AGO2-RIP and Luciferase reporter assay were adopted. Loss of function study was applied to assess the role of hsa_circ_0056836 in HCC and to determine tumorigenesis in nude mice.
Collapse
Affiliation(s)
- Zhiqin Li
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yang Liu
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jingya Yan
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Qinglei Zeng
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yushu Hu
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hongyan Wang
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hua Li
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Juan Li
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zujiang Yu
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
27
|
Park AY, Han MR, Park KH, Kim JS, Son GS, Lee HY, Chang YW, Park EK, Cha SH, Cho Y, Hong H, Cho KR, Song SE, Woo OH, Lee JH, Cha J, Seo BK. Radiogenomic Analysis of Breast Cancer by Using B-Mode and Vascular US and RNA Sequencing. Radiology 2020; 295:24-34. [PMID: 32013793 DOI: 10.1148/radiol.2020191368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Radiogenomic investigations for breast cancer provide an understanding of tumor heterogeneity and discover image phenotypes of genetic variation. However, there is little research on the correlations between US features of breast cancer and whole-transcriptome profiling. Purpose To explore US phenotypes reflecting genetic alteration relevant to breast cancer treatment and prognosis by comparing US images of tumor with their RNA sequencing results. Materials and Methods From January to October 2016, B-mode and vascular US images in 31 women (mean age, 49 years ± 9 [standard deviation]) with breast cancer were prospectively analyzed. B-mode features included size, shape, echo pattern, orientation, margin, and calcifications. Vascular features were evaluated by using microvascular US and contrast agent-enhanced US: vascular index, vessel morphologic features, distribution, penetrating vessels, enhancement degree, order, margin, internal homogeneity, and perfusion defect. RNA sequencing was conducted with total RNA obtained from a surgical specimen by using next-generation sequencing. US features were compared with gene expression profiles, and ingenuity pathway analysis was used to analyze gene networks, enriched functions, and canonical pathways associated with breast cancer. The P value for differential expression was extracted by using a parametric F test comparing nested linear models. Results Thirteen US features were associated with various patterns of 340 genes (P < .05). Nonparallel orientation at B-mode US was associated with upregulation of TFF1 (log twofold change [log2FC] = 4.0; P < .001), TFF3 (log2FC = 2.5; P < .001), AREG (log2FC = 2.6; P = .005), and AGR3 (log2FC = 2.6; P = .003). Complex vessel morphologic structure was associated with upregulation of FZD8 (log2FC = 2.0; P = .01) and downregulation of IGF1R (log2FC = -2.0; P = .006) and CRIPAK (log2FC = -2.4; P = .01). The top networks with regard to orientation or vessel morphologic structure were associated with cell cycle, death, and proliferation. Conclusion Compared with RNA sequencing, B-mode and vascular US features reflected genomic alterations associated with hormone receptor status, angiogenesis, or prognosis in breast cancer. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ah Young Park
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Mi-Ryung Han
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Kyong Hwa Park
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Jung Sun Kim
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Gil Soo Son
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Hye Yoon Lee
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Young Woo Chang
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Eun Kyung Park
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Sang Hoon Cha
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Yunjung Cho
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Hyosun Hong
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Kyu Ran Cho
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Sung Eun Song
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Ok Hee Woo
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Ju-Han Lee
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Jaehyung Cha
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Bo Kyoung Seo
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| |
Collapse
|
28
|
Guo J, Shen S, Liu X, Ruan X, Zheng J, Liu Y, Liu L, Ma J, Ma T, Shao L, Wang D, Yang C, Xue Y. Role of linc00174/miR-138-5p (miR-150-5p)/FOSL2 Feedback Loop on Regulating the Blood-Tumor Barrier Permeability. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:1072-1090. [PMID: 31791014 PMCID: PMC6906710 DOI: 10.1016/j.omtn.2019.10.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 09/27/2019] [Accepted: 10/26/2019] [Indexed: 01/11/2023]
Abstract
The blood-tumor barrier (BTB) limits the transport of chemotherapeutic drugs to brain tumor tissues and impacts the treatment of glioma. Long non-coding RNAs play critical roles in various biological processes of tumors; however, the function of these in BTB permeability is still unclear. In this study, we have identified that long intergenic non-protein coding RNA 174 (linc00174) was upregulated in glioma endothelial cells (GECs) from glioma tissues. Additionally, linc00174 was also upregulated in GECs from the BTB model in vitro. Knock down of linc00174 increased BTB permeability and reduced the expression of the tight junction-related proteins ZO-1, occludin, and claudin-5. Both bioinformatics data and results of luciferase reporter assays demonstrated that linc00174 regulated BTB permeability by binding to miR-138-5p and miR-150-5p. Furthermore, knock down of linc00174 inhibited FOSL2 expression via upregulating miR-138-5p and miR-150-5p. FOSL2 interacted with the promoter regions and upregulated the promoter activity of ZO-1, occludin, claudin-5, and linc00174 in GECs. In conclusion, the present study demonstrated that the linc00174/miR-138-5p (miR-150-5p)/FOSL2 feedback loop played an essential role in regulating BTB permeability.
Collapse
Affiliation(s)
- Jizhe Guo
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Shuyuan Shen
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China; Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang 110004, People's Republic of China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China; Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang 110004, People's Republic of China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China; Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang 110004, People's Republic of China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Jun Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Teng Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Lianqi Shao
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China; Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang 110004, People's Republic of China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China; Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang 110004, People's Republic of China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China.
| |
Collapse
|
29
|
Li S, Liu Z, Fang XD, Wang XY, Fei BY. MicroRNA (miR)-597-5p Inhibits Colon Cancer Cell Migration and Invasion by Targeting FOS-Like Antigen 2 ( FOSL2). Front Oncol 2019; 9:495. [PMID: 31245295 PMCID: PMC6581747 DOI: 10.3389/fonc.2019.00495] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/24/2019] [Indexed: 12/18/2022] Open
Abstract
Our previous work had shown that FOS-like antigen 2 (FOSL2) is regulated by miR-143-5p in colorectal cancer (CRC). Given that it has been shown by others that FOSL2 is also a target of miR-597-5p in breast adenocarcinoma, the objective of the current work was to determine whether FOSL2 is regulated by miR-597-5p in CRC and the role of miR-597-5p in CRC. MiR-597-5p expression was determined in RNA obtained from 30 paired samples of colon cancer and tumor adjacent normal tissue, as well as in the LoVo (CRC cell line) and FHC (normal colonic epithelial cells) by quantitative real time polymerase chain reaction (qRT-PCR). MiR-597-5p expression was significantly downregulated in both CRC tissue and LoVo cells. Reporter assays using wild-type and miR-597-5p seed mutant FOSL2 confirmed that FOSL2 is a bona fide target of miR-597-5p. Modulating miR-597-5p expression levels in FHC and LoVo cells using antagomir and mimic, respectively, impacted expression of epithelial and mesenchymal cell markers as well as in vitro migration and invasion, without any effect on cell proliferation, showing that miR-597-5p functions as a suppressor of epithelial to mesenchymal transition. Restoration of FOSL2 expression rescued pro-metastatic functional properties of LoVo cells conforming that effect of miR-597-5p was being mediated by targeting FOSL2. Xenograft assays in athymic nude mice showed that miR-597-5p mimic did not reduce tumor incidence or growth in LoVo cells. However, using a hepatic metastasis model showed that miR-597-5p mimic can significantly prevent hepatic metastatic nodule formation as well as FOSL2 expression in these metastatic nodules. Importantly, FOSL2 mRNA and miR-597-5p expression was found to be inversely correlated in an independent cohort of 21 CRC patients Cumulatively our results show that miR-597-5p functions as a suppressor of metastatic progression in CRC by targeting FOSL2. Replenishment of miR-597-5p can be a potential therapeutic target where its expression along with FOSL2 can serve as potential diagnostic markers in CRC.
Collapse
Affiliation(s)
- Shuo Li
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhuo Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xue-Dong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiu-Ying Wang
- Medical Record Department, China Japan Union Hospital of Jilin University, Changchun, China
| | - Bing-Yuan Fei
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Agrawal M, Rastogi M, Dogra S, Pandey N, Basu A, Singh SK. Chandipura virus changes cellular miRNome in human microglial cells. J Med Virol 2019; 94:480-490. [PMID: 31017674 DOI: 10.1002/jmv.25491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/17/2019] [Accepted: 04/21/2019] [Indexed: 12/16/2022]
Abstract
Chandipura virus (CHPV) is a neurotropic virus, known to cause encephalitis in humans. The microRNAs (miRNA/miR) play an important role in the pathogenesis of viral infection. The present study is focused on the role of miRNAs during CHPV (strain 1653514) infection in human microglial cells. The deep sequencing of CHPV-infected human microglial cells identified a total of 12 differentially expressed miRNA (DEMs). To elucidate the role of DEMs, the target gene prediction, Gene Ontology term (GO Term), pathway enrichment analysis, and miRNA-messenger RNA (mRNA) interaction network analysis was performed. The GO terms and pathway enrichment analysis provided 146 enriched genes; which were involved in interferon response, cytokine and chemokine signaling. Further, the WGCNA (weighted gene coexpression network analysis) of the enriched genes were discretely categorized into three modules (blue, brown, and turquoise). The hub genes in the blue module may correlate to CHPV induced neuroinflammation. Altogether, the miRNA-mRNA interaction network and WGCNA study revealed the following pairs, hsa-miR-542-3p and FAF1, hsa-miR-92a-1-5p and MYD88, and hsa-miR-3187-3p and TNFRSF21, which may contribute to neuroinflammation during CHPV infection in human microglial cells.
Collapse
Affiliation(s)
- Meghna Agrawal
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Meghana Rastogi
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Smriti Dogra
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Neha Pandey
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Anirban Basu
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, India
| | - Sunit K Singh
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
31
|
Sun L, Guo Z, Sun J, Li J, Dong Z, Zhang Y, Chen J, Kan Q, Yu Z. MiR-133a acts as an anti-oncogene in Hepatocellular carcinoma by inhibiting FOSL2 through TGF-β/Smad3 signaling pathway. Biomed Pharmacother 2018; 107:168-176. [DOI: 10.1016/j.biopha.2018.07.151] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 01/27/2023] Open
|
32
|
Ling L, Zhang SH, Zhi LD, Li H, Wen QK, Li G, Zhang WJ. MicroRNA-30e promotes hepatocyte proliferation and inhibits apoptosis in cecal ligation and puncture-induced sepsis through the JAK/STAT signaling pathway by binding to FOSL2. Biomed Pharmacother 2018; 104:411-419. [PMID: 29787988 DOI: 10.1016/j.biopha.2018.05.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 05/02/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Hepatocyte proliferation and apoptosis are critical cellular behaviors in rat liver as a result of a liver injury. Herein, we performed this study in order to evaluate the role of miR-30e and its target Fos-Related Antigen-2 (FOSL2) in septic rats through the JAK/STAT signaling pathway. METHODS Rat models of sepsis were induced by cecal ligation and puncture. Enzyme-linked immunosorbent assay (ELISA) was performed to access serum levels of lipopolysaccharide (LPS), inflammatory factors, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) to confirm the successful establishment of the model. The hepatocytes were subject to miR-30e mimics, miR-30e inhibitors or siRNA-FOSL2. The expressions of miR-30e, FOSL2, apoptosis- and, JAK/STAT signaling pathway-related genes in liver tissues and hepatocytes were determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. MTT assay and flow cytometry were performed to evaluate hepatocyte viability and apoptosis, respectively. RESULTS The results obtained revealed that in the septic rats, serum levels of inflammatory factors, LPS, ALT and AST, as well as the expression of FOSL2 were elevated and the JAK/STAT signaling pathway was activated, while there was a reduction in the expression of miR-30e. An initial bioinformatics prediction followed by a confirmatory dual-luciferase reporter assay determined that miR-30e targeted and negatively regulated FOSL2 expression. MiR-30e inhibited the activation of JSK2/STAT3 signaling pathway by reducing FOSL2 expression, while miR-30e enhanced hepatocyte proliferation and decreased hepatocyte cell apoptosis in septic rats. CONCLUSION These findings indicated that miR-30e may serve as an independent therapeutic target for sepsis, due to its ability to inhibit apoptosis and induce proliferation of hepatocytes by targeted inhibition of FOSL2 through the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Lan Ling
- Emergency Department, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Shan-Hong Zhang
- Emergency Department, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Li-Da Zhi
- Emergency Department, China-Japan Friendship Hospital, Beijing 100029, PR China.
| | - Hong Li
- Department of Vascular Surgery, Jilin University, Changchun 130012, PR China
| | - Qian-Kuan Wen
- Emergency Department, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Gang Li
- Emergency Department, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Wen-Jia Zhang
- Emergency Department, China-Japan Friendship Hospital, Beijing 100029, PR China
| |
Collapse
|
33
|
miR-143-3p inhibits the proliferation, migration and invasion in osteosarcoma by targeting FOSL2. Sci Rep 2018; 8:606. [PMID: 29330462 PMCID: PMC5766605 DOI: 10.1038/s41598-017-18739-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/16/2017] [Indexed: 12/22/2022] Open
Abstract
Osteosarcoma (OS) is the most common type of primary malignant bone tumor and mainly occurs in children and adolescent. Because of its early migration and invasion, OS has a poor prognosis. It has been reported that mircoRNAs (miRNAs) play a crucial role in the occurrence and development of multiple tumors. In this study, we identified the aberrant-expression of miR-143-3p in osteosarcoma and examined the role of miR-143-3p in OS development. Further, we searched the miR-143-3p target gene and verified its accuracy by luciferase experiments. Finally, we explored the relationship between miR-143-3p and FOS-Like antigen 2 (FOSL2). Our data indicated that miR-143-3p expression was substantially lower in OS tissues and cell-line compared with normal tissues, and was lower in patients with poor prognosis. In addition miR-143-3p inhibited OS cell proliferation and metastasis while promoting apoptosis. We next showed that FOSL2 was directly targeted by miR-143-3p and could reverse the inhibition caused by miR-143-3p. Finally, we found FOSL2 expression in OS cells was significantly higher compared with normal cells and negatively correlated with miR-143-3p. Thus, miR-143-3p directly and negatively targets FOSL2 to affect OS characteristics. This provides a new target for the treatment of OS and deserves further study.
Collapse
|
34
|
Gao Z, Wang L, Wang J, Yang F, Qu J. Molecular mechanism of miR-181b in heart disease due to pregnancy-induced hypertension syndrome. Exp Ther Med 2017; 14:2953-2959. [PMID: 28912851 PMCID: PMC5585881 DOI: 10.3892/etm.2017.4882] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 01/06/2017] [Indexed: 12/26/2022] Open
Abstract
The present study aimed to investigate the molecular mechanisms of microRNA (miR)-181b in heart disease due to hypertensive disorders complicating pregnancy (HDCP) through regulating the expression of metallopeptidase inhibitor 3 (TIMP3). miR-181b expression was detected by reverse transcription-quantitative polymerase chain reaction in peripheral blood samples from patients with HDCP. These samples were analyzed for clinical pathological characteristics. The primary cardiomyocytes of rats were cultured in hypoxic conditions for 24 h, in which miR-181b expression was detected at different time points. The expression of TIMP3 was assessed in normal rat cardiomyocytes following transfection with miR-181b mimics by western blot analysis. The TIMP3 protein was also detected in cardiomyocytes following transfection with TIMP3 short interfering-RNA. The apoptosis rate of transfected cardiomyocytes was detected by flow cytometry following 24 h of culture in a hypoxic environment. Luciferase assay was applied to validate whether miR-181b binds to the 3' untranslated region of TIMP3 mRNA. miR-181b expression was significantly downregulated in the peripheral blood of patients with HDCP and the miR-181b expression was negatively associated with the grades of hypertension (P<0.05). The results of luciferase assay indicated that miR-181b directly targets TIMP3. The apoptosis rates of rat cardiomyocytes in the group transfected with miR-181b or TIMP3 siRNA was significantly lower than the normal control group (P<0.05). miR-181b may inhibit apoptosis of cardiomyocytes to protect myocardial cells through directly targeting TIMP3 genes, which serve important roles in HDCP.
Collapse
Affiliation(s)
- Zheng Gao
- Department of Obstetrics and Gynecology, Laiwu City People's Hospital, Laiwu, Shandong 271100, P.R. China
| | - Li Wang
- Department of Gynecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Jinyun Wang
- Department of Obstetrics and Gynecology, Laiwu City People's Hospital, Laiwu, Shandong 271100, P.R. China
| | - Fengyong Yang
- Intensive Care Unit (ICU), Laiwu City People's Hospital, Laiwu, Shandong 271100, P.R. China
| | - Jin Qu
- Ministry of Painless Endoscopy, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| |
Collapse
|
35
|
Xu S, Zhao C, Jia Z, Wang X, Han Y, Yang Z. Down-regulation of PARP1 by miR-891b sensitizes human breast cancer cells to alkylating chemotherapeutic drugs. Arch Gynecol Obstet 2017; 296:543-549. [PMID: 28660502 DOI: 10.1007/s00404-017-4444-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 06/22/2017] [Indexed: 11/30/2022]
Abstract
PURPOSE Breast cancer is the most common invasive type of cancer among women. Role of different microRNAs (miRNAs) and poly(ADP-ribose) polymerases (PARPs) in breast cancer has been well established. This study aimed to explore the effects of miR-891b on sensitizing breast cancer cells to alkylating chemotherapeutic drugs through PARPs. METHODS The expression of miR-891b and PARP1 in human breast cancer cells HCC1806 was overexpressed by transfection with their mimics or expressing vector. Then, the transfected cells were exposed to 40 µM N-methyl-N-nitro-N-nitrosoguanidine (MNNG) for 1 h. The correlation between miR-891b and PARP1 was detected by RT-qPCR, western blot, and dual-luciferase reporter assay. Besides, MTT assay and Annexin V assay were done to measure cell proliferation and apoptosis, respectively. RESULTS PARP1 was a target of miR-891b, and it was negatively regulated by miR-891b. MiR-891b increased the sensitivity of the HCC1806 cells to the cytotoxic effects of MNNG through suppressing cell proliferation and increasing the percentage of apoptotic cells. Restoration of PARP1 activity in the HCC1806 cells led to loss of miR-891b mediated sensitivity of the HCC1806 cells to MNNG. CONCLUSION MiR-891b increases the sensitivity of the breast cancer cells (HCC1806) to the cytotoxic effects of the chemotherapeutic agent MNNG by suppressing the expression of PARP1.
Collapse
Affiliation(s)
- Shujian Xu
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China.,Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China
| | - Cui Zhao
- Department of Rehabilitation Medicine, The Affiliated Hospital to Binzhou Medical University, Binzhou, 256603, China
| | - Zhongming Jia
- Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China
| | - Xilong Wang
- Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China
| | - Yong Han
- Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China
| | - Zhenlin Yang
- Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China.
| |
Collapse
|