1
|
Wei J, Wang X, Yu D, Tu Y, Yu Y. MicroRNA-mediated autophagy and drug resistance in cancer: mechanisms and therapeutic strategies. Discov Oncol 2024; 15:662. [PMID: 39549162 PMCID: PMC11569378 DOI: 10.1007/s12672-024-01525-9] [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: 07/29/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
This paper provides an exhaustive overview of the intricate interplay between microRNAs (miRNAs) and autophagy in the context of human cancers, underscoring the pivotal role these non-coding RNAs play in modulating autophagic pathways and their implications for cancer development, progression, and resistance to therapy. MiRNAs, as critical regulators of gene expression post-transcription, influence various biological processes, including autophagy, a catabolic mechanism essential for cellular homeostasis, stress response, and survival. The review meticulously delineates the mechanisms through which miRNAs impact autophagy by targeting specific genes and signaling pathways, thereby affecting cancer cell proliferation, metastasis, and response to chemotherapy. It highlights several miRNAs with dual roles, acting either as oncogenes or tumor suppressors based on the cellular context and the specific autophagic pathways they regulate. The paper further explores the therapeutic potential of targeting miRNA-autophagy axis, offering insights into novel strategies for cancer treatment through modulation of this axis. Emphasizing the complexity of the miRNA-autophagy relationship, the review calls for more in-depth studies to unravel the nuanced regulatory networks between miRNAs and autophagy in cancer, which could pave the way for the development of innovative therapeutic interventions and diagnostic tools.
Collapse
Affiliation(s)
- Jinxing Wei
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China
| | - Xianghui Wang
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China
| | - Duo Yu
- Department of Biopharmaceutics School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yanyang Tu
- Research Center, The Huizhou Central People's Hospital, Guangdong Medical University, No. 41 Eling North Road, Huizhou, Guangdong, China.
| | - Yaoyu Yu
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China.
| |
Collapse
|
2
|
Tian Y, Han W, Fu L, Zhang J, Zhou X. IGF2 is upregulated by its antisense RNA to potentiate pancreatic cancer progression. Funct Integr Genomics 2023; 23:348. [PMID: 38036690 DOI: 10.1007/s10142-023-01277-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/24/2023] [Accepted: 11/26/2023] [Indexed: 12/02/2023]
Abstract
Pancreatic cancer is a deadly cancer. More and more long noncoding RNAs (lncRNAs) have received confirmation to be dysregulated in tumors and exert the regulatory function. Studies have suggested that lncRNA insulin-like growth factor 2 antisense RNA (IGF2-AS) participates in the development of some cancers. Thus, we attempted to clarify its function in pancreatic cancer. Reverse-transcription quantitative polymerase chain reaction was applied for testing IGF2-AS expression in pancreatic cancer cells. Colony formation and Transwell wound experiments were applied for determining cell proliferative, migratory, and invasive capabilities. The alteration of epithelial-mesenchymal transition (EMT)-related gene level was tested via western blot. The mice model was established for measuring the tumor growth and metastasis. RIP validated the interaction of RNAs. IGF2-AS displays high expression in pancreatic cancer cells. IGF2-AS depletion repressed PC cell proliferative, migratory, invasive capabilities, and EMT process. Furthermore, pancreatic cancer tumor growth and metastasis were also inhibited by IGF2-AS depletion. Additionally, IGF2-AS positively regulated IGF2 level via recruiting HNRNPC. IGF2 overexpression counteracted the functions of IGF2-AS deficiency on pancreatic cancer cell behaviors. Moreover, IGF2R deletion was found to inhibit the positive effect of IGF2 on pancreatic cancer progression. IGF2-AS potentiates pancreatic cancer cell proliferation, tumor growth, and metastasis by recruiting HNRNPC via the IGF2-IGF2R regulatory pathway. These discoveries might offer a novel insight for treatment of PC, which may facilitate targeted therapies of PC in clinical practice.
Collapse
Affiliation(s)
- Yuan Tian
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315100, China
| | - Wenwen Han
- Department of Emergency, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315100, China
| | - Long Fu
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315100, China
| | - Jing Zhang
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315100, China
| | - Xinhua Zhou
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315100, China.
| |
Collapse
|
3
|
Ning H, Tao H. Small RNA sequencing of exosomal microRNAs reveals differential expression of microRNAs in preeclampsia. Medicine (Baltimore) 2023; 102:e35597. [PMID: 37861520 PMCID: PMC10589583 DOI: 10.1097/md.0000000000035597] [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: 12/26/2022] [Accepted: 09/20/2023] [Indexed: 10/21/2023] Open
Abstract
Preeclampsia (PE) is one of the most common hypertensive disorders of pregnancy. It is a dangerous condition with a high mortality rate in mothers and fetuses and is associated with a lack of early diagnosis and effective treatment. While the etiology of the disease is complex and obscure, it is now clear that the placenta is central to disease progression. Exosomal microRNAs (miRNAs) are possible mediators that regulate placenta-related physiological and pathological processes. Placental mesenchymal stem cells have considerable potential to help us understand the pathogenesis and treatment of pregnancy-related diseases. Here, we investigate the exosomal miRNA profiles of human placenta-derived mesenchymal stem cells between healthy pregnant women and those with PE. We performed small RNA sequencing to obtain miRNA profiles, and conducted enrichment analysis of the miRNA target genes to identify differentially expressed miRNAs associated with PE. Overall, we detected 1795 miRNAs; among them, 206 were differentially expressed in women with PE, including 35 upregulated and 171 downregulated miRNAs, when compared with healthy pregnant women. Moreover, we identified possible functions and pathways associated with PE, including angiogenesis, cell proliferation, migration and invasion, and the coagulation-fibrinolysis balance. Eventually, we proposed hsa-miR-675-5p, hsa-miR-3614-5p, and hsa-miR-615-5p as potential regulators of the pathogenesis of PE, and constructed a miRNA-target gene network. Our study identifies possible candidate biomarkers for the diagnosis of PE, and introduces a new direction for further understanding the pathogenesis of PE.
Collapse
Affiliation(s)
- Hui Ning
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, China
| | - Hong Tao
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
4
|
Wu YH, Huang YF, Wu PY, Chang TH, Huang SC, Chou CY. The downregulation of miR-509-3p expression by collagen type XI alpha 1-regulated hypermethylation facilitates cancer progression and chemoresistance via the DNA methyltransferase 1/Small ubiquitin-like modifier-3 axis in ovarian cancer cells. J Ovarian Res 2023; 16:124. [PMID: 37386587 DOI: 10.1186/s13048-023-01191-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/18/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND MicroRNAs are a group of small non-coding RNAs that are involved in development and diseases such as cancer. Previously, we demonstrated that miR-335 is crucial for preventing collagen type XI alpha 1 (COL11A1)-mediated epithelial ovarian cancer (EOC) progression and chemoresistance. Here, we examined the role of miR-509-3p in EOC. METHODS The patients with EOC who underwent primary cytoreductive surgery and postoperative platinum-based chemotherapy were recruited. Their clinic-pathologic characteristics were collected, and disease-related survivals were determined. The COL11A1 and miR-509-3p mRNA expression levels of 161 ovarian tumors were determined by real-time reverse transcription-polymerase chain reaction. Additionally, miR-509-3p hypermethylation was evaluated by sequencing in these tumors. The A2780CP70 and OVCAR-8 cells transfected with miR-509-3p mimic, while the A2780 and OVCAR-3 cells transfected with miR-509-3p inhibitor. The A2780CP70 cells transfected with a small interference RNA of COL11A1, and the A2780 cells transfected with a COL11A1 expression plasmid. Site-directed mutagenesis, luciferase, and chromatin immunoprecipitation assays were performed in this study. RESULTS Low miR-509-3p levels were correlated with disease progression, a poor survival, and high COL11A1 expression levels. In vivo studies reinforced these findings and indicated that the occurrence of invasive EOC cell phenotypes and resistance to cisplatin are decreased by miR-509-3p. The miR-509-3p promoter region (p278) is important for miR-509-3p transcription regulation via methylation. The miR-509-3p hypermethylation frequency was significantly higher in EOC tumors with a low miR-509-3p expression than in those with a high miR-509-3p expression. The patients with miR-509-3p hypermethylation had a significantly shorter overall survival (OS) than those without miR-509-3p hypermethylation. Mechanistic studies further indicated that miR-509-3p transcription was downregulated by COL11A1 through a DNA methyltransferase 1 (DNMT1) stability increase. Moreover, miR-509-3p targets small ubiquitin-like modifier (SUMO)-3 to regulate EOC cell growth, invasiveness, and chemosensitivity. CONCLUSION The miR-509-3p/DNMT1/SUMO-3 axis may be an ovarian cancer treatment target.
Collapse
Affiliation(s)
- Yi-Hui Wu
- Department of Medical Research, Chi Mei Medical Center, Liouying, Tainan, 73657, Taiwan
- Department of Nursing, Min-Hwei Junior College of Health Care Management, Tainan, 73658, Taiwan
| | - Yu-Fang Huang
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 70403, Tainan, Taiwan
| | - Pei-Ying Wu
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 70403, Tainan, Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, 110, Taiwan
| | - Soon-Cen Huang
- Department of Obstetrics and Gynecology, Chi Mei Medical Center, Liouying, Tainan, 73657, Taiwan.
| | - Cheng-Yang Chou
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, 70403, Tainan, Taiwan.
| |
Collapse
|
5
|
Scalia P, Williams SJ, Fujita-Yamaguchi Y. Human IGF2 Gene Epigenetic and Transcriptional Regulation: At the Core of Developmental Growth and Tumorigenic Behavior. Biomedicines 2023; 11:1655. [PMID: 37371750 DOI: 10.3390/biomedicines11061655] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Regulation of the human IGF2 gene displays multiple layers of control, which secures a genetically and epigenetically predetermined gene expression pattern throughout embryonal growth and postnatal life. These predominantly nuclear regulatory mechanisms converge on the function of the IGF2-H19 gene cluster on Chromosome 11 and ultimately affect IGF2 gene expression. Deregulation of such control checkpoints leads to the enhancement of IGF2 gene transcription and/or transcript stabilization, ultimately leading to IGF-II peptide overproduction. This type of anomaly is responsible for the effects observed in terms of both abnormal fetal growth and increased cell proliferation, typically observed in pediatric overgrowth syndromes and cancer. We performed a review of relevant experimental work on the mechanisms affecting the human IGF2 gene at the epigenetic, transcriptional and transcript regulatory levels. The result of our work, indeed, provides a wider and diversified scenario for IGF2 gene activation than previously envisioned by shedding new light on its extended regulation. Overall, we focused on the functional integration between the epigenetic and genetic machinery driving its overexpression in overgrowth syndromes and malignancy, independently of the underlying presence of loss of imprinting (LOI). The molecular landscape provided at last strengthens the role of IGF2 in cancer initiation, progression and malignant phenotype maintenance. Finally, this review suggests potential actionable targets for IGF2 gene- and regulatory protein target-degradation therapies.
Collapse
Key Words
- (IGF2/H19) IG-DMR, intergenic differentially methylated region
- BWS, Beckwith–Wiedemann syndrome
- CCD, centrally conserved domain
- CNV, copy number variation
- CTCF, CCCTC binding factor
- DMD, differentially methylated domain
- DMR, differentially methylated region
- GOM, gain of methylation
- ICR1, imprinting control region 1
- IGF-II, insulin-like growth factor-2 peptide
- IGF2, insulin-like growth factor 2 gene
- LOI, loss of imprinting
- LOM, loss of methylation
- MOI, maintenance of imprinting
- SRS, Silver Russel Syndrome
- TF: transcription factor
- UPD, uniparental disomy
- WT1, Wilms Tumor protein 1
- mRNA transcript
- p0–p4: IGF2 promoters 0–4
Collapse
Affiliation(s)
- Pierluigi Scalia
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA 19102, USA, and 93100 Caltanissetta, Italy
- Sbarro Cancer Institute for Cancer Research and Molecular Medicine, CST, Biology Department, Temple University, Philadelphia, PA 19122, USA
| | - Stephen J Williams
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA 19102, USA, and 93100 Caltanissetta, Italy
- Sbarro Cancer Institute for Cancer Research and Molecular Medicine, CST, Biology Department, Temple University, Philadelphia, PA 19122, USA
| | - Yoko Fujita-Yamaguchi
- Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
6
|
Li Q, Zhang L, Yang Q, Li M, Pan X, Xu J, Zhong C, Yao F, Zhang R, Zhou S, Dai X, Shi X, Dai Y, Xu J, Cheng X, Xiao W, She Z, Wang K, Qian X, Pu L, Zhang P, Wang X. Thymidine kinase 1 drives hepatocellular carcinoma in enzyme-dependent and -independent manners. Cell Metab 2023; 35:912-927.e7. [PMID: 37071992 DOI: 10.1016/j.cmet.2023.03.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/17/2022] [Accepted: 03/24/2023] [Indexed: 04/20/2023]
Abstract
Metabolic reprogramming plays a crucial role in the development of hepatocellular carcinoma (HCC). However, the key drivers of metabolic reprogramming underlying HCC progression remain unclear. Using a large-scale transcriptomic database and survival correlation screening, we identify thymidine kinase 1 (TK1) as a key driver. The progression of HCC is robustly mitigated by TK1 knockdown and significantly aggravated by its overexpression. Furthermore, TK1 promotes the oncogenic phenotypes of HCC not only through its enzymatic activity and production of deoxythymidine monophosphate (dTMP) but also by promoting glycolysis via binding with protein arginine methyltransferase 1 (PRMT1). Mechanistically, TK1 directly binds PRMT1 and stabilizes it by interrupting its interactions with tripartite-motif-containing 48 (TRIM48), which inhibits its ubiquitination-mediated degradation. Subsequently, we validate the therapeutic capacity of hepatic TK1 knockdown in a chemically induced HCC mouse model. Therefore, targeting both the enzyme-dependent and -independent activity of TK1 may be therapeutically promising for HCC treatment.
Collapse
Affiliation(s)
- Qing Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Liren Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Qin Yang
- Department of Cardiovascular Surgery, Huanggang Central Hospital, Huanggang Institute of Translational Medicine, Huanggang, China
| | - Mei Li
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Xiongxiong Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiali Xu
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Zhong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Feifan Yao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Ruizhi Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Suiqing Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Xinzheng Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Xiaoli Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Yongjiu Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Jing Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Cheng
- School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenchang Xiao
- Department of Cardiovascular Surgery, Huanggang Central Hospital, Huanggang Institute of Translational Medicine, Huanggang, China
| | - Zhigang She
- School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ke Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| | - Xiaofeng Qian
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| | - Peng Zhang
- School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| |
Collapse
|
7
|
Saviana M, Le P, Micalo L, Del Valle-Morales D, Romano G, Acunzo M, Li H, Nana-Sinkam P. Crosstalk between miRNAs and DNA Methylation in Cancer. Genes (Basel) 2023; 14:1075. [PMID: 37239435 PMCID: PMC10217889 DOI: 10.3390/genes14051075] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
miRNAs are some of the most well-characterized regulators of gene expression. Integral to several physiological processes, their aberrant expression often drives the pathogenesis of both benign and malignant diseases. Similarly, DNA methylation represents an epigenetic modification influencing transcription and playing a critical role in silencing numerous genes. The silencing of tumor suppressor genes through DNA methylation has been reported in many types of cancer and is associated with tumor development and progression. A growing body of literature has described the crosstalk between DNA methylation and miRNAs as an additional layer in the regulation of gene expression. Methylation in miRNA promoter regions inhibits its transcription, while miRNAs can target transcripts and subsequently regulate the proteins responsible for DNA methylation. Such relationships between miRNA and DNA methylation serve an important regulatory role in several tumor types and highlight a novel avenue for potential therapeutic targets. In this review, we discuss the crosstalk between DNA methylation and miRNA expression in the pathogenesis of cancer and describe how miRNAs influence DNA methylation and, conversely, how methylation impacts the expression of miRNAs. Finally, we address how these epigenetic modifications may be leveraged as biomarkers in cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Patrick Nana-Sinkam
- Department of Internal Medicine, Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, 1250 E. Marshall Street, Richmond, VA 23298, USA
| |
Collapse
|
8
|
O’Neill F, Allen-Coyle TJ, Roche S, Meiller J, Conlon NT, Swan N, Straubinger RM, Geoghegan J, Straubinger NL, Conlon K, McDermott R, O’Sullivan F, Henry M, Meleady P, McVey G, O’Connor R, Moriarty M, Clynes M. Alteration in Levels of Specific miRNAs and Their Potential Protein Targets between Human Pancreatic Cancer Samples, Adjacent Normal Tissue, and Xenografts Derived from These Tumors. Life (Basel) 2023; 13:608. [PMID: 36983764 PMCID: PMC10057657 DOI: 10.3390/life13030608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Herein, we describe the global comparison of miRNAs in human pancreatic cancer tumors, adjacent normal tissue, and matched patient-derived xenograft models using microarray screening. RNA was extracted from seven tumor, five adjacent normal, and eight FI PDX tumor samples and analyzed by Affymetrix GeneChip miRNA 4.0 array. A transcriptome analysis console (TAC) was used to generate comparative lists of up- and downregulated miRNAs for the comparisons, tumor vs. normal and F1 PDX vs. tumor. Particular attention was paid to miRNAs that were changed in the same direction in both comparisons. We identified the involvement in pancreatic tumor tissue of several miRNAs, including miR4534, miR3154, and miR4742, not previously highlighted as being involved in this type of cancer. Investigation in the parallel mRNA and protein lists from the same samples allowed the elimination of proteins where altered expression correlated with corresponding mRNA levels and was thus less likely to be miRNA regulated. Using the remaining differential expression protein lists for proteins predicted to be targeted for differentially expressed miRNA on our list, we were able to tentatively ascribe specific protein changes to individual miRNA. Particularly interesting target proteins for miRs 615-3p, 2467-3p, 4742-5p, 509-5p, and 605-3p were identified. Prominent among the protein targets are enzymes involved in aldehyde metabolism and membrane transport and trafficking. These results may help to uncover vulnerabilities that could enable novel approaches to treating pancreatic cancer.
Collapse
Affiliation(s)
- Fiona O’Neill
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
| | - Taylor-Jade Allen-Coyle
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
- SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, V94 T9PX Limerick, Ireland
| | - Sandra Roche
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
| | - Justine Meiller
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
| | - Neil T. Conlon
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
| | - Niall Swan
- St. Vincent’s University Hospital, D04 T6F4 Dublin, Ireland
| | - Robert M. Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, NY 14214, USA
| | | | - Ninfa L. Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, NY 14214, USA
| | - Kevin Conlon
- St. Vincent’s University Hospital, D04 T6F4 Dublin, Ireland
| | - Ray McDermott
- St. Vincent’s University Hospital, D04 T6F4 Dublin, Ireland
| | - Finbarr O’Sullivan
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
- SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, V94 T9PX Limerick, Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
- SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, V94 T9PX Limerick, Ireland
- School of Biotechnology, Dublin City University, D09 K2OV Dublin, Ireland
| | - Gerard McVey
- St. Luke’s Hospital, Rathgar, D06 HH36 Dublin, Ireland
| | - Robert O’Connor
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
| | - Michael Moriarty
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
- St. Luke’s Hospital, Rathgar, D06 HH36 Dublin, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland
- SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, V94 T9PX Limerick, Ireland
| |
Collapse
|
9
|
Wu YH, Huang YF, Wu PY, Chang TH, Huang SC, Chou CY. The Downregulation of miR-509-3p Expression by Collagen Type XI Alpha 1-Regulated Hypermethylation Facilitates Cancer Progression and Chemoresistance via the DNA Methyltransferase 1/Small Ubiquitin-like Modifier-3 Axis in Ovarian Cancer Cells. RESEARCH SQUARE 2023:rs.3.rs-2592453. [PMID: 36865240 PMCID: PMC9980191 DOI: 10.21203/rs.3.rs-2592453/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Background MicroRNAs are a group of small non-coding RNAs that are involved in development and diseases such as cancer. Previously, we demonstrated that miR-335 is crucial for preventing collagen type XI alpha 1 (COL11A1)-mediated epithelial ovarian cancer (EOC) progression and chemoresistance. Here, we examined the role of miR-509-3p in EOC. Methods The patients with EOC who underwent primary cytoreductive surgery and postoperative platinum-based chemotherapy were recruited. Their clinic-pathologic characteristics were collected, and disease-related survivals were determined. The COL11A1 and miR-509-3p mRNA expression levels of 161 ovarian tumors were determined by real-time reverse transcription-polymerase chain reaction. Additionally, miR-509-3p hypermethylation was evaluated by sequencing in these tumors. The A2780CP70 and OVCAR-8 cells transfected with miR-509-3p mimic, while the A2780 and OVCAR-3 cells transfected with miR-509-3p inhibitor. The A2780CP70 cells transfected with a small interference RNA of COL11A1, and the A2780 cells transfected with a COL11A1 expression plasmid. Site-directed mutagenesis, luciferase, and chromatin immunoprecipitation assays were performed in this study. Results Low miR-509-3p levels were correlated with disease progression, a poor survival, and high COL11A1 expression levels. In vivo studies reinforced these findings and indicated that the occurrence of invasive EOC cell phenotypes and resistance to cisplatin are decreased by miR-509-3p. The miR-509-3p promoter region (p278) is important for miR-509-3p transcription regulation via methylation. The miR-509-3p hypermethylation frequency was significantly higher in EOC tumors with a low miR-509-3p expression than in those with a high miR-509-3p expression. The patients with miR-509-3p hypermethylation had a significantly shorter overall survival (OS) than those without miR-509-3p hypermethylation. Mechanistic studies further indicated that miR-509-3p transcription was downregulated by COL11A1 through a DNA methyltransferase 1 (DNMT1) phosphorylation and stability increase. Moreover, miR-509-3p targets small ubiquitin-like modifier (SUMO)-3 to regulate EOC cell growth, invasiveness, and chemosensitivity. Conclusion The miR-509-3p/DNMT1/SUMO-3 axis may be an ovarian cancer treatment target.
Collapse
Affiliation(s)
| | - Yu-Fang Huang
- National Cheng Kung University Hospital, National Cheng Kung University
| | - Pei-Ying Wu
- National Cheng Kung University Hospital, National Cheng Kung University
| | | | | | - Cheng-Yang Chou
- National Cheng Kung University Hospital, National Cheng Kung University
| |
Collapse
|
10
|
Li D, Yan L, Zhang J, Gu F. Circular RNA hsa_circ_0004396 acts as a sponge of miR-615-5p to promote non-small cell lung cancer progression and radioresistance through the upregulation of P21-Activated Kinase 1. J Clin Lab Anal 2022; 36:e24463. [PMID: 35500159 PMCID: PMC9169218 DOI: 10.1002/jcla.24463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUNDS CircRNA hsa_circ_0004396 has been confirmed to be upregulated in human non-small cell lung cancer (NSCLC). The aim of his study was to evaluate its mechanism in the radioresistance and progression of NSCLC. METHODS Hsa_circ_0004396, miR-615-5p, and P21-Activated Kinase 1 (PAK1) were measured by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR). The binding between miR-615-5p and hsa_circ_0004396 or PAK1 was predicted by circinteractome or Targetscan, as verified by dual-luciferase reporter assay and RIP assay. Proliferation, clonogenicity capacity, cell cycle progression, apoptosis, migration, and invasion were assessed by CCK-8, colony formation, flow cytometry, and Transwell assay. Bcl-2, Bcl-2 associated protein X (Bax), MMP-2, and PAK1 protein levels were detected using western blot assay. In addition, in vivo function of hsa_circ_0004396 was evaluated by tumor xenograft assay. RESULTS Hsa_circ_0004396 and PAK1 levels were upregulated, while miR-615-5p was declined in NSCLC. Hsa_circ_0004396 silencing inhibited NSCLC cell malignant behavior and induced radiosensitivity. Hsa_circ_0004396 functions as a molecular sponge of miR-615-5p to regulate PAK1 expression. Moreover, hsa_circ_0004396 knockdown inhibited NSCLC tumor growth in vivo. CONCLUSION Our findings demonstrated that hsa_circ_0004396 promoted NSCLC development and radioresistance through the miR-615-5p/PAK1 axis, which might provide a new therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Dong Li
- Department of Thoracic Surgery, Gansu Provincial Tumor Hospital, Lanzhou, Gansu, China
| | - Lin Yan
- Department of Anesthesiology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Junhan Zhang
- Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Feng Gu
- Department of Aspiration Oncology, Gansu Provincial Tumor Hospital, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Gao F, Wang X, Fan T, Luo Z, Ma M, Hu G, Li Y, Liang Y, Lin X, Xu B. LncRNA LINC00461 exacerbates myocardial ischemia-reperfusion injury via microRNA-185-3p/Myd88. Mol Med 2022; 28:33. [PMID: 35272621 PMCID: PMC8908691 DOI: 10.1186/s10020-022-00452-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Long non-coding RNAs (lncRNAs) play critically in the pathogenesis of myocardial ischemia-reperfusion (I/R) injury. Thus, it was proposed to investigate the mechanism of LINC00461 in the disease through mediating microRNA-185-3p (miR-185-3p)/myeloid differentiation primary response gene 88 (Myd88) axis. METHODS miR-185-3p, LINC00461 and Myd88 expression in mice with I/R injury was measured. Mice with I/R injury were injected with the gene expression-modified vectors, after which cardiac function, hemodynamics, myocardial enzyme, oxidative stress, and cardiomyocyte apoptosis were analyzed. RESULTS I/R mice showed LINC00461 and Myd88 up-regulation and miR-185-3p down-regulation. Down-regulating LINC00461 or up-regulating miR-185-3p recovered cardiac function, reduced myocardial enzyme levels, and attenuated oxidative stress and cardiomyocyte apoptosis in mice with I/R. miR-185-3p overexpression rescued the promoting effect of LINC00461 upregulation on myocardial injury in I/R mice. CONCLUSION LINC00461 knockdown attenuates myocardial I/R injury via elevating miR-185-3p expression to suppress Myd88 expression.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiology, Economic Development District, Second Affiliated Hospital of Anhui Medical University, No.678 Furong Road, Hefei, 230601, Anhui, China
| | - Xiaochen Wang
- Department of Cardiology, Economic Development District, Second Affiliated Hospital of Anhui Medical University, No.678 Furong Road, Hefei, 230601, Anhui, China
| | - Tingting Fan
- Department of Cardiology, Economic Development District, Second Affiliated Hospital of Anhui Medical University, No.678 Furong Road, Hefei, 230601, Anhui, China
| | - Zhidan Luo
- Department of Geriatrics, Chongqing People's Hospital, Chongqing, 400013, China
| | - Mengqing Ma
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Guangquan Hu
- Department of Cardiology, Economic Development District, Second Affiliated Hospital of Anhui Medical University, No.678 Furong Road, Hefei, 230601, Anhui, China
| | - Yue Li
- Department of Cardiology, Economic Development District, Second Affiliated Hospital of Anhui Medical University, No.678 Furong Road, Hefei, 230601, Anhui, China
| | - Yi Liang
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Xianhe Lin
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
| | - Banglong Xu
- Department of Cardiology, Economic Development District, Second Affiliated Hospital of Anhui Medical University, No.678 Furong Road, Hefei, 230601, Anhui, China
| |
Collapse
|
12
|
Ren F, Li B, Wang C, Wang Y, Cui B. Iodine-125 seed represses the growth and facilitates the apoptosis of colorectal cancer cells by suppressing the methylation of miR-615 promoter. BMC Cancer 2022; 22:49. [PMID: 34998382 PMCID: PMC8742920 DOI: 10.1186/s12885-021-09141-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/21/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) represents a common malignancy in gastrointestinal tract. Iodine-125 (125I) seed implantation is an emerging treatment technology for unresectable tumors. This study investigated the mechanism of 125I seed in the function of CRC cells. METHODS The CRC cells were irradiated with different doses of 125I seed (0.4, 0.6 and 0.8 mCi). miR-615 expression in CRC tissues and adjacent tissues was detected by RT-qPCR. miR-615 expression was intervened with miR-615 mimic or miR-615 inhibitor, and then the CRC cells were treated with 5-AZA (methylation inhibitor). The CRC cell growth, invasion and apoptosis were measured. The methylation level of miR-615 promoter region was detected. The xenograft tumor model irradiated by 125I seed was established in nude mice. The methylation of miR-615, Ki67 expression and CRC cell apoptosis were detected. RESULTS 125I seed irradiation repressed the growth and facilitated apoptosis of CRC cells in a dose-dependent manner. Compared with adjacent tissues, miR-615 expression in CRC tissues was downregulated and miR-615 was poorly expressed in CRC cells. Overexpression of miR-615 suppressed the growth of CRC cells. 125I seed-irradiated CRC cells showed increased miR-615 expression, reduced growth rate and enhanced apoptosis. The methylation level of miR-615 promoter region in CRC cells was decreased after 125I seed treatment. In vivo experiments confirmed that 125I seed-irradiated xenograft tumors showed reduced methylation of the miR-615 promoter and increased miR-615 expression, as well as decreased Ki67 expression and enhanced apoptosis. The target genes of miR-615 and its regulatory downstream pathway were further predicted by bioinformatics analysis. CONCLUSIONS 125I seed repressed the growth and facilitated the apoptosis of CRC cells by suppressing the methylation of the miR-615 promoter and thus activating miR-615 expression. The possible mechanism was that miR-615-5p targeted MAPK13, thus affecting the MAPK pathway and the progression of CRC.
Collapse
Affiliation(s)
- Fenghai Ren
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Baojun Li
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Chao Wang
- Department of Prenatal Diagnosis, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yanbo Wang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Binbin Cui
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
13
|
Chhichholiya Y, Suryan AK, Suman P, Munshi A, Singh S. SNPs in miRNAs and Target Sequences: Role in Cancer and Diabetes. Front Genet 2021; 12:793523. [PMID: 34925466 PMCID: PMC8673831 DOI: 10.3389/fgene.2021.793523] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/28/2021] [Indexed: 12/27/2022] Open
Abstract
miRNAs are fascinating molecular players for gene regulation as individual miRNA can control multiple targets and a single target can be regulated by multiple miRNAs. Loss of miRNA regulated gene expression is often reported to be implicated in various human diseases like diabetes and cancer. Recently, geneticists across the world started reporting single nucleotide polymorphism (SNPs) in seed sequences of miRNAs. Similarly, SNPs are also reported in various target sequences of these miRNAs. Both the scenarios lead to dysregulated gene expression which may result in the progression of diseases. In the present paper, we explore SNPs in various miRNAs and their target sequences reported in various human cancers as well as diabetes. Similarly, we also present evidence of these mutations in various other human diseases.
Collapse
Affiliation(s)
- Yogita Chhichholiya
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Aman Kumar Suryan
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Prabhat Suman
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| |
Collapse
|
14
|
Heinrich MA, Mostafa AMRH, Morton JP, Hawinkels LJAC, Prakash J. Translating complexity and heterogeneity of pancreatic tumor: 3D in vitro to in vivo models. Adv Drug Deliv Rev 2021; 174:265-293. [PMID: 33895214 DOI: 10.1016/j.addr.2021.04.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive type of cancer with an overall survival rate of less than 7-8%, emphasizing the need for novel effective therapeutics against PDAC. However only a fraction of therapeutics which seemed promising in the laboratory environment will eventually reach the clinic. One of the main reasons behind this low success rate is the complex tumor microenvironment (TME) of PDAC, a highly fibrotic and dense stroma surrounding tumor cells, which supports tumor progression as well as increases the resistance against the treatment. In particular, the growing understanding of the PDAC TME points out a different challenge in the development of efficient therapeutics - a lack of biologically relevant in vitro and in vivo models that resemble the complexity and heterogeneity of PDAC observed in patients. The purpose and scope of this review is to provide an overview of the recent developments in different in vitro and in vivo models, which aim to recapitulate the complexity of PDAC in a laboratory environment, as well to describe how 3D in vitro models can be integrated into drug development pipelines that are already including sophisticated in vivo models. Hereby a special focus will be given on the complexity of in vivo models and the challenges in vitro models face to reach the same levels of complexity in a controllable manner. First, a brief introduction of novel developments in two dimensional (2D) models and ex vivo models is provided. Next, recent developments in three dimensional (3D) in vitro models are described ranging from spheroids, organoids, scaffold models, bioprinted models to organ-on-chip models including a discussion on advantages and limitations for each model. Furthermore, we will provide a detailed overview on the current PDAC in vivo models including chemically-induced models, syngeneic and xenogeneic models, highlighting hetero- and orthotopic, patient-derived tissues (PDX) models, and genetically engineered mouse models. Finally, we will provide a discussion on overall limitations of both, in vitro and in vivo models, and discuss necessary steps to overcome these limitations to reach an efficient drug development pipeline, as well as discuss possibilities to include novel in silico models in the process.
Collapse
Affiliation(s)
- Marcel A Heinrich
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Ahmed M R H Mostafa
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow G61 1QH, UK
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Centre, PO-box 9600, 2300 RC Leiden, the Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| |
Collapse
|
15
|
Abdelaleem OO, Shaker OG, AbdelHafez MN, Abdelghaffar NK, Eid HM, Zaidan M, Khalefa AA, Ahmed NA, Hemeda NF, Zaki OM, Awaji AAA, Mohammed SR. The Influence of rs1859168 Polymorphism on Serum Expression of HOTTIP and Its Target miR-615-3p in Egyptian Patients with Breast Cancer. Biomolecules 2021; 11:733. [PMID: 34069089 PMCID: PMC8156858 DOI: 10.3390/biom11050733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/02/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Polymorphisms of long noncoding RNAs are lately documented as hazardous factors for the development of numerous tumors. Furthermore, the evaluation of noncoding RNAs has emerged as a novel detector of breast cancer patients. We aimed to genotype the HOXA transcript at the distal tip (HOTTIP) rs1859168 and assess its relationship with the levels of the serum HOTTIP and its target miR-615-3p in patients with breast cancer (BC). METHODS One hundred and fifty-one patients with BC, 139 patients with fibroadenoma (FA), and 143 healthy participants were incorporated into the current study. The genotyping of rs1859168 and the measurements of the HOTTIP and miR-615-3p levels were assessed using quantitative real-time PCR. RESULTS We revealed a significant association between each of the CC genotypes, C allele, dominant and recessive models, and the increased risk of BC (p = 0.013, p < 0.001, p < 0.001, and p < 0.001, respectively) relative to the healthy controls. Similarly, the CC genotype, C allele, and recessive model were observed to be related to the increased incidence of BC with respect to FA (p < 0.001 for all). A significant upregulation of HOTTIP and a marked decrease of miR-615-3p were verified in patients with BC compared to each of the healthy individuals, patients with FA, and the non-BC group (healthy subjects + FA) (p < 0.001 for all). A significant negative correlation was demonstrated between the expression of HOTTIP and miR-615-3p in the serum of patients with BC. The HOTTIP expression was upregulated, while that of miR-615-3p was downregulated in patients with BC who carried the CC genotype with respect to those who carried the AA or AC genotypes (p < 0.05 for all). CONCLUSIONS The genetic variants of rs1859168 are linked to an increased susceptibility to BC. Moreover, HOTTIP and miR-615-3p may be used as novel indicators and targets for the treatment of patients with BC.
Collapse
Affiliation(s)
- Omayma O. Abdelaleem
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt; (O.O.A.); (S.R.M.)
| | - Olfat G. Shaker
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo 11511, Egypt;
| | - Marwa N. AbdelHafez
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo 11511, Egypt;
| | - Noha K. Abdelghaffar
- Department of Clinical pathology, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt;
| | - Hanaa M. Eid
- Department of Microbiology and Immunology, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt
| | - Mohamed Zaidan
- Department of General Surgery, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt;
| | - Abeer A. Khalefa
- Department of Physiology, Faculty of Medicine, Zagazig University, Zagazig 44523, Egypt; (A.A.K.); (N.A.A.)
| | - Naglaa A. Ahmed
- Department of Physiology, Faculty of Medicine, Zagazig University, Zagazig 44523, Egypt; (A.A.K.); (N.A.A.)
| | - Nada F. Hemeda
- Department of Genetics, Faculty of Agriculture, Fayoum University, Fayoum 63511, Egypt;
| | - Othman M. Zaki
- Department of Clinical Pathology, Faculty of Medicine, Damietta University, Damietta 34511, Egypt;
| | - Aeshah Ali A. Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, Tabuk University, Tabuk 47711, Saudi Arabia;
| | - Shereen R. Mohammed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt; (O.O.A.); (S.R.M.)
| |
Collapse
|
16
|
Liang Z, Zhao B, Hou J, Zheng J, Xin G. CircRNA circ-OGDH (hsa_circ_0003340) Acts as a ceRNA to Regulate Glutamine Metabolism and Esophageal Squamous Cell Carcinoma Progression by the miR-615-5p/PDX1 Axis. Cancer Manag Res 2021; 13:3041-3053. [PMID: 33854374 PMCID: PMC8039021 DOI: 10.2147/cmar.s290088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/01/2021] [Indexed: 01/17/2023] Open
Abstract
Background Circular RNA hsa_circ_0003340 (circ-OGDH) has been uncovered to be involved in esophageal squamous cell carcinoma (ESCC) progression. However, the mechanism by which circ-OGDH regulates ESCC progression is unclear. Methods Expression levels of circ-OGDH, microRNA (miR)-615-5p, and PDX1 (pancreatic and duodenal homeobox 1) mRNA were evaluated with quantitative real-time polymerase chain reaction (qRT-PCR). The proliferation, apoptosis, migration, invasion, and cell cycle progression of ESCC cells were analyzed by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide), colony formation, flow cytometry, and transwell assays. Measurement of glutamine consumption, α-KG (α-ketoglutarate) production, and ATP (Adenosine Triphosphate) content using corresponding kits. Protein levels were analyzed by Western blotting. The targeting relationship between circ-OGDH or PDX1 and miR-615-5p was verified by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. The function of circ-OGDH in ESCC was confirmed by animal experiments. Results Circ-OGDH was upregulated in ESCC. Circ-OGDH inhibition reduced ESCC growth in vivo and accelerated cell apoptosis, cell cycle arrest, repressed cell proliferation, migration, invasion, and reduced cell glutamine metabolism in ESCC cells in vitro. MiR-615-5p was downregulated in ESCC, while PDX1 had an opposite result. Circ-OGDH sponged miR-615-5p to regulate PDX1 expression. MiR-615-5p inhibitor neutralized the repressive effect of circ-OGDH knockdown on malignancy and glutamine metabolism of ESCC cells. PDX1 overexpression counteracted the inhibitory impact of miR-615-5p mimic on malignancy and glutamine metabolism of ESCC cells. Conclusion Circ-OGDH sponged miR-615-5p to elevate PDX1 expression, thus elevating glutamine metabolism and promoting tumor growth in ESCC. The study offered evidence to support circ-OGDH as a promising target for ESCC therapy.
Collapse
Affiliation(s)
- Zongying Liang
- Department of Thoracic Surgery, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, People's Republic of China
| | - Baoshan Zhao
- Department of Thoracic Surgery, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, People's Republic of China
| | - Jishen Hou
- Department of Thoracic Surgery, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, People's Republic of China
| | - Jingxiong Zheng
- Department of Thoracic Surgery, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, People's Republic of China
| | - Guohua Xin
- Department of Thoracic Surgery, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, People's Republic of China
| |
Collapse
|
17
|
Ilkhani K, Delgir S, Safi A, Seif F, Samei A, Bastami M, Alivand MR. Clinical and In Silico Outcomes of the Expression of miR-130a-5p and miR-615-3p in Tumor Compared with Non-Tumor Adjacent Tissues of Patients with BC. Anticancer Agents Med Chem 2021; 21:927-935. [PMID: 32972352 DOI: 10.2174/1871520620666200924105352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/23/2020] [Accepted: 07/31/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Breast Cancer (BC) is the most common malignancy among women with a high mortality rate. The blockade of asparagine-related pathways may be an effective measure to control the progression and reduction of BC metastasis potential. Recently, it has been shown that various miRNAs, as part of small non-coding RNAs, have a great role in cancer development, especially asparagine-related pathways, to modulate the invasiveness. OBJECTIVE This study aimed to evaluate the expression of miR-130a-5p and miR-615-3p in tumoral and nontumoral adjacent tissues of patients with BC. METHODS There is a chance that asparagine metabolism is influenced by miR-130a-5p and miR-615-3p as confirmed by bioinformatics analysis. Hence, real-time PCR was conducted on eighty BC tumoral and non-tumoral adjacent tissues to evaluate the expression level of the two miRNAs. To predict the potential biological process and molecular pathways of miR-130a-5p, an in silico analysis was performed. RESULTS This study indicated that miR-130a was downregulated in tumoral tissues compared to non-tumoral adjacent tissues (P-value= 0.01443 and fold change= -2.5137), while miR-615-3p did not show a significant difference between the two groups. Furthermore, the subgroup studies did not reveal any significant correlation between the expression of these two miRNAs and subfactors. Furthermore, in silico studies unraveled several biological processes related to amino-acid metabolism, as well as pathways related to tumor development such as Phosphatase and Tensin Homolog (PTEN) and JAK-STAT pathways among miR-130a-5p target genes. CONCLUSION Our findings indicate that miRNA-130a-5p is downregulated in BC tissues and may play a tumor suppressor role in patients with BC. Therefore, it may be suggested as a potential diagnostic and therapeutic target for BC.
Collapse
Affiliation(s)
- Khandan Ilkhani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Delgir
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asma Safi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Seif
- Department of Immunology & Allergy, Academic Center for Education, Culture, and Research, Tehran, Iran
| | - Azam Samei
- Department of Laboratory Sciences, School of Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Milad Bastami
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Hai X, Zhao G, Li Z, Wu J, Xu X, Yang Y. Effects of microRNA-103 on the Proliferation and Apoptosis of Pancreatic Cancer Cells via Targeting Phosphatase and Tensin Homolog Deleted on Chromosome Ten (PTEN) and Activating Phosphoinositide 3-Kinase/A Serine/Threonine Kinase (PI3K/Akt) Signaling Pathway. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objective: To investigate whether micro ribonucleic acid (miR)-103 affects pancreatic cancer (PaCa) cells via PTEN-activated PI3K/Akt signaling pathway. Methods: Differences in miR-103 expression in 35 pairs of PaCa tissues and cell lines (SW1990 and PATU8988S) were detected
by RT-qPCR. miR-103 inhibitor was transfected into PaCa PATU8988S cell followed by analysis of proliferation and apoptosis of PaCa cells by MTT assay and flow cytometry, respectively. Results: MiR-103 exhibited a significantly high expression in PaCa tissues and cell lines (p
< 0.05). Besides, the exogenous inhibition of miR-103 expression in PATU8988S cells significantly inhibited cell proliferation and migration but increased apoptosis activity (p < 0.05). According to the prediction of TargetScan biological database, miR-103 could bind PTEN 3′
untranslated region (3′UTR) and miR-103 was confirmed to suppress PTEN expression in a targeted way (p<0.05). Furthermore, down-regulation of PTEN activated PI3K/Akt signaling to affect the proliferation and apoptosis of PaCa cells (p < 0.05 or p <0.01). Conclusion:
MiR-103 displays a significantly increased expression in PaCa cells and targets PTEN to activate PI3K/Akt signaling pathway, thus promoting malignant phenotype formation.
Collapse
Affiliation(s)
- Xiaoyu Hai
- Department of General Surgery, Ningxia Fifth People’s Hospital, Shizuishan, 753000, Ningxia, China
| | - Guozhong Zhao
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Zhaolong Li
- Department of General Surgery, Ningxia Fifth People’s Hospital, Shizuishan, 753000, Ningxia, China
| | - Junli Wu
- Department of Pancreatic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 753000, Jiangsu, China
| | - Xiangzhao Xu
- Department of Anesthesiology, Ningxia Fifth People’s Hospital, Shizuishan, 753000, Ningxia, China
| | - Yaowen Yang
- Department of Radiology, Fifth People’s Hospital of Ningxia, Shizuishan, 753000, Ningxia, China
| |
Collapse
|
19
|
Hussen BM, Hidayat HJ, Salihi A, Sabir DK, Taheri M, Ghafouri-Fard S. MicroRNA: A signature for cancer progression. Biomed Pharmacother 2021; 138:111528. [PMID: 33770669 DOI: 10.1016/j.biopha.2021.111528] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are a group of small non-coding RNAs that post-transcriptionally control expression of genes by targeting mRNAs. miRNA alterations partake in the establishment and progression of different types of human cancer. Consequently, expression profiling of miRNA in human cancers has correlations with cancer detection, staging, progression, and response to therapies. Particularly, amplification, deletion, abnormal pattern of epigenetic factors and the transcriptional factors that mediate regulation of primary miRNA frequently change the landscape of miRNA expression in cancer. Indeed, changes in the quantity and quality of miRNAs are associated with the initiation of cancer, its progression and metastasis. Additionally, miRNA profiling has been used to categorize genes that can affect oncogenic pathways in cancer. Here, we discuss several circulating miRNA signatures, their expression profiles in different types of cancer and their impacts on cellular processes.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Iraq
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq; Department of Medical Analysis, Faculty of Science, Tishk International University, Erbil, Iraq
| | - Dana K Sabir
- Department of Medical Laboratory Sciences, Charmo University, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Liu K, Ma R. MicroRNA-615-5p regulates the proliferation and apoptosis of breast cancer cells by targeting HSF1. Exp Ther Med 2021; 21:192. [PMID: 33488801 DOI: 10.3892/etm.2021.9624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/31/2019] [Indexed: 12/22/2022] Open
Abstract
Breast cancer, which commonly occurs in the epithelium of the mammary gland, is a malignant tumor. MicroRNAs are involved in various cancer-associated processes, and microRNA-615-5p has been identified to be decreased in the pathological tissues from patients with breast cancer. In the present study, the possible mechanism of microRNA-615-5p in the progression of breast cancer was investigated in order to identify potential novel targets for clinical treatment. Heat shock factor 1 (HSF1) was identified as a predictive target gene of microRNA-615-5p using TargetScan analysis. The expression levels of microRNA-615-5p and its target gene, HSF1, were measured in breast cancer tissues and normal adjacent tissues. Additionally, the effects of microRNA-615-5p on MCF-7 breast cancer cell growth and apoptosis were examined. Furthermore, the interaction between HSF1 and microRNA-615-5p was investigated by a dual luciferase gene reporter assay. The expression levels of HSF1 were measured following transfection with microRNA-615-5p or pcDNA3.1-HSF1. Finally, the expression levels of proliferation- and apoptosis-associated factors such as B-cell lymphoma 2 (Bcl-2), cyclin D1, proliferating cell nuclear antigen (PCNA) and bcl-2-like protein 4 (Bax) were determined. The results demonstrated that lower microRNA-615-5p expression and higher HSF1 mRNA expression were present in tumor tissues compared with adjacent tissues (P<0.01). HSF1 was verified as a direct target of microRNA-615-5p using the dual luciferase gene reporter assay. In comparison with untransfected control and mimic-transfected negative control (NC) cells, MCF-7 cells transfected with microRNA-615-5p mimics exhibited reduced cell proliferation and increased apoptosis (P<0.01). However, the overexpression of HSF1 using a vector reversed the suppression of HSF1 induced by microRNA-615-5p mimics (P<0.01). The mRNA and protein expression levels of Bax were significantly increased, whereas those of Bcl-2, cyclin D1 and PCNA were decreased in the cells transfected with microRNA-615-5p mimics compared with the control and NC cells (P<0.01). Collectively, the present study indicated that microRNA-615-5p may mediate the progression of breast cancer by targeting HSF1.
Collapse
Affiliation(s)
- Kaisheng Liu
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Rong Ma
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
21
|
circSETD3 regulates MAPRE1 through miR-615-5p and miR-1538 sponges to promote migration and invasion in nasopharyngeal carcinoma. Oncogene 2020; 40:307-321. [PMID: 33122825 DOI: 10.1038/s41388-020-01531-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/25/2020] [Accepted: 10/15/2020] [Indexed: 12/22/2022]
Abstract
Circular RNAs (circRNAs) play an essential role in tumorigenesis and development. However, they have rarely been investigated in nasopharyngeal carcinoma (NPC). This study aimed to investigate the role of circRNA in the invasion and metastasis of NPC. We screened and verified the high expression of circSETD3 in NPC cell lines using RNA sequencing (RNA-Seq) and verified the results of NPC biopsy samples using real-time quantitative polymerase chain reaction (qRT-PCR) and in situ hybridization (ISH). In vivo and in vitro experiments indicated that circSETD3 could promote NPC cell invasion and migration. We compared the proteomic data of NPC cells before and after the overexpression or knockdown of circSETD3 in combination with bioinformatics prediction and experimental verification. It was found that circSETD3 competitively adsorbs to miR-615-5p and miR-1538 and negates their inhibitory effect on MAPRE1 mRNA, thereby upregulating the expression of MAPRE1. The upregulated MAPRE1 then inhibits the acetylation of α-tubulin, promotes the dynamic assembly of microtubules, and enhances the invasion and migration capabilities of NPC cells. The results of this study suggest that circSETD3 is a novel molecular marker and a potential target for NPC diagnosis and treatment.
Collapse
|
22
|
Gu Y, Zhang B, Gu G, Yang X, Qian Z. Metformin Increases the Chemosensitivity of Pancreatic Cancer Cells to Gemcitabine by Reversing EMT Through Regulation DNA Methylation of miR-663. Onco Targets Ther 2020; 13:10417-10429. [PMID: 33116621 PMCID: PMC7569251 DOI: 10.2147/ott.s261570] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/06/2020] [Indexed: 12/12/2022] Open
Abstract
Background Pancreatic cancer is a devastating malignancy with poor prognosis. Metformin, a classic anti-diabetes drug, seems to improve survival of pancreatic cancer patients in some studies. Methods Cell counting kit-8 assay was used to detect the BxPC-3 and MIAPaCa-2 cell viability after treatment with gemcitabine only or with different concentrations of metformin. The methylation state and expression level of miR-663 were detected by methylation analysis and RT-PCR. Dual-luciferase reporter gene analysis, Western blot and RT-PCR were used to confirm the target of miR-663. Moreover, xenograft experiment was also performed to validate the role of metformin in chemosensitivity in vivo. Results We found that metformin increased the chemosensitivity of pancreatic cancer cells to gemcitabine, and epithelial-mesenchymal transition (EMT) progress caused by gemcitabine was suppressed by metformin. We further explored the possible molecular mechanisms and it was demonstrated that CpG islands of miR-663 were hypomethylated and relative expression level of miR-663 was up-regulated after treatment of metformin. miR-663, an important cancer suppressor miRNA, was confirmed to increase the chemosensitivity of pancreatic cancer cells by reversing EMT directly targeted TGF-β1. Moreover, we identified that metformin increased the chemosensitivity through up-regulating expression of miR-663. Conclusion We demonstrated that metformin increased the chemosensitivity of pancreatic cancer cells to gemcitabine by reversing EMT through regulation DNA methylation of miR-663.
Collapse
Affiliation(s)
- Yuqing Gu
- Pancreas Center, The Second Affiliated Hospital to Nanjing Medical University, Nanjing 210003, People's Republic of China
| | - Bin Zhang
- Pancreas Center, The Second Affiliated Hospital to Nanjing Medical University, Nanjing 210003, People's Republic of China
| | - Guangliang Gu
- Pancreas Center, The Second Affiliated Hospital to Nanjing Medical University, Nanjing 210003, People's Republic of China
| | - Xiaojun Yang
- Pancreas Center, The Second Affiliated Hospital to Nanjing Medical University, Nanjing 210003, People's Republic of China
| | - Zhuyin Qian
- Pancreas Center, The Second Affiliated Hospital to Nanjing Medical University, Nanjing 210003, People's Republic of China
| |
Collapse
|
23
|
Sun L, Wang P, Zhang Z, Zhang K, Xu Z, Li S, Mao J. MicroRNA-615 functions as a tumor suppressor in osteosarcoma through the suppression of HK2. Oncol Lett 2020; 20:226. [PMID: 32968448 PMCID: PMC7500052 DOI: 10.3892/ol.2020.12089] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
At present, the regulatory mechanisms of various microRNAs (miRNAs/miRs) have been elucidated in human cancers including osteosarcoma (OS). This study mainly focused on the role of miR-615 in OS, which has not yet been reported. Ninety-two OS tissues and normal samples were used in this study. Human osteoblast hFOB1.19 cells and OS cell line HOS were utilized to detect the expression of miR-615. The expression of miR-615 and gene expression were assessed by RT-qPCR and western blot analysis. Transwell, MTT and luciferase reporter assays were used to investigate the regulatory mechanism of miR-615 in OS. The results revealed that miR-615 expression was reduced in OS tissues and cells, and was associated with poor clinical outcomes and prognosis in OS patients. In addition, overexpression of miR-615 restrained cell viability and metastasis in OS. Furthermore, hexokinase 2 (HK2) was confirmed as a direct target of miR-615. Upregulation of HK2 was detected in OS tissues. The upregulation of HK2 weakened the tumor-suppressive effect of miR-615 in OS. Moreover, miR-615 blocked epithelial-mesenchymal transition (EMT) and inactivated the PI3K/AKT pathway in OS. To conclude, miR-615 acts as a tumor suppressor in OS, thus miR-615 can be used as a target for OS treatment.
Collapse
Affiliation(s)
- Limin Sun
- Department of Orthopedics, Shandong Provincial Third Hospital, Jinan, Shandong 250031, P.R. China
| | - Peng Wang
- Department of Orthopedics, Shandong Provincial Third Hospital, Jinan, Shandong 250031, P.R. China
| | - Zhiqiang Zhang
- Department of Orthopedics, Shandong Provincial Third Hospital, Jinan, Shandong 250031, P.R. China
| | - Kai Zhang
- Department of Orthopedics, Shandong Provincial Third Hospital, Jinan, Shandong 250031, P.R. China
| | - Zheng Xu
- Department of Orthopedics, Shandong Provincial Third Hospital, Jinan, Shandong 250031, P.R. China
| | - Siyuan Li
- Department of Orthopedics, Shandong Provincial Third Hospital, Jinan, Shandong 250031, P.R. China
| | - Junsheng Mao
- Department of Orthopedic Surgery, Taishan Hospital of Shandong Province, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
24
|
He M, Henderson M, Muth S, Murphy A, Zheng L. Preclinical mouse models for immunotherapeutic and non-immunotherapeutic drug development for pancreatic ductal adenocarcinoma. ACTA ACUST UNITED AC 2020; 3. [PMID: 32832900 PMCID: PMC7440242 DOI: 10.21037/apc.2020.03.03] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is in urgent need of better diagnostic and therapeutic methods due to its late diagnosis, limited treatment options and poor prognosis. Finding the right animal models to recapitulate the tumor molecular pathogenesis and tumor microenvironment (TME) complexity is critical for preclinical immunotherapeutic and non-immunotherapeutic treatment developments. In this review, we summarize and evaluate popular preclinical animal models including patient-derived xenograft models, humanized mouse models, genetically engineered mouse models, and syngeneic mouse models. Through comparisons between these models in different research settings, we hope to provide guidance in finding the most relevant preclinical models to suit various research purposes.
Collapse
Affiliation(s)
- Mengni He
- Department of Cell Biology, Baltimore, MD, USA
| | - MacKenzie Henderson
- Department of Oncology, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen Muth
- Department of Oncology, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adrian Murphy
- Department of Oncology, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,The Precision Medicine Center of Excellence (PMCoE) Program for Pancreatic Cancer, Baltimore, MD, USA
| | - Lei Zheng
- Department of Oncology, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,The Precision Medicine Center of Excellence (PMCoE) Program for Pancreatic Cancer, Baltimore, MD, USA
| |
Collapse
|
25
|
miR-615 Fine-Tunes Growth and Development and Has a Role in Cancer and in Neural Repair. Cells 2020; 9:cells9071566. [PMID: 32605009 PMCID: PMC7408929 DOI: 10.3390/cells9071566] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that function as epigenetic modulators regulating almost any gene expression. Similarly, other noncoding RNAs, as well as epigenetic modifications, can regulate miRNAs. This reciprocal interaction forms a miRNA-epigenetic feedback loop, the deregulation of which affects physiological processes and contributes to a great diversity of diseases. In the present review, we focus on miR-615, a miRNA highly conserved across eutherian mammals. It is involved not only during embryogenesis in the regulation of growth and development, for instance during osteogenesis and angiogenesis, but also in the regulation of cell growth and the proliferation and migration of cells, acting as a tumor suppressor or tumor promoter. It therefore serves as a biomarker for several types of cancer, and recently has also been found to be involved in reparative processes and neural repair. In addition, we present the pleiad of functions in which miR-615 is involved, as well as their multiple target genes and the multiple regulatory molecules involved in its own expression. We do this by introducing in a comprehensible way the reported knowledge of their actions and interactions and proposing an integral view of its regulatory mechanisms.
Collapse
|
26
|
Yang N, Dong B, Song Y, Li Y, Kou L, Yang J, Qin Q. Downregulation of miR-637 promotes vascular smooth muscle cell proliferation and migration via regulation of insulin-like growth factor-2. Cell Mol Biol Lett 2020; 25:30. [PMID: 32399056 PMCID: PMC7203897 DOI: 10.1186/s11658-020-00222-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/13/2020] [Indexed: 02/08/2023] Open
Abstract
Background Dysregulation of the proliferation and migration of vascular smooth muscle cells (VSMCs) is a crucial cause of atherosclerosis. MiR-637 exerts an antiproliferative effect on multiple human cells. Its impact on atherosclerosis remains largely unexplored. Methods Real-time PCR was used to determine miR-637 expression in samples from atherosclerosis patients and animal models. Its expression in VSMC dysfunction models (induced by ox-LDL) was also measured. The proliferation and migration of VSMCs were respectively tested using CCK-8 and Transwell assays, and apoptosis was measured using flow cytometry. The Targetscan database was used to predict the target genes of miR-637. Interaction between miR-637 and the potential target gene was validated via real-time PCR, western blotting and a luciferase reporter assay. Results MiR-637 expression was significantly lower in atherosclerosis patient and animal model samples. It also decreased in a dose- and time-dependent manner in animal models with ox-LDL-induced atherosclerosis. Transfection with miR-637 mimics suppressed the proliferation and migration of VSMCs while promoting apoptosis, while transfection with miR-637 inhibitors had the opposite effects. We also validated that insulin-like growth factor-2 (IGF-2), a crucial factor in the pathogenesis of atherosclerosis, serves as a target gene for miR-637. Conclusion MiR-637 targeting IGF-2 contributes to atherosclerosis inhibition and could be a potential target for this disease.
Collapse
Affiliation(s)
- Ning Yang
- Department of Cardiology, Tianjin Chest hospital, Taierzhuang South Road No.261, Jinnan District, Tianjin, 300222 China
| | - Bo Dong
- Department of Cardiology, Tianjin Chest hospital, Taierzhuang South Road No.261, Jinnan District, Tianjin, 300222 China
| | - Yanqiu Song
- Department of Cardiology, Tianjin Chest hospital, Taierzhuang South Road No.261, Jinnan District, Tianjin, 300222 China
| | - Yang Li
- Department of Cardiology, Tianjin Chest hospital, Taierzhuang South Road No.261, Jinnan District, Tianjin, 300222 China
| | - Lu Kou
- Department of Cardiology, Tianjin Chest hospital, Taierzhuang South Road No.261, Jinnan District, Tianjin, 300222 China
| | - Jingyu Yang
- Department of Cardiology, Tianjin Chest hospital, Taierzhuang South Road No.261, Jinnan District, Tianjin, 300222 China
| | - Qin Qin
- Department of Cardiology, Tianjin Chest hospital, Taierzhuang South Road No.261, Jinnan District, Tianjin, 300222 China
| |
Collapse
|
27
|
Lv Y, Duanmu J, Fu X, Li T, Jiang Q. Identifying a new microRNA signature as a prognostic biomarker in colon cancer. PLoS One 2020; 15:e0228575. [PMID: 32049961 PMCID: PMC7015317 DOI: 10.1371/journal.pone.0228575] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/17/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The aim was to identify a novel prognostic miRNA signature for colon cancer (CC) in silico. METHODS Data on the expression of miRNAs and relevant clinical information for 407 patients were obtained from The Cancer Genome Atlas (TCGA), and the samples were randomly split into a validation set (n = 203) and training set (n = 204). The differential expression of miRNAs between normal tissues and patients with CC was analyzed. We detected a miRNA expression signature in the training dataset by using a Cox proportional hazard regression model. Then, we verified the signature in the validation set. Association of the miRNA signature with overall survival was assessed in the validation cohort and combined cohort by log-rank test and based on Kaplan-Meier curves. The receiver operating characteristic and disease-free survival analyses were performed to evaluate the miRNA signature of CC in the combined cohort. Multivariate and univariate Cox analyses related to survival for the miRNA signature were performed, and a nomogram was built as a prognostic model for CC. To explore the function of target genes of the miRNA signature, Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis were used. RESULTS Between the matched normal tissues and colon cancer tissues, 267 differentially expressed miRNAs were detected, and a single-factor CoxPH model showed that 13 miRNAs were related to overall survival in the training cohort. Then, a five-miRNA signature was identified using a CoxPH regression model with multiple factors. The five-miRNA signature had significant prognostic value in the training cohort and was validated in the validation cohort and combined cohort. A total of 193 target genes of the miRNA signature were identified. According to the results of functional analysis of the target genes, the signaling pathways MAPK, AMPK and PI3K-Akt, focal adhesion, and microRNAs in cancer were remarkably enriched. CONCLUSION A five-miRNA signature had increased prognostic value for CC, which may provide important biological insights for the discovery and development of molecular predictors to improve the prognosis of patients with CC.
Collapse
Affiliation(s)
- Yunxia Lv
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Jinzhong Duanmu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Xiaorui Fu
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Taiyuan Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Qunguang Jiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
- * E-mail:
| |
Collapse
|
28
|
The IGF-II-Insulin Receptor Isoform-A Autocrine Signal in Cancer: Actionable Perspectives. Cancers (Basel) 2020; 12:cancers12020366. [PMID: 32033443 PMCID: PMC7072655 DOI: 10.3390/cancers12020366] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 12/18/2022] Open
Abstract
Insulin receptor overexpression is a common event in human cancer. Its overexpression is associated with a relative increase in the expression of its isoform A (IRA), a shorter variant lacking 11 aa in the extracellular domain, conferring high affinity for the binding of IGF-II along with added intracellular signaling specificity for this ligand. Since IGF-II is secreted by the vast majority of malignant solid cancers, where it establishes autocrine stimuli, the co-expression of IGF-II and IRA in cancer provides specific advantages such as apoptosis escape, growth, and proliferation to those cancers bearing such a co-expression pattern. However, little is known about the exact role of this autocrine ligand–receptor system in sustaining cancer malignant features such as angiogenesis, invasion, and metastasis. The recent finding that the overexpression of angiogenic receptor kinase EphB4 along with VEGF-A is tightly dependent on the IGF-II/IRA autocrine system independently of IGFIR provided new perspectives for all malignant IGF2omas (those aggressive solid cancers secreting IGF-II). The present review provides an updated view of the IGF system in cancer, focusing on the biology of the autocrine IGF-II/IRA ligand–receptor axis and supporting its underscored role as a malignant-switch checkpoint target.
Collapse
|
29
|
Dalton S, Smith K, Singh K, Kaiser H, Kolhe R, Mondal AK, Khayrullin A, Isales CM, Hamrick MW, Hill WD, Fulzele S. Accumulation of kynurenine elevates oxidative stress and alters microRNA profile in human bone marrow stromal cells. Exp Gerontol 2020; 130:110800. [PMID: 31790802 PMCID: PMC6998036 DOI: 10.1016/j.exger.2019.110800] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 02/09/2023]
Abstract
Kynurenine, a metabolite of tryptophan breakdown, has been shown to increase with age, and plays a vital role in a number of age-related pathophysiological changes, including bone loss. Accumulation of kynurenine in bone marrow stromal cells (BMSCs) has been associated with a decrease in cell proliferation and differentiation, though the exact mechanism by which kynurenine mediates these changes is poorly understood. MiRNAs have been shown to regulate BMSC function, and accumulation of kynurenine may alter the miRNA expression profile of BMSCs. The aim of this study was to identify differentially expressed miRNAs in human BMSCs in response to treatment with kynurenine, and correlate miRNAs function in BMSCs biology through bioinformatics analysis. Human BMSCs were cultured and treated with and without kynurenine, and subsequent miRNA isolation was performed. MiRNA array was performed to identify differentially expressed miRNA. Microarray analysis identified 50 up-regulated, and 36 down-regulated miRNAs in kynurenine-treated BMSC cultures. Differentially expressed miRNA included miR-1281, miR-330-3p, let-7f-5p, and miR-493-5p, which are important for BMSC proliferation and differentiation. KEGG analysis found up-regulated miRNA targeting glutathione metabolism, a pathway critical for removing oxidative species. Our data support that the kynurenine dependent degenerative effect is partially due to changes in the miRNA profile of BMSCs.
Collapse
Affiliation(s)
- Sherwood Dalton
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Kathryn Smith
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Kanwar Singh
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Helen Kaiser
- Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Ravindra Kolhe
- Departments of Pathology, Augusta University, Augusta, GA 30912, United States of America
| | - Ashis K Mondal
- Departments of Pathology, Augusta University, Augusta, GA 30912, United States of America
| | - Andrew Khayrullin
- Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC, 29403, United States of America
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America.
| |
Collapse
|
30
|
Du J, Zhang G, Qiu H, Yu H, Yuan W. The novel circular RNA circ-CAMK2A enhances lung adenocarcinoma metastasis by regulating the miR-615-5p/fibronectin 1 pathway. Cell Mol Biol Lett 2019; 24:72. [PMID: 31889960 PMCID: PMC6933717 DOI: 10.1186/s11658-019-0198-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Circular RNA (circRNA) has recently been considered as a key regulator in carcinogenesis. In this study, we investigated the functional significance and regulatory role of circ-CAMK2A (hsa_circ_0128332) in lung adenocarcinoma (LUAD). METHODS GSE101586 was employed to screen differentially expressed circRNAs. = Relative expression levels of circ-CAMK2A, miR-615-5p, fibronectin 1 (FN1), MMP2, and MMP9 were tested by quantitative reverse transcription PCR (qRT-PCR) or western blotting. Functional experiments were performed by CCK-8, wound healing, and transwell assays. Luciferase reporter and biotin-labeled RNA pull-down assays were carried out to evaluate the interaction between circ-CAMK2A, miR-615-5p, and fibronectin 1. In addition, a lung metastasis model was constructed to determine the metastasis-promoting role of circ-CAMK2A in vivo. RESULTS Circ-CAMK2A overexpression was observed in LUAD and was closely associated with lymph node metastasis, distant metastasis, advanced clinical stage, and poor prognosis. Circ-CAMK2A silencing evidently inhibited LUAD cell migration and invasion, whereas circ-CAMK2A overexpression had an opposite effect. Importantly, overexpression of circ-CAMK2A also enhanced LUAD metastasis in vivo. Mechanistically, miR-615-5p was identified as a direct target of circ-CAMK2A. Circ-CAMK2A up-regulates the expression level of fibronectin 1 by sponging miR-615-5p, thereby increasing MMP2 and MMP9 expression to promote the metastasis of LUAD. CONCLUSION Circ-CAMK2A plays a crucial role in the metastasis of LUAD, at least partially, by regulating the miR-615-5p/fibronectin 1 axis.
Collapse
Affiliation(s)
- Jiahui Du
- Minimally invasive surgery, Henan Provincial Chest Hospital, No. 1 Weiwu Road, Jinshui District, Zhengzhou, 450000 People’s Republic of China
| | - Guangzhao Zhang
- Minimally invasive surgery, Henan Provincial Chest Hospital, No. 1 Weiwu Road, Jinshui District, Zhengzhou, 450000 People’s Republic of China
| | - Hongli Qiu
- Minimally invasive surgery, Henan Provincial Chest Hospital, No. 1 Weiwu Road, Jinshui District, Zhengzhou, 450000 People’s Republic of China
| | - Haifeng Yu
- Minimally invasive surgery, Henan Provincial Chest Hospital, No. 1 Weiwu Road, Jinshui District, Zhengzhou, 450000 People’s Republic of China
| | - Wuying Yuan
- Minimally invasive surgery, Henan Provincial Chest Hospital, No. 1 Weiwu Road, Jinshui District, Zhengzhou, 450000 People’s Republic of China
| |
Collapse
|
31
|
Elevated miR-615-3p Expression Predicts Adverse Clinical Outcome and Promotes Proliferation and Migration of Prostate Cancer Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2377-2388. [DOI: 10.1016/j.ajpath.2019.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 12/11/2022]
|
32
|
circPUM1 Promotes Tumorigenesis and Progression of Ovarian Cancer by Sponging miR-615-5p and miR-6753-5p. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:882-892. [PMID: 31751911 PMCID: PMC6881671 DOI: 10.1016/j.omtn.2019.09.032] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 07/31/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023]
Abstract
Circular RNAs (circRNAs) have been reported to participate in the molecular mechanism of human cancers. The PUM1 gene has been confirmed to be closely related to tumorigenesis and progression of ovarian cancer. In the present study, we explored the function and underlying molecular mechanism of circPUM1 in ovarian cancer. qRT-PCR analysis showed upregulation of circPUM1 in ovarian cancer tissues compared with normal ovaries. Gain- and loss-of-function experiments indicated that circPUM1 increased cell proliferation, migration, and invasion and inhibited cell apoptosis. Intraperitoneal injection of circPUM1-knockout tumor cells in nude mice resulted in a decrease in the metastatic ability of the tumor. Bioinformatics analysis and dual-luciferase reporter assays revealed that circPUM1 upregulated the expression of nuclear factor kappa B (NF-κB) and MMP2 by sponging miR-615-5p and miR-6753-5p. Further studies showed that exosomal circPUM1 acted on peritoneal mesothelial cells and increased tumor metastasis. In conclusion, our study indicates that circPUM1 not only promotes ovarian cancer proliferation, migration and invasion, but also acts on the peritoneum and contributes to metastasis of cancer in the form of cancer-derived exosomes.
Collapse
|
33
|
Rafieenia F, Abbaszadegan MR, Poursheikhani A, Razavi SMS, Jebelli A, Molaei F, Aghaee‐Bakhtiari SH. In silico evidence of high frequency of miRNA‐related SNPs in Esophageal Squamous Cell Carcinoma. J Cell Physiol 2019; 235:966-978. [DOI: 10.1002/jcp.29012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/31/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Fatemeh Rafieenia
- Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Arash Poursheikhani
- Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad Iran
| | | | - Amir Jebelli
- Stem Cell and Regenerative Medicine Research Department Iranian Academic Center for Education, Culture and Research (ACECR), Mashhad Branch Mashhad Iran
| | - Fatemeh Molaei
- Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Hamid Aghaee‐Bakhtiari
- Bioinformatics Research Group Mashhad University of Medical Sciences Mashhad Iran
- Department of Medical Biotechnology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
34
|
Icli B, Wu W, Ozdemir D, Li H, Cheng HS, Haemmig S, Liu X, Giatsidis G, Avci SN, Lee N, Guimaraes RB, Manica A, Marchini JF, Rynning SE, Risnes I, Hollan I, Croce K, Yang X, Orgill DP, Feinberg MW. MicroRNA-615-5p Regulates Angiogenesis and Tissue Repair by Targeting AKT/eNOS (Protein Kinase B/Endothelial Nitric Oxide Synthase) Signaling in Endothelial Cells. Arterioscler Thromb Vasc Biol 2019; 39:1458-1474. [PMID: 31092013 PMCID: PMC6594892 DOI: 10.1161/atvbaha.119.312726] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 04/29/2019] [Indexed: 12/16/2022]
Abstract
Objective- In response to tissue injury, the appropriate progression of events in angiogenesis is controlled by a careful balance between pro and antiangiogenic factors. We aimed to identify and characterize microRNAs that regulate angiogenesis in response to tissue injury. Approach and Results- We show that in response to tissue injury, microRNA-615-5p (miR-615-5p) is rapidly induced and serves as an antiangiogenic microRNA by targeting endothelial cell VEGF (vascular endothelial growth factor)-AKT (protein kinase B)/eNOS (endothelial nitric oxide synthase) signaling in vitro and in vivo. MiR-615-5p expression is increased in wounds of diabetic db/db mice, in plasma of human subjects with acute coronary syndromes, and in plasma and skin of human subjects with diabetes mellitus. Ectopic expression of miR-615-5p markedly inhibited endothelial cell proliferation, migration, network tube formation in Matrigel, and the release of nitric oxide, whereas miR-615-5p neutralization had the opposite effects. Mechanistic studies using transcriptomic profiling, bioinformatics, 3' untranslated region reporter and microribonucleoprotein immunoprecipitation assays, and small interfering RNA dependency studies demonstrate that miR-615-5p inhibits the VEGF-AKT/eNOS signaling pathway in endothelial cells by targeting IGF2 (insulin-like growth factor 2) and RASSF2 (Ras-associating domain family member 2). Local delivery of miR-615-5p inhibitors, markedly increased angiogenesis, granulation tissue thickness, and wound closure rates in db/db mice, whereas miR-615-5p mimics impaired these effects. Systemic miR-615-5p neutralization improved skeletal muscle perfusion and angiogenesis after hindlimb ischemia in db/db mice. Finally, modulation of miR-615-5p expression dynamically regulated VEGF-induced AKT signaling and angiogenesis in human skin organoids as a model of tissue injury. Conclusions- These findings establish miR-615-5p as an inhibitor of VEGF-AKT/eNOS-mediated endothelial cell angiogenic responses and that manipulating miR-615-5p expression could provide a new target for angiogenic therapy in response to tissue injury. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Basak Icli
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Winona Wu
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Denizhan Ozdemir
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Department of Medical Biology, Hacettepe University, Ankara, Turkey
| | - Hao Li
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Henry S. Cheng
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Stefan Haemmig
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Xin Liu
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Giorgio Giatsidis
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Seyma Nazli Avci
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Nathan Lee
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Raphael Boesch Guimaraes
- Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia (ICFUC), Porto Alegre, RS, Brazil
| | - Andre Manica
- Instituto de Cardiologia do Rio Grande do Sul, Fundação Universitária de Cardiologia (ICFUC), Porto Alegre, RS, Brazil
| | - Julio F Marchini
- Heart Institute, University of São Paulo Medical School, São Paulo, Brazil
| | - Stein Erik Rynning
- Rheumatology, Lillehamer Hospital for Rheumatic Diseases, Lillehamer, Norway
| | - Ivar Risnes
- Rheumatology, Lillehamer Hospital for Rheumatic Diseases, Lillehamer, Norway
| | - Ivana Hollan
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- Research Department, Lillehamer Hospital for Rheumatic Diseases, Lillehamer, Norway
| | - Kevin Croce
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | | | - Dennis P. Orgill
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
35
|
Xing Q, Li R, Xu A, Qin Z, Tang J, Zhang L, Tang M, Han P, Wang W, Qin C, Du M, Zhang W. Genetic variants in a long noncoding RNA related to Sunitinib Resistance predict risk and survival of patients with renal cell carcinoma. Cancer Med 2019; 8:2886-2896. [PMID: 31038847 PMCID: PMC6558481 DOI: 10.1002/cam4.2160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 02/22/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022] Open
Abstract
Objective LncARSR (lncRNA Activated in RCC with Sunitinib Resistance, ENST00000424980) is a newly identified lncRNA to promote the sunitinib resistance of renal cell carcinoma (RCC), which may contribute to tumorigenesis and progression. This study aimed to explore the association of lncARSR tagSNPs with the risk and prognosis of RCC. Methods In this study, a 2‐stage case‐control study was performed to evaluate the association between 2 tagging SNPs (rs1417080 and rs7859384) and RCC susceptibility. Odds ratios (ORs) and 95% confidence intervals (CIs) were obtained by unconditional logistic regression analyses. Different survival time was estimated by the Kaplan‐Meier method and compared by the Log‐rank test. Hazard ratios (HRs) and their 95% CIs were calculated to determine predictive factors by Cox proportion hazards model. Results When combing discovery and validation sets together, rs7859384 was determined to be significantly associated with the decreased RCC risk with all P < 0.05 in 4 models (co‐dominant model, additive model, dominant model and recessive model). stratified analyses showed prominent risk effect of SNP rs7859384 GA/GG genotypes was found in clinical subgroups of stage I and stage II (P = 0.009, OR = 0.77, 95% CI = 0.64‐0.94) and individuals with clear cell RCC (P = 0.014, OR = 0.79, 95% CI = 0.65‐0.95). A protective effect of SNP rs7859384 GA/GG genotypes was observed among individuals with BMI > 24 (P = 0.025, OR = 0.74, 95% CI = 0.56‐0.96), without hypertension (P = 0.037, OR = 0.79, 95% CI = 0.63‐0.99), without family history of cancer (P = 0.048, OR = 0.83, 95% CI = 0.68‐1.00). Survival analyses revealed individuals with GA/GG genotypes had higher survival rate compared with the corresponding AA wild genotypes in the dominant model (log‐rank P = 0.005, adjusted HR = 0.34, 95% CI = 0.16‐0.73). Conclusion This study suggests that rs7859384 of lncARSR was associated with RCC susceptibility and may act as a prognostic biomarker for patients with RCC.
Collapse
Affiliation(s)
- Qianwei Xing
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, Affiliated Hospital of Nantong University, Nantong, China
| | - Ran Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Aiming Xu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiqiang Qin
- Department of Urology Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jinyuan Tang
- Department of Urology, Jiangsu Province Hospital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing, China
| | - Lei Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Tang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peng Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chao Qin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mulong Du
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
36
|
Mungamuri SK. Targeting the epigenome as a therapeutic strategy for pancreatic tumors. THERANOSTIC APPROACH FOR PANCREATIC CANCER 2019:211-244. [DOI: 10.1016/b978-0-12-819457-7.00011-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
37
|
Dong Y, Huo X, Sun R, Liu Z, Huang M, Yang S. lncRNA Gm15290 promotes cell proliferation and invasion in lung cancer through directly interacting with and suppressing the tumor suppressor miR-615-5p. Biosci Rep 2018; 38:BSR20181150. [PMID: 30287504 PMCID: PMC6209606 DOI: 10.1042/bsr20181150] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 01/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been involved in occurrence and progression of multiple cancers. In the present study, we investigated the role of lncRNA Gm15290 in the proliferation and invasion of non-small cell lung cancer (NSCLC) cells. First, we found that lncRNA Gm15290 was markedly up-regulated in tumor tissues from NSCLC patients and NSCLC cell lines, compared with adjacent normal tissues and normal lung cell line HBE respectively. Then, different concentrations of pcDNA-Gm15290 expression vector and Gm15290 siRNA were respectively transfected into A549 NSCLC cells. Our results showed that overexpression of Gm15290 significantly increased the proliferation and invasion of A549 cells and suppressed cell apoptosis. Knockdown of Gm15290 suppressed A549 cell proliferation and invasion and promoted cell apoptosis. Subsequently, we explored the underlying mechanism through which Gm15290 promoted cell proliferation and invasion. The output of RNA hybrid bioinformatic tool revealed that Gm15290 potentially interacted with tumor suppressor miR-615-5p which displayed an opposite expression pattern in the cell lines and a strong negative correlation with the levels of Gm15290 in NSCLC patients (r2 = 0.9677, P<0.0001). The results of RNA pull-down assays confirmed that Gm15290 directly bound with miR-615-5p Gm15290 negatively regulated the expression of miR-615-5p and increased the protein levels of miR-615-5p target genes, including IGF2, AKT2, and SHMT2 Moreover, miR-615-5p mimic could antagonize the promoting effect of Gm15290 on cell proliferation and invasion.
Collapse
MESH Headings
- Adult
- Antagomirs/genetics
- Antagomirs/metabolism
- Apoptosis
- Base Sequence
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/surgery
- Cell Line, Tumor
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Glycine Hydroxymethyltransferase/genetics
- Glycine Hydroxymethyltransferase/metabolism
- Humans
- Insulin-Like Growth Factor II/genetics
- Insulin-Like Growth Factor II/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/surgery
- Male
- MicroRNAs/agonists
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Neoplasm Invasiveness
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Yu Dong
- Department of Respiratory Medicine, Xi'an Central Hospital, Xi'an, China
- Department of Respiration Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoying Huo
- Department of Respiratory Medicine, Fourth Hospital of Xi'an, Xi'an, China
| | - Ruiying Sun
- Department of Respiration Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhiyan Liu
- Department of Respiratory Medicine, Xi'an Central Hospital, Xi'an, China
| | - Miaoyi Huang
- Department of Respiratory Medicine, Xi'an Central Hospital, Xi'an, China
| | - Shuanying Yang
- Department of Respiration Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
38
|
Epigenetically regulated miR-1247 functions as a novel tumour suppressor via MYCBP2 in methylator colon cancers. Br J Cancer 2018; 119:1267-1277. [PMID: 30318507 PMCID: PMC6251029 DOI: 10.1038/s41416-018-0249-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/24/2018] [Accepted: 08/03/2018] [Indexed: 01/27/2023] Open
Abstract
Background Colorectal cancer (CRC) is a heterogeneous disease with distinct clinical subsets based on underlying genetic and epigenetic changes. DNA hypermethylation yields a unique CRC subset with a distinct phenotype and clinical behaviour, but this oncogenic pathway is not fully characterised. This study identifies and characterises miR-1247 as a novel tumour suppressor microRNA in methylated human colon cancers. Method Tumour samples from patients with hypermethylated and non-methylated colon cancer and cell lines were evaluated for miR-1247 expression and function. A murine subcutaneous xenograft model was used for in vivo functional studies. Results miR-1247 was methylated and underexpressed in methylator colon cancers. Overexpression of miR-1247 significantly inhibited cell proliferation, decreased tumour cell motility, induced apoptosis, and mitigated tumour formation capacity both in vivo and in vitro. Pharmacologic demethylation increased miR-1247 expression and produced similar anti-tumour activities. Mechanistic investigations revealed that MYCBP2, a member of the c-myc oncogene family, is a direct functional target of miR-1247. Furthermore, in CRC patients, MYCBP2 protein levels are associated with miR-1247 levels and survival. Conclusions miR-1247 acts as a tumour suppressor by inhibiting MYCBP2 in methylator colon cancer. The MYCBP2/c-myc axis may underlie the anti-tumour activities of miR-1247 and is a potential therapeutic target via demethylation agents.
Collapse
|
39
|
Wang Q, Wu G, Zhang Z, Tang Q, Zheng W, Chen X, Chen F, Li Q, Che X. Long non-coding RNA HOTTIP promotes renal cell carcinoma progression through the regulation of the miR-615/IGF-2 pathway. Int J Oncol 2018; 53:2278-2288. [PMID: 30226576 DOI: 10.3892/ijo.2018.4539] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 07/27/2018] [Indexed: 11/06/2022] Open
Abstract
Emerging evidence has indicated that long non‑coding RNA (lncRNA) HOXA transcript at the distal tip (HOTTIP) regulates cell growth, differentiation, apoptosis and cancer progression. However, the expression and function of HOTTIP in the progression of renal cell carcinoma (RCC) remain largely unknown. In this study, we investigated the role of the lncRNA HOTTIP in RCC. The expression levels of HOTTIP in RCC tissues and cell lines were determined by RT‑qPCR. The association between HOTTIP expression and clinicopathological characteristics and prognosis was analyzed in patients with RCC from the TCGA database. Loss‑of‑ function assays were designed and conducted to verify the oncogenic function of HOTTIP in RCC progression. Luciferase assay was performed to explore the mechanisms of the miRNA‑lncRNA sponge. The results revealed that HOTTIP expression was upregulated in RCC. An increased HOTTIP expression in RCC was associated with a larger tumor size and a higher clinical stage, lymph node metastasis and vascular invasion. Additionally, patients RCC with a high HOTTIP expression had a significantly shorter overall survival (OS) and disease‑free survival (DFS). HOTTIP knockdown significantly inhibited cell proliferation, migration and invasion, and increased the apoptosis of RCC cells in vitro. Mechanistic analyses revealed that HOTTIP functioned as a competing endogenous RNA (ceRNA) for hsa‑miR‑615‑3p, and led to the derepression of its endogenous target, insulin‑like growth factor-2 (IGF‑2), which is a protein hormone that exerts a stimulatory effect on tumor cell growth. miR‑615 inhibition reversed the suppressive effects of HOTTIP knockdown on RCC cell progression. HOTTIP regulated IGF‑2 expression in a miR‑615‑dependent manner in RCC cells. In addition, IGF‑2 expression was significantly upregulated in the RCC specimens and a positive association between the expression of HOTTIP and IGF‑2 in RCC tissues was detected. The effect of HOTTIP was abolished by the siRNA‑mediated silencing of IGF-2 in RCC cells. On the whole, this study demonstrates, for the first time, at least to the best of our knowledge, that the HOTTIP/miR‑615/IGF‑2 axis plays an important role in RCC progression and potentially contributes to the improvement of RCC diagnosis and therapy.
Collapse
Affiliation(s)
- Qifei Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Zhiwei Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Qizhen Tang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Wei Zheng
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xiaochi Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Feng Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Quanlin Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
40
|
Botti G, De Chiara A, Di Bonito M, Cerrone M, Malzone MG, Collina F, Cantile M. Noncoding RNAs within the
HOX
gene network in tumor pathogenesis and progression. J Cell Physiol 2018; 234:395-413. [DOI: 10.1002/jcp.27036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Gerardo Botti
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Anna De Chiara
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Maurizio Di Bonito
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Margherita Cerrone
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Maria Gabriella Malzone
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Francesca Collina
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Monica Cantile
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| |
Collapse
|
41
|
Ho J, Li X, Zhang L, Liang Y, Hu W, Yau JCW, Chan H, Gin T, Chan MTV, Tse G, Wu WKK. Translational genomics in pancreatic ductal adenocarcinoma: A review with re-analysis of TCGA dataset. Semin Cancer Biol 2018; 55:70-77. [PMID: 29705685 DOI: 10.1016/j.semcancer.2018.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/16/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Malignancy of the pancreas is a leading cause of cancer-related mortality, with the highest case-fatality of all cancers. Nevertheless, the lack of sensitive biomarkers and presence of biological heterogeneity precludes its early detection and effective treatment. The recent introduction of next-generation sequencing allows characterization of multiple driver mutations at genome- and exome-wide levels. Sequencing of DNA and RNA from circulating tumour cells has also opened an exciting era of non-invasive procedures for tumour detection and prognostication. This massively-parallel sequencing technology has uncovered the previously obscure molecular mechanisms, providing clues for better stratification of patients and identification of druggable targets for the disease. Identification of active oncogenic pathways and gene-gene interactions may reveal oncogene addiction and synthetic lethality. Relevant findings can be extrapolated to develop targeted and personalized therapeutic interventions. In addition to known mutational events, the role of chromosomal rearrangements in pancreatic neoplasms is gradually uncovered. Coupled with bioinformatics pipelines and epidemiological analyses, a better framework for risk stratification and prognostication of pancreatic cancer will be possible in the near future. In this review, we discuss how translational genomic studies facilitate our understanding of pathobiology, and development of novel diagnostics and therapeutics for pancreatic ductal adenocarcinoma with emphases on whole genome sequencing, whole exome sequencing, and liquid biopsies. We have also re-analyzed The Cancer Genome Atlas (TCGA) dataset to look for genetic features associated with altered survival in patients with pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Jeffery Ho
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Xianchun Li
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 00060, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Yonghao Liang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Hu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Johnny C W Yau
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Hung Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Tony Gin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China.
| | - Gary Tse
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - William K K Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
42
|
Ji Y, Sun Q, Zhang J, Hu H. MiR-615 inhibits cell proliferation, migration and invasion by targeting EGFR in human glioblastoma. Biochem Biophys Res Commun 2018; 499:719-726. [PMID: 29605294 DOI: 10.1016/j.bbrc.2018.03.217] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023]
Abstract
MiR-615 and epidermal growth factor receptor (EGFR) are associated with a number of disease processes and pathogenesis. However, little is known about the mechanisms of miR-615 and EGFR in human glioblastoma multiforme (GBM). Here, we found that down-regulation of miR-615 expression occurred in GBM tissues and cells, and was inversely correlated with overall survival, relapse-free survival, WHO grade as well as EGFR expression. We further determined that miR-615 functions as a tumor suppressor by inhibiting GBM cell proliferation, cell cycle, migration and invasion, and promoting cell apoptosis. In-vivo assay validated the inhibition effect of miR-615 on tumor growth and EGFR expression. Luciferase reporter assays demonstrated that miR-615 targeted the 3'-untranslated region (3'-UTR) of EGFR. Besides, over-expression of EGFR reversed the inhibition effects of miR-615, while silencing of EGFR aggravated these inhibition effects. In conclusions, we identified that miR-615 plays a tumor suppressor role in GBM cell proliferation, migration and invasion by targeting EGFR expression, and miR-615 may act as a novel biomarker for early diagnosis or therapeutic targets of GBM.
Collapse
Affiliation(s)
- Yanwei Ji
- Department of Neurosurgery, Shandong Provincial Third Hospital, No.12, Central Wuying Hill Road, Jinan 250031, Shandong, China
| | - Qingshan Sun
- Department of Orthopedics, Shandong Provincial Third Hospital, No.12, Central Wuying Hill Road, Jinan 250031, Shandong, China
| | - Jianbin Zhang
- Department of Neurosurgery, Shandong Provincial Third Hospital, No.12, Central Wuying Hill Road, Jinan 250031, Shandong, China
| | - Haoran Hu
- Department of Gerontology, Shandong Provincial Third Hospital, No.12, Central Wuying Hill Road, Jinan 250031, Shandong, China.
| |
Collapse
|
43
|
|
44
|
Epigenetic inhibition of miR-663b by long non-coding RNA HOTAIR promotes pancreatic cancer cell proliferation via up-regulation of insulin-like growth factor 2. Oncotarget 2018; 7:86857-86870. [PMID: 27895308 PMCID: PMC5349959 DOI: 10.18632/oncotarget.13490] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/06/2016] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer is one of the most deadly cancers with a poor prognosis. Although microRNAs are involving in the carcinogenesis and development of pancreatic cancer, little information is known regarding the role of miR-663b in pancreatic cancer. In this study, the expression of miR-663b in pancreatic cancer cells was down-regulated by hypermethylation in its putative promoter region, and overexpression of miR-663b repressed cell proliferation, invasion and migration, and induced apoptosis in pancreatic cancer cells. Bioinformatics analysis, luciferase report assay and rescue experiments showed that insulin-like growth factor 2 (IGF2) was a direct target of miR-663b. Results from clinical samples showed that the expression level of miR-663b correlated with the pathological grading, and the expression of miR-663b was down-regulated and was inversely correlated with IGF2 expression level in pancreatic cancer tissues. More importantly, the long non-coding RNA, HOX transcript antisense RNA (HOTAIR), was up-regulated in both pancreatic cancer cells and tissues, and HOTAIR suppressed the expression of miR-663b in pancreatic cancer by histone modification on H3K4me3 and H3K27me3 on miR-663b promoter. Further in vivo studies demonstrated that the stable overexpression of miR-663b or knock-down of HOTAIR inhibited tumor growth and was associated with IGF2 expression. In summary, our studies demonstrated that miR-663b is epigenetically repressed by HOTAIR and exerts its tumor-suppressive function via targeting IGF2 in pancreatic cancer.
Collapse
|
45
|
Epigenetics and MicroRNAs in Cancer. Int J Mol Sci 2018; 19:ijms19020459. [PMID: 29401683 PMCID: PMC5855681 DOI: 10.3390/ijms19020459] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 02/08/2023] Open
Abstract
The ability to reprogram the transcriptional circuitry by remodeling the three-dimensional structure of the genome is exploited by cancer cells to promote tumorigenesis. This reprogramming occurs because of hereditable chromatin chemical modifications and the consequent formation of RNA-protein-DNA complexes that represent the principal actors of the epigenetic phenomena. In this regard, the deregulation of a transcribed non-coding RNA may be both cause and consequence of a cancer-related epigenetic alteration. This review summarizes recent findings that implicate microRNAs in the aberrant epigenetic regulation of cancer cells.
Collapse
|
46
|
A 4-miRNA signature predicts the therapeutic outcome of glioblastoma. Oncotarget 2018; 7:45764-45775. [PMID: 27302927 PMCID: PMC5216759 DOI: 10.18632/oncotarget.9945] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/22/2016] [Indexed: 01/15/2023] Open
Abstract
Multimodal therapy of glioblastoma (GBM) reveals inter-individual variability in terms of treatment outcome. Here, we examined whether a miRNA signature can be defined for the a priori identification of patients with particularly poor prognosis. FFPE sections from 36 GBM patients along with overall survival follow-up were collected retrospectively and subjected to miRNA signature identification from microarray data. A risk score based on the expression of the signature miRNAs and cox-proportional hazard coefficients was calculated for each patient followed by validation in a matched GBM subset of TCGA. Genes potentially regulated by the signature miRNAs were identified by a correlation approach followed by pathway analysis. A prognostic 4-miRNA signature, independent of MGMT promoter methylation, age, and sex, was identified and a risk score was assigned to each patient that allowed defining two groups significantly differing in prognosis (p-value: 0.0001, median survival: 10.6 months and 15.1 months, hazard ratio = 3.8). The signature was technically validated by qRT-PCR and independently validated in an age- and sex-matched subset of standard-of-care treated patients of the TCGA GBM cohort (n=58). Pathway analysis suggested tumorigenesis-associated processes such as immune response, extracellular matrix organization, axon guidance, signalling by NGF, GPCR and Wnt. Here, we describe the identification and independent validation of a 4-miRNA signature that allows stratification of GBM patients into different prognostic groups in combination with one defined threshold and set of coefficients that could be utilized as diagnostic tool to identify GBM patients for improved and/or alternative treatment approaches.
Collapse
|
47
|
Epigenetic modification of miR-141 regulates SKA2 by an endogenous 'sponge' HOTAIR in glioma. Oncotarget 2017; 7:30610-25. [PMID: 27121316 PMCID: PMC5058705 DOI: 10.18632/oncotarget.8895] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 03/31/2016] [Indexed: 12/21/2022] Open
Abstract
Aberrant expression of miR-141 has recently implicated in the occurrence and development of various types of malignant tumors. However whether the involvement of miR-141 in the pathogenesis of glioma remains unknown. Here, we showed that miR-141 was markedly downregulated in glioma tissues and cell lines compared with normal brain tissues, and its expression correlated with the pathological grading. Enforced expression of miR-141 in glioma cells significantly inhibited cell proliferation, migration and invasion, whereas knockdown of miR-141 exerted opposite effect. Mechanistic investigations revealed that HOTAIR might act as an endogenous 'sponge' of miR-141, thereby regulating the derepression of SKA2. Further, we explored the molecular mechanism by which miR-141 expression was regulated, and found that the miR-141 promoter was hypermethylated and that promoter methylation of miR-141 was mediated by DNMT1 in glioma cells. Finally, both overexpression of miR-141 and knockdown of HOTAIR in a mouse model of human glioma resulted in significant reduction of tumor growth in vivo. Collectively, these results suggest that epigenetic modification of miR-141 and the interaction of ceRNA regulatory network will provide a new approach for therapeutics against glioma.
Collapse
|
48
|
KDM4B-mediated epigenetic silencing of miRNA-615-5p augments RAB24 to facilitate malignancy of hepatoma cells. Oncotarget 2017; 8:17712-17725. [PMID: 27487123 PMCID: PMC5392280 DOI: 10.18632/oncotarget.10832] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 06/17/2016] [Indexed: 11/25/2022] Open
Abstract
Emerging evidence indicates that dysregulation of microRNAs (miRNAs) contributes to hepatocellular carcinoma (HCC) tumorigenesis and development. Here, we found that miR-615-5p was obviously downregulated in HCC. Furthermore, the deficiency of demethylase KDM4B stimulated the CpG methylation of miR-615-5p promoter and then decreased the miR-615-5p expression. The Ras-related protein RAB24 was found to be downregulated by miR-615-5p. The low level of miR-615-5p increased the expression of RAB24 and facilitated HCC growth and metastasis in vitro and in vivo. Moreover, miR-615-5p suppresses HCC cell growth by influencing cell cycle progression and apoptosis. Downregulation of miR-615-5p and upregulation of RAB24 promotes the epithelial-mesenchymal transition (EMT), adhesion and vasculogenic mimicry (VM) of HCC cells, all of which contribute to cell motility and metastasis. Thus, miR-615-5p, who is downregulated by KDM4B-mediated hypermethylation in its promoter, functions as a tumor suppressor by inhibiting RAB24 expression in HCC. In conclusion, our findings characterize miR-615-5p as an important epigenetically silenced miRNA involved in the Rab-Ras pathway in hepatocellular carcinoma and expand our understanding of the molecular mechanism underlying hepatocarcinogenesis and metastasis.
Collapse
|
49
|
You Y, Tan J, Gong Y, Dai H, Chen H, Xu X, Yang A, Zhang Y, Bie P. MicroRNA-216b-5p Functions as a Tumor-suppressive RNA by Targeting TPT1 in Pancreatic Cancer Cells. J Cancer 2017; 8:2854-2865. [PMID: 28928875 PMCID: PMC5604218 DOI: 10.7150/jca.18931] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 05/01/2017] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miRNAs) are increasingly recognized as being involved in pancreatic cancer progression by directly regulating the expression of their targets. In this study, we showed that miR-216b-5p expression was significantly decreased in pancreatic cancer tissues and cell lines. In addition, low miR-216b-5p expression was significantly associated with large tumor size and advanced TNM stage. Meanwhile, both Kaplan-Meier and multivariate survival analysis showed that decreased miR-216b-5p expression was associated with overall survival. miR-216b-5p over-expression repressed pancreatic cancer cell proliferation and induced cell cycle arrest and cell apoptosis in vitro and inhibited tumorigenesis in vivo. The translationally controlled tumor protein (TPT1) was identified as a novel direct target of miR-216b-5p. miR-216b-5p up-regulation suppressed TPT1 expression. Moreover, TPT1 mRNA expression levels were increased in pancreatic cancer tissues, and were inversely correlated with miR-216b-5p expression. TPT1 down-regulation had similar effects as miR-216b-5p up-regulation on pancreatic cancer cell progression. The restoration of TPT1 reversed the effect of miR-216b-5p on pancreatic cancer cell progression. Furthermore, we found that miR-216b-5p up-regulation suppressed Pim-3, Cyclin B1, p-Bad and Bcl-xL protein expression. However, the effect of miR-216b-5p up-regulation was partly reversed by TPT1 up-regulation in vitro. Taken together, our findings suggested that miR-216b-5p functions as a potential tumor suppressor by regulating TPT1 in pancreatic cancer cells, and it may represent a potential therapeutic target for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Yu You
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Jiaxin Tan
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yi Gong
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Haisu Dai
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Haowei Chen
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Xuejun Xu
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Aigang Yang
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yujun Zhang
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Ping Bie
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
50
|
Wu F, Yuan G, Chen J, Wang C. Network analysis based on TCGA reveals hub genes in colon cancer. Contemp Oncol (Pozn) 2017; 21:136-144. [PMID: 28947883 PMCID: PMC5611503 DOI: 10.5114/wo.2017.68622] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/17/2017] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is the third most widespread cancer in the world. Although many advances have been made in molecular biology, novel approaches are still required to reveal molecular mechanisms for the diagnosis and therapy of colon cancer. In this study, we aimed to determine and analyse the hub genes of CRC. First, we explored the mRNA and microRNA (miRNA) expression profiles of colon carcinoma, then we screened target genes of differentially expressed miRNAs and obtained the intersection between differently expressed genes and target genes. Gene Ontology (GO) classification and KEGG pathway analysis of differently expressed genes were performed, and gene-miRNA and TF-gene-miRNA networks were constructed to identify hub genes, miRNAs, and TFs. In total, 3436 significant differentially expressed genes (1709 upregulated and 1727 downregulated) and 216 differentially expressed miRNAs (99 upregulated and 117 downregulated) were identified in colon cancer. These differentially expressed genes were significantly enriched in GO terms and KEGG pathways, such as cell proliferation, cell adhesion, and cytokine-cytokine receptor interaction signalling pathways. GCNT4, EDN2, and so on were located in the central hub of the co-expression network. MYC, WT1, mir-34a, and LEF1 were located in the central hub of the network of TF-gene-miRNA. These findings increase our understanding of the molecular mechanisms of colon cancer and will aid in identifying potential targets for diagnostic and therapeutic usage.
Collapse
Affiliation(s)
- Fenzan Wu
- Science and Education Division, Cixi Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Guoping Yuan
- Clinical Laboratory, Cixi Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Junjie Chen
- Clinical Laboratory, Cixi Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Chengzu Wang
- Clinical Laboratory, Cixi Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|