1
|
Al-Shibli R, AlSuleimani M, Ahmed I, Al Lawati A, Das S. Association of miRNA and Bone Tumors: Future Therapeutic Inroads. Curr Med Chem 2025; 32:1103-1120. [PMID: 38299295 DOI: 10.2174/0109298673284932231226110754] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 02/02/2024]
Abstract
Small endogenous non-coding RNA molecules known as micro-ribonucleic acids (miRNAs) control post-transcriptional gene regulation. A change in miRNA expression is related to various diseases, including bone tumors. Benign bone tumors are categorized based on matrix production and predominant cell type. Osteochondromas and giant cell tumors are among the most common bone tumors. Interestingly, miRNAs can function as either tumor suppressor genes or oncogenes, thereby determining the fate of a tumor. In the present review, we discuss various bone tumors with regard to their prognosis, pathogenesis, and diagnosis. The association between miRNAs and bone tumors, such as osteosarcoma, Ewing's sarcoma, chondrosarcoma, and giant-cell tumors, is also discussed. Moreover, miRNA may play an important role in tumor proliferation, growth, and metastasis. Knowledge of the dysregulation, amplification, and deletion of miRNA can be beneficial for the treatment of various bone cancers. The miRNAs could be beneficial for prognosis, treatment, future drug design, and treatment of resistant cases of bone cancer.
Collapse
Affiliation(s)
- Rashid Al-Shibli
- Department of Medical, Sultan Qaboos University Hospital, Muscat, 123, Oman
| | | | - Ibrahim Ahmed
- Department of Medical, Sultan Qaboos University Hospital, Muscat, 123, Oman
| | - Abdullah Al Lawati
- Department of Medical, Sultan Qaboos University Hospital, Muscat, 123, Oman
| | - Srijit Das
- Department of Human & Clinical Anatomy, Sultan Qaboos University, Muscat, 123, Oman
| |
Collapse
|
2
|
Zhai F, Wang J, Luo X, Ye M, Jin X. Roles of NOLC1 in cancers and viral infection. J Cancer Res Clin Oncol 2023; 149:10593-10608. [PMID: 37296317 DOI: 10.1007/s00432-023-04934-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND The nucleolus is considered the center of metabolic control and an important organelle for the biogenesis of ribosomal RNA (rRNA). Nucleolar and coiled-body phosphoprotein 1(NOLC1), which was originally identified as a nuclear localization signal-binding protein is a nucleolar protein responsible for nucleolus construction and rRNA synthesis, as well as chaperone shuttling between the nucleolus and cytoplasm. NOLC1 plays an important role in a variety of cellular life activities, including ribosome biosynthesis, DNA replication, transcription regulation, RNA processing, cell cycle regulation, apoptosis, and cell regeneration. PURPOSE In this review, we introduce the structure and function of NOLC1. Then we elaborate its upstream post-translational modification and downstream regulation. Meanwhile, we describe its role in cancer development and viral infection which provide a direction for future clinical applications. METHODS The relevant literatures from PubMed have been reviewed for this article. CONCLUSION NOLC1 plays an important role in the progression of multiple cancers and viral infection. In-depth study of NOLC1 provides a new perspective for accurate diagnosis of patients and selection of therapeutic targets.
Collapse
Affiliation(s)
- Fengguang Zhai
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China
| | - Jie Wang
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China
| | - Xia Luo
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China.
| |
Collapse
|
3
|
Shi WZ, Li W, Cheng Y, Zhang M, Niu XC, Gao QW, Lu Y, Tian T, Du S, Mi Y, Chang MZ, Tian Y. The cytoprotective role of omentin against oxidative stress-induced PC12 apoptosis. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2021; 49:483-492. [PMID: 34151664 DOI: 10.1080/21691401.2021.1892707] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/15/2021] [Indexed: 10/21/2022]
Abstract
Oxidative stress has been proven to play a critical role in the pathogenesis of neuronal injury. As a novel adipocytokine, omentin is produced by visceral adipose with insulin sensitizing effects and has been revealed to possess anti-inflammatory effects. However, the possible effect of omentin on oxidative stress remains unknown. The present study aimed to detect the potential protective effect of omentin against hydrogen peroxide (H2O2)-induced cytotoxicity of PC12 cells. The results showed that no cytotoxic effect was shown in PC12 cells co-cultured with omentin alone at a concentration of 50-1000 ng/mL. The CCK8 and TUNEL assays suggested that omentin could remarkably attenuate apoptosis induced by 100 μM H2O2. The PCR and western blotting showed that the expression levels of Bax was significantly inhibited by omentin via the upregulation of miR-128-3p at its 3'-UTR. Taken together, these results indicated that omentin protects PC12 cells against H2O2-induced apoptosis, and further studies need to be conducted before utilization in the clinic for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Wen-Zhen Shi
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
| | - Wu Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
| | - Ye Cheng
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
| | - Meng Zhang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
| | - Xiao-Chen Niu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
| | - Qi-Wei Gao
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Ying Lu
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
- Medical Research and Experimental Center, School of Medicine, Yan'an University, Yan'an, Shaanxi, China
| | - Tian Tian
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
- Medical Research and Experimental Center, School of Medicine, Yan'an University, Yan'an, Shaanxi, China
| | - Shan Du
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
- Medical Research and Experimental Center, School of Medicine, Yan'an University, Yan'an, Shaanxi, China
| | - Yan Mi
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
- Medical Research and Experimental Center, School of Medicine, Yan'an University, Yan'an, Shaanxi, China
| | - Ming-Ze Chang
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
| | - Ye Tian
- Medical Research Center, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, The College of Life Sciences, Northwest University, Xi'an, China
- Medical Research and Experimental Center, School of Medicine, Yan'an University, Yan'an, Shaanxi, China
| |
Collapse
|
4
|
Fonseca A, Ramalhete SV, Mestre A, Pires das Neves R, Marreiros A, Castelo-Branco P, Roberto VP. Identification of colorectal cancer associated biomarkers: an integrated analysis of miRNA expression. Aging (Albany NY) 2021; 13:21991-22029. [PMID: 34547721 PMCID: PMC8507258 DOI: 10.18632/aging.203556] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022]
Abstract
Colorectal cancer is one of the leading causes of cancer-related deaths worldwide. This complex disease still holds severe problems concerning diagnosis due to the high invasiveness nature of colonoscopy and the low accuracy of the alternative diagnostic methods. Additionally, patient heterogeneity even within the same stage is not properly reflected in the current stratification system. This scenario highlights the need for new biomarkers to improve non-invasive screenings and clinical management of patients. MicroRNAs (miRNAs) have emerged as good candidate biomarkers in cancer as they are stable molecules, easily measurable and detected in body fluids thus allowing for non-invasive diagnosis and/or prognosis. In this study, we performed an integrated analysis first using 4 different datasets (discovery cohorts) to identify miRNAs associated with colorectal cancer development, unveil their role in this disease by identifying putative targets and regulatory networks and investigate their ability to serve as biomarkers. We have identified 26 differentially expressed miRNAs which interact with frequently deregulated genes known to participate in commonly altered pathways in colorectal cancer. Most of these miRNAs have high diagnostic power, and their prognostic potential is evidenced by panels of 5 miRNAs able to predict the outcome of stage II and III colorectal cancer patients. Notably, 8 miRNAs were validated in three additional independent cohorts (validation cohorts) including a plasma cohort thus reinforcing the value of miRNAs as non-invasive biomarkers.
Collapse
Affiliation(s)
- André Fonseca
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal
| | - Sara Ventura Ramalhete
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), Faro 8005-139, Portugal
| | - André Mestre
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), Faro 8005-139, Portugal
| | - Ricardo Pires das Neves
- CNC, Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-517, Portugal
- IIIUC-Institute of Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| | - Ana Marreiros
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), Faro 8005-139, Portugal
| | - Pedro Castelo-Branco
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), Faro 8005-139, Portugal
- Champalimaud Research Program, Champalimaud Center for the Unknown, Lisbon 1400-038, Portugal
| | - Vânia Palma Roberto
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), Faro 8005-139, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro 8005-139, Portugal
| |
Collapse
|
5
|
Wang JH, Shi CW, Lu YY, Zeng Y, Cheng MY, Wang RY, Sun Y, Jiang YL, Yang WT, Zhao DD, Huang HB, Ye LP, Cao X, Yang GL, Wang CF. MicroRNA and circRNA Expression Analysis in a Zbtb1 Gene Knockout Monoclonal EL4 Cell Line. Front Cell Infect Microbiol 2021; 11:706919. [PMID: 34290994 PMCID: PMC8287301 DOI: 10.3389/fcimb.2021.706919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/21/2021] [Indexed: 12/30/2022] Open
Abstract
Zinc finger and BTB domain containing 1(Zbtb1) is a transcriptional suppressor protein, and a member of the mammalian Zbtb gene family. Previous studies have shown that Zbtb1 is essential for T-cell development. However, the role of Zbtb1 in T-cell lymphoma is undetermined. In this study, an EL4 cell line with Zbtb1 deletion was constructed using the CRISPR-Cas9 technique. The expression profiles of microRNA and circRNA produced by the control and gene deletion groups were determined by RNA-seq. In general, 24 differentially expressed microRNA and 16 differentially expressed circRNA were found between normal group and gene deletion group. Through further analysis of differentially expressed genes, GO term histogram and KEGG scatter plot were drawn, and three pairs of miRNA and circRNA regulatory relationships were found. This study describes the differentially expressed microRNA and circRNA in normal and Zbtb1-deficient EL4 cell lines, thus providing potential targets for drug development and clinical treatment of T-cell lymphoma.
Collapse
Affiliation(s)
- Jun-Hong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yi-Yuan Lu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ming-Yang Cheng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ru-Yu Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yu Sun
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Dan-Dan Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Li-Ping Ye
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.,Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China.,Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
6
|
A Zic2/Runx2/NOLC1 signaling axis mediates tumor growth and metastasis in clear cell renal cell carcinoma. Cell Death Dis 2021; 12:319. [PMID: 33767130 PMCID: PMC7994417 DOI: 10.1038/s41419-021-03617-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/26/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most common malignancies with rapid growth and high metastasis, but lacks effective therapeutic targets. Here, using public sequencing data analyses, quantitative real-time PCR assay, western blotting, and IHC staining, we characterized that runt-related transcription factor 2 (Runx2) was significantly upregulated in ccRCC tissues than that in normal renal tissues, which was associated with the worse survival of ccRCC patients. Overexpression of Runx2 promoted malignant proliferation and migration of ccRCC cells, and inversely, interfering Runx2 with siRNA attenuates its oncogenic ability. RNA sequencing and functional studies revealed that Runx2 enhanced ccRCC cell growth and metastasis via downregulation of tumor suppressor nucleolar and coiled-body phosphoprotein 1 (NOLC1). Moreover, increased Zic family member 2 (Zic2) was responsible for the upregulation of Runx2 and its oncogenic functions in ccRCC. Kaplan-Meier survival analyses indicated that ccRCC patients with high Zic2/Runx2 and low NOLC1 had the worst outcome. Therefore, our study demonstrates that Zic2/Runx2/NOLC1 signaling axis promotes ccRCC progression, providing a set of potential targets and prognostic indicators for patients with ccRCC.
Collapse
|
7
|
CircPPP1R12A promotes the progression of colon cancer through regulating CTNNB1 via sponging miR-375. Anticancer Drugs 2021; 32:635-646. [PMID: 33595945 DOI: 10.1097/cad.0000000000001037] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Circular RNAs (circRNAs) have been identified as potential biomarkers for many cancer, including colon cancer (CC). However, the function and mechanism of circPPP1R12A in CC have not been fully elucidated. Quantitative real-time PCR was employed to assess the expression of circPPP1R12A, microRNA (miR)-375 and catenin beta-1 (CTNNB1). The proliferation, apoptosis, migration and invasion of cells were determined using colony formation assay, flow cytometry, wound healing assay and transwell assay. The protein levels of cell cyclin-related markers and CTNNB1 were detected by western blot analysis. The interaction between miR-375 and circPPP1R12A or CTNNB1 was verified by dual-luciferase reporter assay. Xenograft models were built to evaluate the effect of circPPP1R12A silencing and CTNNB1 overexpression on CC tumor growth in vivo. Our results showed that circPPP1R12A was a highly expressed circRNA in CC tissues and cells. Silenced circPPP1R12A suppressed the proliferation, promoted the apoptosis, and inhibited the migration and invasion of CC cells. MiR-375 could be sponged by circPPP1R12A, and its inhibitor could reverse the inhibition of circPPP1R12A silencing on CC progression. Furthermore, CTNNB1 was a target of miR-375, and its overexpression also abolished the suppression of miR-375 on CC progression. Moreover, circPPP1R12A indirectly regulated CTNNB1 expression by sponging miR-375. Importantly, circPPP1R12A knockdown reduced the tumor growth of CC in vivo, and this effect also could be reversed by overexpressing CTNNB1. Our study proposed that circPPP1R12A might play an oncogenic role in CC, which could act as a potential therapeutic target for CC.
Collapse
|
8
|
Morris NL, Choudhry MA. Maintenance of gut barrier integrity after injury: Trust your gut microRNAs. J Leukoc Biol 2021; 110:979-986. [PMID: 33577717 DOI: 10.1002/jlb.3ru0120-090rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract is a highly dynamic structure essential for digestion, nutrient absorption, and providing an interface to prevent gut bacterial translocation. In order to maintain the barrier function, the gut utilizes many defense mechanisms including proliferation, apoptosis, and apical junctional complexes. Disruption of any of these parameters due to injury or disease could negatively impact the intestinal barrier function and homeostasis resulting in increased intestine inflammation, permeability, bacterial dysbiosis, and tissue damage. MicroRNAs are small noncoding RNA sequences that are master regulators of normal cellular homeostasis. These regulatory molecules affect cellular signaling pathways and potentially serve as candidates for providing a mechanism of impaired gut barrier integrity following GI-related pathologic conditions, ethanol exposure, or trauma such as burn injury. MicroRNAs influence cellular apoptosis, proliferation, apical junction complex expression, inflammation, and the microbiome. Due to their widespread functional affiliations, altered expression of microRNAs are associated with many pathologic conditions. This review explores the role of microRNAs in regulation of intestinal barrier integrity. The studies reviewed demonstrate that microRNAs largely impact intestine barrier function and provide insight behind the observed adverse effects following ethanol and burn injury. Furthermore, these studies suggest that microRNAs are excellent candidates for therapeutic intervention or for biomarkers to manage gut barrier integrity following trauma such as burn injury and other GI-related pathologic conditions.
Collapse
Affiliation(s)
- Niya L Morris
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Current address: Department of Medicine: Pulmonary, Allergy, Critical Care and Sleep, Emory University/Atlanta VA Medical Center, Decatur, Geogia, USA
| | - Mashkoor A Choudhry
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| |
Collapse
|
9
|
Sharma T, Radosevich JA, Mandal CC. Dual Role of microRNAs in Autophagy of Colorectal Cancer. Endocr Metab Immune Disord Drug Targets 2021; 21:56-66. [DOI: 10.2174/1871530320666200519075908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/14/2020] [Accepted: 02/19/2020] [Indexed: 12/24/2022]
Abstract
Autophagy is an evolutionarily conserved pathway that eliminates unwanted proteins out of
the cell and increases cell survival. However, dysfunctional autophagy is associated with cancer progression,
cellular adaptation, cancer metastasis and makes it an attractive therapeutic target. MicroRNAs
(miRNAs) are small single-stranded non-coding RNA molecules that usually bind to 3’UTR of
mRNAs. This interaction eventually inhibits protein synthesis by repressing translation and/or by degrading
mRNAs. miRNAs play a crucial role in the regulation of autophagy and also behave as both
tumor suppressors and promoters in colorectal cancer. This paper defines an overall molecular view of
how miRNAs regulate the dual role of autophagy in colorectal cancer. It also highlights how long noncoding
RNAs modulate miRNAs expression to regulate autophagy in colorectal cancer. Thus, targeting
autophagy by miRNAs seems to be a potential therapeutic strategy for colorectal cancer.
Collapse
Affiliation(s)
- Tanu Sharma
- Department of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, 305817, India
| | - James A. Radosevich
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, University of Illinois, Chicago, 60612, Illinois, United States
| | - Chandi C. Mandal
- Department of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, 305817, India
| |
Collapse
|
10
|
Qi J, Hu Z, Liu S, Li F, Wang S, Wang W, Sheng X, Feng L. Comprehensively Analyzed Macrophage-Regulated Genes Indicate That PSMA2 Promotes Colorectal Cancer Progression. Front Oncol 2021; 10:618902. [PMID: 33537240 PMCID: PMC7849682 DOI: 10.3389/fonc.2020.618902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/30/2020] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. Here, we identified tumor-associated macrophages (TAMs) as regulators of genes in CRC. In total, the expressions of 457 genes were dysregulated after TAM coculture; specifically, 344 genes were up-regulated, and 113 genes were down-regulated. Bioinformatic analysis implied that these TAM-related genes were associated with regulation of the processes of macromolecule metabolism, apoptosis, cell death, programmed cell death, and the response to stress. To further uncover the interplay among these proteins, we constructed a PPI network; 15 key regulators were identified in CRC, including VEGFA, FN1, JUN, CDH1, MAPK8, and FOS. Among the identified genes, we focused on PSMA2 and conducted loss-of-function experiments to validate the functions of PSMA2 in CRC. To further determine the mechanism by which PSMA2 affected CRC, we conducted multiple assays in CRC cell lines and tissues. PSMA2 enhanced the proliferation, migration and invasion of CRC cells. Moreover, our data indicated that PSMA2 expression was dramatically increased in stage 1, stage 2, stage 3, and stage 4 CRC samples. Our data indicated that PSMA2 was one target of miR-132. A miR-132 mimic greatly hindered CRC cell proliferation. In addition, the luciferase assay results revealed that miR-132 directly regulated PSMA2. Moreover, our data indicated that miR-132 expression was greatly decreased in CRC samples, which was associated with longer survival times of CRC patients, implying that miR-132 was a probable biomarker for CRC. Collectively, these data indicate that PSMA2 is a promising target for the therapy of CRC.
Collapse
Affiliation(s)
- Jingbo Qi
- Endoscopy Center, Minhang Hospital, Fudan University and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhiqiu Hu
- Department of Surgical, Minhang Hospital, Fudan University, Shanghai, China
| | - Shaoqun Liu
- Department of Surgical, Minhang Hospital, Fudan University, Shanghai, China
| | - Fan Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Sheng Wang
- Endoscopy Center, Minhang Hospital, Fudan University and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wuqing Wang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Xia Sheng
- Department of Pathology, Minhang Hospital, Fudan University, Shanghai, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Suppression of HELLS by miR-451a represses mTOR pathway to hinder aggressiveness of SCLC. Genes Genomics 2021; 43:105-114. [PMID: 33460027 DOI: 10.1007/s13258-020-01028-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/11/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Uncovering molecular pathogenesis and mechanisms of small cell lung cancer (SCLC) will contribute to SCLC therapy. Multiple studies demonstrated that miR-451a acts as an anti-tumor miRNA in non-small cell lung cancer. However, the mechanism of miR-451a in SCLC was ambiguous. OBJECTIVE We aimed to explore the function of miR-451a in SCLC and decipher the underlying mechanisms. METHODS TargetScan and dual-luciferase reporter assays were used to analyze the target genes of miR-451a. Cell counting kit-8 and colony formation assays were performed to assess the roles of miR-451a on cell growth. Gene set enrichment analysis (GSEA) was utilized to enrich biological pathways. Western blot was used to measure protein expression. RESULTS MiR-451a expression was reduced dramatically in SCLC tissues and cell lines (NCI-H1688 and NCI-H446). Helicase, Lymphoid Specific (HELLS) was proved to be a target gene of miR-451a. In addition, cell proliferation assays in SCLC cells transfected with miR-451a mimic and/or HELLS revealed that miR-451a inhibited cell proliferation via targeting HELLS. Moreover, the roles of miR-451a/HELLS in expression of key proteins in mTOR and apoptosis signaling pathways suggested that miR-451a inactivated mTOR and activated apoptosis signaling pathway via directly silencing HELLS. CONCLUSIONS Our study indicated that miR-451a hinders SCLC cell proliferation in vitro through regulating mTOR and apoptosis signaling pathways via silencing HELLS, suggesting that miR-451a could be a promising tumor suppressor in SCLC. And there is a potential for miR-451a to be a drug target and biomarker for SCLC.
Collapse
|
12
|
Shiosaki J, Tiirikainen M, Peplowska K, Shaeffer D, Machida M, Sakamoto K, Takahashi M, Kojima K, Machi J, Bryant-Greenwood P, Kuwada SK. Serum micro-RNA Identifies Early Stage Colorectal Cancer in a Multi-Ethnic Population. Asian Pac J Cancer Prev 2020; 21:3019-3026. [PMID: 33112562 PMCID: PMC7798181 DOI: 10.31557/apjcp.2020.21.10.3019] [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] [Received: 05/10/2020] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Certain microRNAs (miR) have been previously described to be dysregulated in cancers and can be detected in blood samples. Studies examining the utility of miRs for colon cancer screening have primarily been performed in ethnically homogeneous groups of patients, thus the performance of miRs in multiethnic populations is unknown. METHODS Four miRs were selected that were shown to be aberrantly expressed in the blood or stool of patients with colorectal cancer (CRC) of various ethnicities. In this study, the ability of these miRs to discern early stage CRC was determined in a previously untested multiethnic population of 73 CRC cases and 18 controls. RESULTS The ratios of non-vesicular to extracellular vesicular levels of miR's -21, -29a, and -92a were statistically and quantitatively related to CRC stage compared to controls. CONCLUSION Serum levels of miR-21, miR-29a and miR-92a were able to significantly detect early stage CRC in a multiethnic and previously untested population.<br />.
Collapse
Affiliation(s)
- Jessica Shiosaki
- John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, MEB, Honolulu, Hawaii, United States
| | - Maarit Tiirikainen
- University of Hawaii Cancer Center, 01 Ilalo Street, Honolulu, Hawaii, United States
| | - Karolina Peplowska
- University of Hawaii Cancer Center, 01 Ilalo Street, Honolulu, Hawaii, United States
| | - David Shaeffer
- John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, MEB, Honolulu, Hawaii, United States
| | - Michio Machida
- Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo, Japan
| | | | | | - Kuniaki Kojima
- Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Junji Machi
- John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, MEB, Honolulu, Hawaii, United States
| | | | - Scott K Kuwada
- John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, MEB, Honolulu, Hawaii, United States.,University of Hawaii Cancer Center, 01 Ilalo Street, Honolulu, Hawaii, United States
| |
Collapse
|
13
|
Luan Y, Li X, Luan Y, Zhao R, Li Y, Liu L, Hao Y, Oleg Vladimir B, Jia L. Circulating lncRNA UCA1 Promotes Malignancy of Colorectal Cancer via the miR-143/MYO6 Axis. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 19:790-803. [PMID: 31955010 PMCID: PMC6970172 DOI: 10.1016/j.omtn.2019.12.009] [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: 06/23/2019] [Revised: 11/22/2019] [Accepted: 12/12/2019] [Indexed: 01/28/2023]
Abstract
Exosomes mediate cell-cell crosstalk in cancer progression by transferring a variety of biomolecules, including long noncoding RNAs (lncRNAs). Long non-coding RNA urothelial carcinoma-associated (UCA1) is a well-known lncRNA associated with the development and progression of various cancers, including colorectal cancer (CRC). However, the presence of UCA1 in exosomes and the roles and clinical values of exosomal UCA1 in CRC remain unknown. In this study, we systematically analyzed the expression profiles of exosomal lncRNAs in CRC patients using a high-throughput microarray assay. Then, we evaluated the UCA1 expression levels in a series of CRC tissues and the serum exosomes of CRC patients using quantitative real-time PCR. The roles of UCA1 on CRC in vitro and in vivo were investigated by MTT, colony formation, Transwell, quantitative real-time PCR, flow cytometry, and western blotting. The miRNA binding sites of UCA1 were predicted using the miRcode online database, and miR-143 was validated to target UCA1 by dual-luciferase activity assay and AGO2 RNA immunoprecipitation. Finally, the role of exosome-mediated UCA1 was further investigated by co-culturing with CRC cells. This study showed that UCA1 was upregulated in CRC tissues and functioned as an oncogene in CRC. Loss-of-function investigations showed that inhibition of UCA1 suppressed CRC cell proliferation and metastasis in vivo and in vitro. Mechanistically, UCA1 was identified as a miR-143 sponge. We also found that MYO6 was a direct target of miR-1205, which functioned as an oncogene in CRC. Moreover, UCA was also upregulated in the serum exosomes of CRC patients and could transfer UCA1 to CRC cells to increase their abilities of cell proliferation and migration. In conclusion, these data suggest that UCA1 could be an oncogene for CRC and may serve as a candidate target for new therapies in human CRC.
Collapse
Affiliation(s)
- Yunpeng Luan
- Key Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, Southwest Forestry University, Kunming 650224, China.
| | - Xiang Li
- Department of Gastrointestinal, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Nanjing 650021, China; Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yunqi Luan
- Department of Physical Education, Yunnan Normal University, Kunming 650500, China
| | - Rong Zhao
- Department of Gastrointestinal, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Nanjing 650021, China
| | - Yanmei Li
- Department of Life Technology Teaching and Research, College of Life Sciences, Southwest Forestry University, Kunming 650224, China
| | - Lili Liu
- Department of Life Technology Teaching and Research, College of Life Sciences, Southwest Forestry University, Kunming 650224, China
| | - Yizhuo Hao
- Department of Gastrointestinal Surgery, Sixth People's Hospital of Dalian City, Dalian 116031, China
| | - Burakovaov Oleg Vladimir
- Department of Bioengineering, College of Life Science, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Lu Jia
- Department of Life Technology Teaching and Research, College of Life Sciences, Southwest Forestry University, Kunming 650224, China
| |
Collapse
|
14
|
Li X, Cao Q, Wang Y, Wang Y. Retracted Article: LncRNA OIP5-AS1 contributes to ox-LDL-induced inflammation and oxidative stress through regulating the miR-128-3p/CDKN2A axis in macrophages. RSC Adv 2019; 9:41709-41719. [PMID: 35541591 PMCID: PMC9076472 DOI: 10.1039/c9ra08322g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/02/2019] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNA OIP5-AS1 (lncRNA OIP5-AS1) and microRNA-128-3p (miRNA-128-3p) have been reported to play significant roles in human diseases. However, their role in atherosclerosis (AS) has been less studied. The aim of this research was to reveal the roles and functional mechanisms of OIP5-AS1 and miRNA-128-3p in AS development. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot assays were performed to detect gene expression. Cell proliferation and apoptosis were assessed using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry analysis, respectively. In addition, ELISA was employed to determine the levels of interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α. Oxidative stress was examined using a relevant kit. Furthermore, the interaction between miR-128-3p and OIP5-AS1 or cyclin-dependent kinase inhibitor 2A (CDKN2A) was predicted using StarBase, and then confirmed using the dual-luciferase reporter assay or the RNA immunoprecipitation (RIP) assay. We found that OIP5-AS1 and CDKN2A levels were upregulated and the miR-128-3p level was downregulated in oxidized low-density lipoprotein (ox-LDL)-induced THP1 cells. OIP5-AS1 knockdown weakened the regulatory effect of ox-LDL on cell progression. Interestingly, OIP5-AS1 directly interacted with miR-128-3p and miR-128-3p-targeted CDKN2A. Furthermore, OIP5-AS1 regulated ox-LDL-induced cell progression through modulating miR-128-3p expression, and miR-128-3p exerted its influence by modulating the CDKN2A level. Finally, we confirmed that OIP5-AS1 suppressed miR-128-3p expression to increase the level of CDKN2A. In conclusion, our findings demonstrate that OIP5-AS1 knockdown repressed the effect of ox-LDL on cell progression through regulating the miR-128-3p/CDKN2A axis, providing a potential target for the treatment of AS.
Collapse
Affiliation(s)
- Xiaojuan Li
- Department of Central Sterile Supply, The First Affiliated Hospital of Henan University of Science and Technology No. 24, Jinghua Road Luoyang 471003 China +86-0379-64830544
| | - Quansheng Cao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Henan University of Science and Technology Luoyang China
| | - Yanyu Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Henan University of Science and Technology Luoyang China
| | - Yongsheng Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Henan University of Science and Technology Luoyang China
| |
Collapse
|
15
|
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]
|
16
|
Han SH, Mo JS, Park WC, Chae SC. Reduced microRNA 375 in colorectal cancer upregulates metadherin-mediated signaling. World J Gastroenterol 2019; 25:6495-6507. [PMID: 31802830 PMCID: PMC6886017 DOI: 10.3748/wjg.v25.i44.6495] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/20/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The human microRNA 375 (MIR375) is significantly downregulated in human colorectal cancer (CRC) and we have previously shown that MIR375 is a CRC-associated miRNA. The metadherin (MTDH) is a candidate target gene of MIR375. AIM To investigate the interaction and function between MIR375 and MTDH in human CRC. METHODS A luciferase reporter system was used to confirm the effect of MIR375 on MTDH expression. The expression levels of MIR375 and the target genes were evaluated by quantitative RT-PCR (qRT-PCR), western blotting, or immunohistochemistry. RESULTS MTDH expression was found to be upregulated in human CRC tissues compared to that in healthy controls. We show that MIR375 regulates the expression of many genes involved in the MTDH-mediated signal transduction pathways [BRAF-MAPK and phosphatidylinositol-4,5-biphosphate-3-kinase catalytic subunit alpha (PIK3CA)-AKT] in CRC cells. Upregulated MTDH expression levels were found to inhibit NF-κB inhibitor alpha, which further upregulated NFKB1 and RELA expression in CRC cells. CONCLUSION Our findings suggest that suppressing MIR375 expression in CRC regulates cell proliferation and angiogenesis by increasing MTDH expression. Thus, MIR375 may be of therapeutic value in treating human CRC.
Collapse
Affiliation(s)
- Seol-Hee Han
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk 54538, South Korea
| | - Ji-Su Mo
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk 54538, South Korea
- Digestive Disease Research Institute, Wonkwang University, Iksan, Chonbuk 54538, South Korea
| | - Won-Cheol Park
- Digestive Disease Research Institute, Wonkwang University, Iksan, Chonbuk 54538, South Korea
| | - Soo-Cheon Chae
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk 54538, South Korea
- Digestive Disease Research Institute, Wonkwang University, Iksan, Chonbuk 54538, South Korea
| |
Collapse
|
17
|
Yu Y, Gao F, He Q, Li G, Ding G. lncRNA UCA1 Functions as a ceRNA to Promote Prostate Cancer Progression via Sponging miR143. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 19:751-758. [PMID: 31954329 PMCID: PMC6962633 DOI: 10.1016/j.omtn.2019.11.021] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/24/2019] [Accepted: 11/01/2019] [Indexed: 01/08/2023]
Abstract
UCA1 (urothelial carcinoma associated 1) is a long non-coding RNA (lncRNA) that was found overexpressed in various human cancers including prostate cancer (PCa). However, the aspect of UCA1-miRNA-mRNA interaction in PCa remains unclear. In this study, we confirmed the role of UCA1 in PCa and found that UCA1 downregulation inhibited cell proliferation of PCa cells. Then we demonstrated that repressed UCA1 promoted the microRNA-143 (miR-143) expression and miR-143 could bind to the predicted binding site of UCA1. We then proved the anti-tumor role of miR-143 in PCa. Furthermore, we found that miR-143 displays its role in PCa via modulating the MYO6 expression. In summary, our study demonstrated that UCA1 exerts oncogenes activity in PCa, acting mechanistically by upregulating MYO6 expression through “sponging” miR-143.
Collapse
Affiliation(s)
- Yanlan Yu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Fengbin Gao
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Qian He
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Guoqing Ding
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| |
Collapse
|
18
|
Rodrigues Lopes I, Silva RJ, Caramelo I, Eulalio A, Mano M. Shedding light on microRNA function via microscopy-based screening. Methods 2019; 152:55-64. [DOI: 10.1016/j.ymeth.2018.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 09/13/2018] [Accepted: 09/28/2018] [Indexed: 12/24/2022] Open
|
19
|
Zheng Y, Tan K, Huang H. Long noncoding RNA HAGLROS regulates apoptosis and autophagy in colorectal cancer cells via sponging miR-100 to target ATG5 expression. J Cell Biochem 2018; 120:3922-3933. [PMID: 30430634 DOI: 10.1002/jcb.27676] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022]
Abstract
The aim of this study was to explore the relationship between the expression of HOXD antisense growth-associated long noncoding RNA (HAGLROS) and prognosis of patients with colorectal cancer (CRC), as well as the roles and regulatory mechanism of HAGLROS in CRC development. The HAGLROS expression in CRC tissues and cells was detected. The correlation between HAGLROS expression and survival time of CRC patients was investigated. Moreover, HAGLROS was overexpressed and suppressed in HCT-116 cells, followed by detection of cell viability, apoptosis, and the expression of apoptosis-related proteins and autophagy markers. Furthermore, the association between HAGLROS and miR-100 and the potential targets of miR-100 were investigated. Besides, the regulatory relationship between HAGLROS and PI3K/AKT/mTOR pathway was elucidated. The results showed that HAGLROS was highly expressed in CRC tissues and cells. Highly expression of HAGLROS correlated with a shorter survival time of CRC patients. Moreover, knockdown of HAGLROS in HCT-116 cells induced apoptosis by increasing the expression of Bax/Bcl-2 ratio, cleaved-caspase-3, and cleaved-caspase-9, and inhibited autophagy by decreasing the expression of LC3II/LC3I and Beclin-1 and increasing P62 expression. Furthermore, HAGLROS negatively regulated the expression of miR-100, and HAGLROS controlled HCT-116 cell apoptosis and autophagy through negatively regulation of miR-100. Autophagy related 5 (ATG5) was verified as a functional target of miR-100 and miR-100 regulated HCT-116 cell apoptosis and autophagy through targeting ATG5. Besides, HAGLROS overexpression activated phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway. In conclusion, a highly expression of HAGLROS correlated with shorter survival time of CRC patients. Downregulation of HAGLROS may induce apoptosis and inhibit autophagy in CRC cells by regulation of miR-100/ATG5 axis and PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Yansheng Zheng
- Department of Gastrointestinal Surgery, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, China
| | - Kanglian Tan
- Department of Gastrointestinal Surgery, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, China
| | - Haipeng Huang
- Department of Gastrointestinal Surgery, Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, China
| |
Collapse
|
20
|
miR-375 is involved in Hippo pathway by targeting YAP1/TEAD4-CTGF axis in gastric carcinogenesis. Cell Death Dis 2018; 9:92. [PMID: 29367737 PMCID: PMC5833783 DOI: 10.1038/s41419-017-0134-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 02/08/2023]
Abstract
miR-375 is a tumor-suppressive microRNA (miRNA) in gastric cancer (GC). However, its molecular mechanism remains unclear. The aim of this study is to comprehensively investigate how miR-375 is involved in Hippo pathway by targeting multiple oncogenes. miR-375 expression in gastric cancer cell lines and primary GC was investigated by qRT-PCR. The regulation of YAP1, TEAD4, and CTGF expression by miR-375 was evaluated by qRT-PCR, western blot, and luciferase reporter assays, respectively. The functional roles of the related genes were examined by siRNA-mediated knockdown or ectopic expression assays. The clinical significance and expression correlation analysis of miR-375, YAP1, and CTGF were performed in primary GCs. TCGA cohort was also used to analyze the expression correlation of YAP1, TEAD4, CTGF, and miR-375 in primary GCs. miR-375 was down-regulated in GC due to promoter methylation and histone deacetylation. miR-375 downregulation was associated with unfavorable outcome and lymph node metastasis. Ectopic expression of miR-375 inhibited tumor growth in vitro and in vivo. Three components of Hippo pathway, YAP1, TEAD4 and CTGF, were revealed to be direct targets of miR-375. The expression of three genes showed a negative correlation with miR-375 expression and YAP1 re-expression partly abolished the tumor-suppressive effect of miR-375. Furthermore, CTGF was confirmed to be the key downstream of Hippo-YAP1 cascade and its knockdown phenocopied siYAP1 or miR-375 overexpression. YAP1 nuclear accumulation was positively correlated with CTGF cytoplasmic expression in primary GC tissues. Verteporfin exerted an anti-oncogenic effect in GC cell lines by quenching CTGF expression through YAP1 degradation. In short, miR-375 was involved in the Hippo pathway by targeting YAP1-TEAD4-CTGF axis and enriched our knowledge on the miRNA dysregulation in gastric tumorigenesis.
Collapse
|
21
|
Shaker OG, Mohammed SR, Mohammed AM, Mahmoud Z. Impact of microRNA-375 and its target gene SMAD-7 polymorphism on susceptibility of colorectal cancer. J Clin Lab Anal 2018; 32:e22215. [PMID: 28374902 PMCID: PMC6817095 DOI: 10.1002/jcla.22215] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/27/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) has a high morbidity and mortality. Many studies reported that mir-375 is frequently down-regulated in many cancers including esophageal cancer, hepatocellular carcinoma, breast cancer and leukemias. AIM Our aim was to study the expression of microRNA-375 and its target gene SMAD-7 polymorphisms (rs4939827) in CRC patients in comparison to control subjects and to correlate these results with clinical data of patients to elucidate their role in pathogenesis and early diagnosis of CRC. MATERIAL AND METHODS The present study was conducted on 122 subjects divided into 86 patients with CRC and 36 age- and sex-matched controls. The followings were done to all subjects: full history taking, full clinical examination, complete blood picture, serum (ALT, AST), serum albumin, CEA, TLC, PLT, and creatinine. Gene expression of miRNA-375 from serum was done by real-time PCR. Gene polymorphism SNPs of SMAD7 (rs4939827) was also done in DNA extracted from blood by real-time PCR. RESULTS As regards the polymorphism of SMAD7, we found that CC (wild) genotype has high percentage in controls compared to CRC cases (36.1% vs 15.1%). Meanwhile, the mutant and heterozygotes genotypes showed high percentage among cases compared to controls (33.7%, and 51.2% respectively) vs (22.2%, and 41.7% respectively) with no significant statistical analysis. There was a statistically significant high T-allelic frequency among cases and C-allelic frequency among controls. There was a statistically significant association between fold change in micro RNA (-375) and the susceptibility to CRC as there is down-regulation of the microRNA-375 in CRC group with fold change in 0.42±0.27. CONCLUSION Micro RNA-375 and rs4939827 SNP in SMAD7 could be considered as potential markers for detecting and early diagnosing CRC patients.
Collapse
Affiliation(s)
- Olfat Gamil Shaker
- Biochemistry and Molecular BiologyFaculty of MedicineCairo UniversityCairoEgypt
| | - Shereen Rashad Mohammed
- Departments of Medical Biochemistry and Molecular BiologyFaculty of MedicineFayoum UniversityAl FayoumEgypt
| | - Asmaa Mohammed Mohammed
- Departments of Medical Biochemistry and Molecular BiologyFaculty of MedicineFayoum UniversityAl FayoumEgypt
| | - Zeinab Mahmoud
- Departments of Tropical MedicineFaculty of MedicineCairo UniversityCairoEgypt
| |
Collapse
|
22
|
Librizzi M, Caradonna F, Cruciata I, Dębski J, Sansook S, Dadlez M, Spencer J, Luparello C. Molecular Signatures Associated with Treatment of Triple-Negative MDA-MB231 Breast Cancer Cells with Histone Deacetylase Inhibitors JAHA and SAHA. Chem Res Toxicol 2017; 30:2187-2196. [PMID: 29129070 DOI: 10.1021/acs.chemrestox.7b00269] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Jay Amin hydroxamic acid (JAHA; N8-ferrocenylN1-hydroxy-octanediamide) is a ferrocene-containing analogue of the histone deacetylase inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA). JAHA's cytotoxic activity on MDA-MB231 triple negative breast cancer (TNBC) cells at 72 h has been previously demonstrated with an IC50 of 8.45 μM. JAHA's lethal effect was found linked to perturbations of cell cycle, mitochondrial activity, signal transduction, and autophagy mechanisms. To glean novel insights on how MDA-MB231 breast cancer cells respond to the cytotoxic effect induced by JAHA, and to compare the biological effect with the related compound SAHA, we have employed a combination of differential display-PCR, proteome analysis, and COMET assay techniques and shown some differences in the molecular signature profiles induced by exposure to either HDACis. In particular, in contrast to the more numerous and diversified changes induced by SAHA, JAHA has shown a more selective impact on expression of molecular signatures involved in antioxidant activity and DNA repair. Besides expanding the biological knowledge of the effect exerted by the modifications in compound structures on cell phenotype, the molecular elements put in evidence in our study may provide promising targets for therapeutic interventions on TNBCs.
Collapse
Affiliation(s)
- Mariangela Librizzi
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo , Viale delle Scienze, 90128 Palermo, Italy
| | - Fabio Caradonna
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo , Viale delle Scienze, 90128 Palermo, Italy
| | - Ilenia Cruciata
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo , Viale delle Scienze, 90128 Palermo, Italy
| | - Janusz Dębski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Supojjanee Sansook
- Department of Chemistry, School of Life Sciences, University of Sussex , Falmer, Brighton BN1 9QJ, United Kingdom
| | - Michał Dadlez
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Pawinskiego 5a, 02-106 Warsaw, Poland
| | - John Spencer
- Department of Chemistry, School of Life Sciences, University of Sussex , Falmer, Brighton BN1 9QJ, United Kingdom
| | - Claudio Luparello
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo , Viale delle Scienze, 90128 Palermo, Italy
| |
Collapse
|
23
|
Alam KJ, Mo JS, Han SH, Park WC, Kim HS, Yun KJ, Chae SC. MicroRNA 375 regulates proliferation and migration of colon cancer cells by suppressing the CTGF-EGFR signaling pathway. Int J Cancer 2017; 141:1614-1629. [PMID: 28670764 DOI: 10.1002/ijc.30861] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/04/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022]
Abstract
MicroRNA 375 (MIR375) is significantly down regulated in human colorectal cancer (CRC) tissues; we have previously identified MIR375 as a colon cancer associated microRNA (miRNA). We identified putative MIR375 target genes by comparing the mRNA microarray analysis data of MIR375-overexpressing cells with the candidate MIR375 target genes predicted by public bioinformatic tools. We investigated that the connective tissue growth factor (CTGF) is a direct target gene of MIR375. Expression of CTGF, a ligand of epidermal growth factor receptor (EGFR), was markedly enhanced in human CRC tissues in comparison with the corresponding normal colon tissues. We demonstrated that the expression levels of molecules in EGFR signaling pathways were regulated by MIR375 in colorectal cells. Using immunohistochemistry and the xenograft of MIR375-overexpressing colorectal cells in mice, we showed that MIR375 regulates cell growth and proliferation, angiogenesis, cell migration, cell cycle arrest, apoptosis, and necrosis in colon cells. Furthermore, results of MIR375 overexpression and cetuximab treatment indicated that the apoptosis and necrosis in colon cells were synergistically enhanced. Our results suggest that the down-regulation of MIR375 modulates EGFR signaling pathways in human colorectal cells and tissues by increasing CTGF expression; therefore, MIR375 may have a therapeutic value in relation to human CRC.
Collapse
Affiliation(s)
- Khondoker Jahengir Alam
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
| | - Ji-Su Mo
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
- Digestive Disease Research Institute, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
| | - Seol-Hee Han
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
| | - Won-Cheol Park
- Digestive Disease Research Institute, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
| | - Hun-Soo Kim
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
| | - Ki-Jung Yun
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
- Digestive Disease Research Institute, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
| | - Soo-Cheon Chae
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
- Digestive Disease Research Institute, School of Medicine, Wonkwang University, Iksan, Chonbuk, Republic of Korea
| |
Collapse
|
24
|
Wan Y, Shen A, Qi F, Chu J, Cai Q, Sferra TJ, Peng J, Chen Y. Pien Tze Huang inhibits the proliferation of colorectal cancer cells by increasing the expression of miR-34c-5p. Exp Ther Med 2017; 14:3901-3907. [PMID: 29042999 DOI: 10.3892/etm.2017.4972] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 05/05/2016] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small, short endogenous non-coding RNA that act as oncogenes or tumor suppressors, and serve an important role in various human malignant cancers, including colorectal cancer (CRC). Evidence has indicated that miRNAs regulate the expression of various genes associated with human cancer, in particular the miR-34 family. A well-known traditional Chinese formula, Pien Tze Huang (PZH), has a significant clinical effect on CRC. Previous studies have demonstrated that PZH inhibits CRC growth in vitro and in vivo via multiple mechanisms, including the induction of apoptosis, inhibition of cell proliferation and tumor angiogenesis. To further elucidate the molecular mechanisms underlying the antitumor activity of PZH, in the present study its effects on cell proliferation and miRNA expression in human colon carcinoma (HCT)-8 cell lines was examined. It was observed that treatment with PZH inhibited cell viability and upregulated the expression of miR-34c-5p in HCT-8 cells. In addition, transfection with an miR-34c-5p mimic and treatment with PZH inhibited cell survival and arrested the cell cycle between the G0/G1 and S phase in HCT-8 cells. Furthermore, PZH treatment and transfection with miR-34c-5p downregulated the expression of cyclin-dependent kinase 4 and cMyc (a promoter of cell proliferation), and increased the expression of p53, which is a promoter of apoptosis. These results suggest that PZH may suppress proliferation in CRC cells by upregulating the expression of miR-34c-5p, which provides a novel perspective for understanding the mode of action of PZH.
Collapse
Affiliation(s)
- Yun Wan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Fei Qi
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jianfeng Chu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Qiaoyan Cai
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Thomas Joseph Sferra
- Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Postdoctoral Workstation, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian 363000, P.R. China
| | - Youqin Chen
- Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
25
|
Elshafei A, Shaker O, Abd El-Motaal O, Salman T. The expression profiling of serum miR-92a, miR-375, and miR-760 in colorectal cancer: An Egyptian study. Tumour Biol 2017; 39:1010428317705765. [PMID: 28618945 DOI: 10.1177/1010428317705765] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dysregulation in microRNA expression is a common feature in colorectal cancer. Due to the inconsistent results regarding serum miR-92a expression pattern and the insufficient studies on serum miR-375 and miR-760, we aimed in this study to investigate their expression profile and diagnostic and prognostic power in Egyptian colorectal cancer patients. The expression profile of miR-92a, miR-375, and miR-760 was determined in the sera of 64 colorectal cancer patients using quantitative real-time reverse transcription polymerase chain reaction in comparison to 27 healthy control subjects. The expression fold change of the studied microRNAs was correlated with patients' clinicopathological features. Receiver operating characteristic curve analysis was done to determine the role of these microRNAs in colorectal cancer diagnosis and follow-up according to the yielded area under the curve. The expression pattern of miR-92a was significantly upregulated (3.38 ± 2.52, p < 0.0001), while both of miR-375 and 760 were significantly downregulated (-1.250 ± 1.80, p< 0.0001; -1.710 ± 1.88, p < 0.0001, respectively) in colorectal cancer than the control. MiR-92a was positively correlated ( r = 0.671, p = 0.0001), while miR-375 and miR-760 were inversely correlated ( r = -0.414, p = 0.001; r = -0.644, p = 0.0001) with advanced colorectal cancer stages. Receiver operating characteristic curve analysis disclosed the highest diagnostic potential for miR-760 to discriminate colorectal cancer patients and early-stage colorectal cancer from the control (area under the curve = 0.922 and 0.875, respectively), while the highest prognostic potential for discrimination between colorectal cancer stages was for miR-92a. In conclusion, serum level of miR-92a, miR-375, and miR-760 may serve as biomarkers of colorectal cancer in Egyptian patients with high diagnostic power for miR-760 and high prognostic power for miR-92a.
Collapse
Affiliation(s)
- Ahmed Elshafei
- 1 Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Olfat Shaker
- 2 Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ossama Abd El-Motaal
- 1 Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Tarek Salman
- 1 Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
26
|
Wei W, Liu Y, Lu Y, Yang B, Tang L. LncRNA XIST Promotes Pancreatic Cancer Proliferation Through miR-133a/EGFR. J Cell Biochem 2017; 118:3349-3358. [PMID: 28295543 DOI: 10.1002/jcb.25988] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 03/09/2017] [Indexed: 12/17/2022]
Abstract
According to recent studies, long non-coding RNA X-inactive specific transcript (XIST) is involved in the development and progression of many malignant tumors including pancreatic cancer. We validated the detailed role of XIST in human pancreatic cancer (PC) cell lines and PC tissues so as to determine its exact function and the mechanism by which it affected PC proliferation. In our research, lncRNA-XIST was specifically upregulated in PC tissues and cell lines, and high XIST expression in PC was related to poorer prognosis (larger tumor size, perineural invasion, lymph node micrometastases, and shorter overall survival). XIST augmented PC cell proliferation. Recently, the interaction between lncRNA and miRNA has been frequently reported to play major role in several biological processes. In the present study, XIST and miR-133a reciprocally inhibited each other in PC cells. Exogenous miR-133a expression significantly inhibited PC cell proliferation. Moreover, as exhibited by luciferase reporter gene assays, miR-133a bound to XIST and the 3'UTR of EGFR by direct targeting. In PC tissues, miR-133a expression was down-regulated and EGFR expression was up-regulated; miR-133a was inversely correlated with EGFR and XIST, respectively; XIST was positively correlated with EGFR. Taken together, these findings will shed light on the role and mechanism of XIST/miR-133a/EGFR in regulating PC cells proliferation. XIST may serve as a potential therapeutic target in PC in the future. J. Cell. Biochem. 118: 3349-3358, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wei Wei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu Liu
- Department of Pathology, Hunan Provincial People's Hospital, Changsha, 410005, China
| | - Yebin Lu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Bo Yang
- Department of General Surgery, The Fourth Hospital of Changde City, Changde, 415000, China
| | - Ling Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| |
Collapse
|
27
|
Umegawachi T, Yoshida H, Koshida H, Yamada M, Ohkawa Y, Sato T, Suyama M, Krause HM, Yamaguchi M. Control of tissue size and development by a regulatory element in the yorkie 3'UTR. Am J Cancer Res 2017; 7:673-687. [PMID: 28401020 PMCID: PMC5385651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 10/31/2016] [Indexed: 06/07/2023] Open
Abstract
Regulation of the Hippo pathway via phosphorylation of Yorkie (Yki), the Drosophila homolog of human Yes-associated protein 1, is conserved from Drosophila to humans. Overexpression of a non-phosphorylatable form of Yki induces severe overgrowth in adult fly eyes. Here, we show that yki mRNA associates with microsomal fractions and forms foci that partially colocalize to processing bodies in the vicinity of endoplasmic reticulum. This localization is dependent on a stem-loop (SL) structure in the 3' untranslated region of yki. Surprisingly, expression of SL deleted yki in eye imaginal discs also results in severe overgrowth phenotypes. When the structure of the SL is disrupted, Yki protein levels increase without a significant effect on RNA levels. When the SL is completely removed, protein levels drastically increase, but in this case, due to increased RNA stability. In the latter case, we show that the increased RNA accumulation is due to removal of a putative miR-8 seed sequence in the SL. These data demonstrate the function of two novel regulatory mechanisms, both controlled by the yki SL element, that are essential for proper Hippo pathway mediated growth regulation.
Collapse
Affiliation(s)
- Takanari Umegawachi
- Department of Applied Biology, Kyoto Institute of TechnologyMatsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Hideki Yoshida
- Department of Applied Biology, Kyoto Institute of TechnologyMatsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
- The Center for Advanced Insect Research Promotion, Kyoto Institute of TechnologyMatsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Hiromu Koshida
- Department of Applied Biology, Kyoto Institute of TechnologyMatsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Momoko Yamada
- Department of Applied Biology, Kyoto Institute of TechnologyMatsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Yasuyuki Ohkawa
- Medical Institute of Bioregulation, Kyushu UniversityFukuoka 812-8582, Japan
| | - Tetsuya Sato
- Medical Institute of Bioregulation, Kyushu UniversityFukuoka 812-8582, Japan
| | - Mikita Suyama
- Medical Institute of Bioregulation, Kyushu UniversityFukuoka 812-8582, Japan
| | - Henry M Krause
- Banting and Best Department of Medical Research, University of TorontoToronto, Ontario M5G 1L6, Canada
- Department of Molecular Genetics, University of TorontoToronto, Ontario M5G 1L6, Canada
- Donnelly Centre, University of TorontoToronto, Ontario M5G 1L6, Canada
| | - Masamitsu Yamaguchi
- Department of Applied Biology, Kyoto Institute of TechnologyMatsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
- The Center for Advanced Insect Research Promotion, Kyoto Institute of TechnologyMatsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| |
Collapse
|
28
|
Jeppsson S, Srinivasan S, Chandrasekharan B. Neuropeptide Y (NPY) promotes inflammation-induced tumorigenesis by enhancing epithelial cell proliferation. Am J Physiol Gastrointest Liver Physiol 2017; 312:G103-G111. [PMID: 27856419 PMCID: PMC5338605 DOI: 10.1152/ajpgi.00410.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 11/10/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
Abstract
UNLABELLED We have demonstrated that neuropeptide Y (NPY), abundantly produced by enteric neurons, is an important regulator of intestinal inflammation. However, the role of NPY in the progression of chronic inflammation to tumorigenesis is unknown. We investigated whether NPY could modulate epithelial cell proliferation and apoptosis, and thus regulate tumorigenesis. Repeated cycles of dextran sodium sulfate (DSS) were used to model inflammation-induced tumorigenesis in wild-type (WT) and NPY knockout (NPY-/-) mice. Intestinal epithelial cell lines (T84) were used to assess the effects of NPY (0.1 µM) on epithelial proliferation and apoptosis in vitro. DSS-WT mice exhibited enhanced intestinal inflammation, polyp size, and polyp number (7.5 ± 0.8) compared with DSS-NPY-/- mice (4 ± 0.5, P < 0.01). Accordingly, DSS-WT mice also showed increased colonic epithelial proliferation (PCNA, Ki67) and reduced apoptosis (TUNEL) compared with DSS-NPY-/- mice. The apoptosis regulating microRNA, miR-375, was significantly downregulated in the colon of DSS-WT (2-fold, P < 0.01) compared with DSS-NPY-/--mice. In vitro studies indicated that NPY promotes cell proliferation (increase in PCNA and β-catenin, P < 0.05) via phosphatidyl-inositol-3-kinase (PI3-K)-β-catenin signaling, suppressed miR-375 expression, and reduced apoptosis (increase in phospho-Bad). NPY-treated cells also displayed increased c-Myc and cyclin D1, and reduction in p21 (P < 0.05). Addition of miR-375 inhibitor to cells already treated with NPY did not further enhance the effects induced by NPY alone. Our findings demonstrate a novel regulation of inflammation-induced tumorigenesis by NPY-epithelial cross talk as mediated by activation of PI3-K signaling and downregulation of miR-375. NEW & NOTEWORTHY Our work exemplifies a novel role of neuropeptide Y (NPY) in regulating inflammation-induced tumorigenesis via two modalities: first by enhanced proliferation (PI3-K/pAkt), and second by downregulation of microRNA-375 (miR-375)-dependent apoptosis in intestinal epithelial cells. Our data establish the existence of a microRNA-mediated cross talk between enteric neurons producing NPY and intestinal epithelial cells, and the potential of neuropeptide-regulated miRNAs as potential therapeutic molecules for the management of inflammation-associated tumors in the gut.
Collapse
Affiliation(s)
- Sabrina Jeppsson
- 1Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia;
| | - Shanthi Srinivasan
- 1Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia; ,2Veterans Affairs Medical Center, Decatur, Atlanta, Georgia; and
| | - Bindu Chandrasekharan
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia; .,Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
29
|
Zheng J, Lin Z, Zhang L, Chen H. MicroRNA-455-3p Inhibits Tumor Cell Proliferation and Induces Apoptosis in HCT116 Human Colon Cancer Cells. Med Sci Monit 2016; 22:4431-4437. [PMID: 27861461 PMCID: PMC5117242 DOI: 10.12659/msm.898452] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND MicroRNAs have been reported to play significant roles in pathogenesis of colorectal cancer (CRC). In the present study, we aimed to investigate the functional role of microRNA-455-3p (miR-455-3p) in CRC, as well as its underlying mechanisms. MATERIAL AND METHODS Human colon cancer cell line HCT116 cells were transfected with miR-455-3p mimics, inhibitors, or controls. After transfection, the effects of miR-455-3p mimics or inhibitors on cell proliferation were analyzed by 3-(4, 5-dimethyl-2- thiazolyl)-2, 5-diphenyl -2-H-tetrazolium bromide (MTT) assay and BrdU assay, and the effects of miR-455-3p mimics or inhibitors on cell apoptosis were determined. In addition, the underlying mechanisms of cell proliferation and apoptosis were explored by assessing the protein levels of cell cycle regulators and apoptosis-related protein. RESULTS The results showed that overexpression of miR-455-3p significantly inhibited the cell proliferation (P<0.05 or <0.01) in HCT116 cells compared with the control group, but significantly increased the apoptosis (P<0.01). On the contrary, suppression of miR-455-3p significantly increased the cell proliferation but decreased the apoptosis. Moreover, we found that overexpression of miR-455-3p significantly elevated the protein levels of p27 kinase inhibition protein (KIP) 1, Bax, pro-caspase-3, and active caspase-3, and markedly downregulated the levels of B-cell lymphoma-2 (Bcl-2). Contrary results were found by suppression of miR-455-3p. However, there were no significant differences in p21 expression. CONCLUSIONS MiRNA-455-3p functions as an anti-oncogene in HCT116 cells by inhibiting cell proliferation and inducing of apoptosis.
Collapse
Affiliation(s)
- Jiantao Zheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China (mainland)
| | - Zhenlv Lin
- Department of Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China (mainland)
| | - Lin Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China (mainland)
| | - Hui Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China (mainland)
| |
Collapse
|
30
|
Ostenfeld MS, Jensen SG, Jeppesen DK, Christensen LL, Thorsen SB, Stenvang J, Hvam ML, Thomsen A, Mouritzen P, Rasmussen MH, Nielsen HJ, Ørntoft TF, Andersen CL. miRNA profiling of circulating EpCAM(+) extracellular vesicles: promising biomarkers of colorectal cancer. J Extracell Vesicles 2016; 5:31488. [PMID: 27576678 PMCID: PMC5005366 DOI: 10.3402/jev.v5.31488] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/14/2016] [Accepted: 07/14/2016] [Indexed: 01/06/2023] Open
Abstract
Cancer cells secrete small membranous extracellular vesicles (EVs) into their microenvironment and circulation. These contain biomolecules, including proteins and microRNAs (miRNAs). Both circulating EVs and miRNAs have received much attention as biomarker candidates for non-invasive diagnostics. Here we describe a sensitive analytical method for isolation and subsequent miRNA profiling of epithelial-derived EVs from blood samples of patients with colorectal cancer (CRC). The epithelial-derived EVs were isolated by immunoaffinity-capture using the epithelial cell adhesion molecule (EpCAM) as marker. This approach mitigates some of the specificity issues observed in earlier studies of circulating miRNAs, in particular the negative influence of miRNAs released by erythrocytes, platelets and non-epithelial cells. By applying this method to 2 small-scale patient cohorts, we showed that blood plasma isolated from CRC patients prior to surgery contained elevated levels of 13 EpCAM+-EV miRNAs compared with healthy individuals. Upon surgical tumour removal, the plasma levels of 8 of these were reduced (miR-16-5p, miR-23a-3p, miR-23b-3p, miR-27a-3p, miR-27b-3p, miR-30b-5p, miR-30c-5p and miR-222-3p). These findings indicate that the miRNAs are of tumour origin and may have potential as non-invasive biomarkers for detection of CRC. This work describes a non-invasive blood-based method for sensitive detection of cancer with potential for clinical use in relation to diagnosis and screening. We used the method to study CRC; however, it is not restricted to this disease. It may in principle be used to study any cancer that release epithelial-derived EVs into circulation.
Collapse
Affiliation(s)
| | - Steffen Grann Jensen
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Stine Buch Thorsen
- Department of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jan Stenvang
- Department of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark
| | - Michael Lykke Hvam
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | | | | | - Hans Jørgen Nielsen
- Department of Surgical Gastroenterology 360, Hvidovre Hospital, Hvidovre, Denmark
| | - Torben Falck Ørntoft
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
31
|
Xu L, Wen T, Liu Z, Xu F, Yang L, Liu J, Feng G, An G. MicroRNA-375 suppresses human colorectal cancer metastasis by targeting Frizzled 8. Oncotarget 2016; 7:40644-40656. [PMID: 27276676 PMCID: PMC5130033 DOI: 10.18632/oncotarget.9811] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 05/17/2016] [Indexed: 01/01/2023] Open
Abstract
microRNAs are aberrantly expressed during the development and progression of a variety of human cancers, including colorectal cancer (CRC). Of these microRNAs, microRNA-375 (miR-375) was previously observed to be downregulated in human colorectal cancer(CRC) plasma and tissues, but its functions are largely unknown. Here, we investigated the impact of miR-375 on CRC metastasis. Specifically, miR-375 expression was significantly decreased in human CRC tissues compared with their matched noncancerous tissues (NCTs), and low levels of miR-375 predicted tumor metastatic potential. The up-regulation of miR-375 suppressed colorectal cancer cell migration and invasion in vitro and reduced tumor metastases in murine models established by both orthotopic implantation and spleen injection. Furthermore, we identified Frizzled 8 (FZD8) as a direct target of miR-375 in CRC, and miR-375 negatively regulated Wnt/β-catenin signaling by suppressing FZD8. More importantly, FZD8 expression inversely correlated with overall survival in human CRC patients and is a likely independent predictor of survival. Therefore, we concluded that miR-375 functions as a tumor-suppressive microRNA by directly acting upon FZD8, which may serve as a new therapeutic target to inhibit tumor metastasis in CRC.
Collapse
Affiliation(s)
- Lingling Xu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Tao Wen
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhe Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Feng Xu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Lei Yang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jian Liu
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Guosheng Feng
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
32
|
A ZEB1-miR-375-YAP1 pathway regulates epithelial plasticity in prostate cancer. Oncogene 2016; 36:24-34. [PMID: 27270433 DOI: 10.1038/onc.2016.185] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/28/2016] [Accepted: 04/12/2016] [Indexed: 02/07/2023]
Abstract
MicroRNA-375 (miR-375) is frequently elevated in prostate tumors and cell-free fractions of patient blood, but its role in genesis and progression of prostate cancer is poorly understood. In this study, we demonstrated that miR-375 is inversely correlated with epithelial-mesenchymal transition signatures (EMT) in clinical samples and can drive mesenchymal-epithelial transition (MET) in model systems. Indeed, miR-375 potently inhibited invasion and migration of multiple prostate cancer lines. The transcription factor YAP1 was found to be a direct target of miR-375 in prostate cancer. Knockdown of YAP1 phenocopied miR-375 overexpression, and overexpression of YAP1 rescued anti-invasive effects mediated by miR-375. Furthermore, transcription of the miR-375 gene was shown to be directly repressed by the EMT transcription factor, ZEB1. Analysis of multiple patient cohorts provided evidence for this ZEB1-miR-375-YAP1 regulatory circuit in clinical samples. Despite its anti-invasive and anti-EMT capacities, plasma miR-375 was found to be correlated with circulating tumor cells in men with metastatic disease. Collectively, this study provides new insight into the function of miR-375 in prostate cancer, and more broadly identifies a novel pathway controlling epithelial plasticity and tumor cell invasion in this disease.
Collapse
|
33
|
Cekaite L, Eide PW, Lind GE, Skotheim RI, Lothe RA. MicroRNAs as growth regulators, their function and biomarker status in colorectal cancer. Oncotarget 2016; 7:6476-505. [PMID: 26623728 PMCID: PMC4872728 DOI: 10.18632/oncotarget.6390] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023] Open
Abstract
Gene expression is in part regulated by microRNAs (miRNAs). This review summarizes the current knowledge of miRNAs in colorectal cancer (CRC); their role as growth regulators, the mechanisms that regulate the miRNAs themselves and the potential of miRNAs as biomarkers. Although thousands of tissue samples and bodily fluids from CRC patients have been investigated for biomarker potential of miRNAs (>160 papers presented in a comprehensive tables), none single miRNA nor miRNA expression signatures are in clinical use for this disease. More than 500 miRNA-target pairs have been identified in CRC and we discuss how these regulatory nodes interconnect and affect signaling pathways in CRC progression.
Collapse
Affiliation(s)
- Lina Cekaite
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Peter W. Eide
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Guro E. Lind
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Rolf I. Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Ragnhild A. Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K.G.Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
34
|
Jepsen RK, Novotny GW, Klarskov LL, Christensen IJ, Riis LB, Høgdall E. Intra-tumor heterogeneity of microRNA-92a, microRNA-375 and microRNA-424 in colorectal cancer. Exp Mol Pathol 2016; 100:125-31. [DOI: 10.1016/j.yexmp.2015.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/03/2015] [Accepted: 12/05/2015] [Indexed: 12/11/2022]
|
35
|
MicroRNAs and Inflammation in Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 937:53-69. [PMID: 27573894 DOI: 10.1007/978-3-319-42059-2_3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Colorectal cancers (CRC) are known to be related to inflammatory conditions, and inflammatory bowel diseases increase the relative risk for developing CRC. The use of anti-inflammatory drugs prevents the development of colorectal cancer.Several molecular mediators are connecting the pathways that are involved in inflammatory conditions and in carcinogenesis. By the way these pathways are tightly interwoven, with the consequence that a deregulation at the level of any of these molecular mediators can affect the others.MiRNAs are demonstrated to be deregulated in inflammatory bowel diseases and in colorectal cancer. Moreover, they target several molecular mediators that connect inflammation to cancer, and they are thus implicated in the route from inflammation to colorectal cancer.This chapter will focus on the miRNAs that are jointly deregulated in inflammatory bowel disease and in colorectal cancer. Their role on the regulation of the molecular mediators and pathways that link inflammation to cancer will be described.
Collapse
|
36
|
Choi YC, Yoon S, Byun Y, Lee G, Kee H, Jeong Y, Yoon J, Baek K. MicroRNA library screening identifies growth-suppressive microRNAs that regulate genes involved in cell cycle progression and apoptosis. Exp Cell Res 2015; 339:320-32. [DOI: 10.1016/j.yexcr.2015.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/21/2015] [Accepted: 10/09/2015] [Indexed: 12/26/2022]
|
37
|
Sager M, Yeat NC, Pajaro-Van der Stadt S, Lin C, Ren Q, Lin J. Transcriptomics in cancer diagnostics: developments in technology, clinical research and commercialization. Expert Rev Mol Diagn 2015; 15:1589-603. [PMID: 26565429 DOI: 10.1586/14737159.2015.1105133] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transcriptomic technologies are evolving to diagnose cancer earlier and more accurately to provide greater predictive and prognostic utility to oncologists and patients. Digital techniques such as RNA sequencing are replacing still-imaging techniques to provide more detailed analysis of the transcriptome and aberrant expression that causes oncogenesis, while companion diagnostics are developing to determine the likely effectiveness of targeted treatments. This article examines recent advancements in molecular profiling research and technology as applied to cancer diagnosis, clinical applications and predictions for the future of personalized medicine in oncology.
Collapse
Affiliation(s)
- Monica Sager
- a College of Arts and Sciences , Washington University in St. Louis , St. Louis , MO , USA.,b Rare Genomics Institute , Bethesda , MD , USA
| | - Nai Chien Yeat
- b Rare Genomics Institute , Bethesda , MD , USA.,c School of Medicine , Washington University in St. Louis , St. Louis , MO , USA
| | - Stefan Pajaro-Van der Stadt
- a College of Arts and Sciences , Washington University in St. Louis , St. Louis , MO , USA.,b Rare Genomics Institute , Bethesda , MD , USA
| | - Charlotte Lin
- b Rare Genomics Institute , Bethesda , MD , USA.,c School of Medicine , Washington University in St. Louis , St. Louis , MO , USA
| | - Qiuyin Ren
- b Rare Genomics Institute , Bethesda , MD , USA.,d Whiting School of Engineering , Johns Hopkins University , Baltimore , MD , USA
| | - Jimmy Lin
- b Rare Genomics Institute , Bethesda , MD , USA
| |
Collapse
|
38
|
XU RAN, ZENG GUANG, GAO JING, REN YUE, ZHANG ZHE, ZHANG QINGNA, ZHAO JINXIU, TAO HONG, LI DAXU. miR-138 suppresses the proliferation of oral squamous cell carcinoma cells by targeting Yes-associated protein 1. Oncol Rep 2015; 34:2171-8. [DOI: 10.3892/or.2015.4144] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/06/2015] [Indexed: 11/05/2022] Open
|
39
|
Jafri MA, Zaidi SK, Ansari SA, Al-Qahtani MH, Shay JW. MicroRNAs as potential drug targets for therapeutic intervention in colorectal cancer. Expert Opin Ther Targets 2015; 19:1705-23. [PMID: 26189482 DOI: 10.1517/14728222.2015.1069816] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION MicroRNAs (miRNAs) are small (19 - 22 nucleotide), non-protein-coding RNA segments that function as master regulators of hundreds of genes simultaneously in both normal and malignant cells. In colorectal cancer (CRC) miRNAs are deregulated and have critical roles in initiation and progression of CRC by interacting with various oncogenes and tumor suppressor genes including APC, KRAS and p53, or by modulating downstream signal transduction pathways. Numerous promising miRNAs have emerged as potential drug targets for therapeutic intervention and possible candidates for replacement therapy in CRC. AREAS COVERED In this review the authors summarize the available information on miRNAs and their role in CRC. The authors point out specific miRNAs as potential drug targets and those having a significant role in gene activation and gene silencing during the process of CRC development, to highlight their importance as possible therapeutic candidates for the treatment of CRC. EXPERT OPINION Targeting miRNAs provides an emerging opportunity to develop effective miRNA-based replacement therapy or antagonists to alter expression in colon cancer patient tumors. However, the biggest challenge is to overcome obstacles associated with pharmacokinetics, delivery and toxicity in order to translate the potential of miRNAs into efficacious anticancer drugs.
Collapse
Affiliation(s)
- Mohammad Alam Jafri
- a 1 King Abdulaziz University, Center of Excellence in Genomic Medicine Research , Jeddah, Saudi Arabia
| | - Syed Kashif Zaidi
- a 1 King Abdulaziz University, Center of Excellence in Genomic Medicine Research , Jeddah, Saudi Arabia
| | - Shakeel Ahmed Ansari
- a 1 King Abdulaziz University, Center of Excellence in Genomic Medicine Research , Jeddah, Saudi Arabia
| | | | - Jerry W Shay
- a 1 King Abdulaziz University, Center of Excellence in Genomic Medicine Research , Jeddah, Saudi Arabia.,b 2 UT Southwestern Medical Center, Department of Cell Biology , Dallas, TX, USA +1 214 648 4201 ; +1 214 648 5814 ;
| |
Collapse
|
40
|
Zhang XW, Liu N, Chen S, Wang YE, Sun KL, Xu ZM, Fu WN. Upregulation of microRNA-23a regulates proliferation and apoptosis by targeting APAF-1 in laryngeal carcinoma. Oncol Lett 2015; 10:410-416. [PMID: 26171041 DOI: 10.3892/ol.2015.3238] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 04/27/2015] [Indexed: 11/06/2022] Open
Abstract
MicroRNA-23a (miR-23a) is a potential biomarker for laryngeal cancer. Apoptotic protease activating factor 1 (APAF-1) was recently demonstrated to be a target of miR-23a. However, whether miR-23a exerts its effects via APAF-1 in laryngeal cancer, remains unknown. In the present study, miR-23a expression was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). APAF-1 mRNA and protein expression levels were assayed by RT-qPCR and western blotting, respectively. Binding of miR-23a to APAF-1 was monitored by a luciferase reporter assay. Gain-of-function and loss-of-function studies were performed in order to investigate the roles of miR-23a and APAF-1 in Hep2 cell proliferation and apoptosis. miR-23a and APAF-1 were found to be significantly upregulated and downregulated, respectively, in laryngeal cancer tissues, and there was a significant negative correlation between APAF-1 and miR-23a expression. The results of the luciferase reporter assay demonstrated that miR-23a bound directly to the APAF-1 mRNA 3'-untranslated region. Ectopic expression of miR-23a and knockdown of APAF-1 significantly promoted cell proliferation and colony formation, and inhibited early apoptosis in Hep2 cells. In conclusion, miR-23a acts as an oncogenic regulator in laryngeal carcinoma by directly targeting APAF-1, and may be a useful biomarker in the diagnosis and treatment of laryngeal carcinoma.
Collapse
Affiliation(s)
- Xiao-Wen Zhang
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ning Liu
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Sheng Chen
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Y E Wang
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Kai-Lai Sun
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhen-Ming Xu
- Department of Otolaryngology, No. 463 Hospital of PLA, Shenyang, Liaoning 110007, P.R. China
| | - Wei-Neng Fu
- Department of Medical Genetics, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
41
|
Eulalio A, Mano M. MicroRNA Screening and the Quest for Biologically Relevant Targets. ACTA ACUST UNITED AC 2015; 20:1003-17. [DOI: 10.1177/1087057115578837] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/03/2015] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are a class of genome-encoded small RNAs that post-transcriptionally regulate gene expression by repressing target transcripts containing partially or fully complementary binding sites. Despite their relatively low number, miRNAs have been shown to directly regulate a large fraction of the transcriptome. In agreement with their pervasive role in the regulation of eukaryotic gene expression, miRNAs have been implicated in virtually all biological processes, including different pathologies. The use of screening technologies to systematically analyze miRNA function in cell-based assays offers a unique opportunity to gain new insights into complex biological and disease-relevant processes. Given the low complexity of the miRNome and the similarities to small interfering RNA (siRNA) screening experimental approaches, phenotypic screening using genome-wide libraries of miRNA mimics or inhibitors is not, per se, technically challenging. The identification of miRNA targets and, more importantly, the characterization of their mechanisms of action through the identification of the key targets underlying observed phenotypes remain the major challenges of this approach. This article provides an overview of cell-based screenings for miRNA function that were performed in different biological contexts. The advantages and limitations of computational and experimental approaches commonly used to identify miRNA targets are also discussed.
Collapse
Affiliation(s)
- Ana Eulalio
- Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- UC-BIOTECH, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
42
|
Peng J, Xie Z, Cheng L, Zhang Y, Chen J, Yu H, Li Z, Kang H. Paired design study by real-time PCR: miR-378* and miR-145 are potent early diagnostic biomarkers of human colorectal cancer. BMC Cancer 2015; 15:158. [PMID: 25896668 PMCID: PMC4436811 DOI: 10.1186/s12885-015-1123-2] [Citation(s) in RCA: 23] [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: 06/27/2014] [Accepted: 02/24/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Although microRNAs offer great potential as cancer biomarkers, effective clinical dignostics and tumor maker have not been verified to diagnose with colorectal cancer (CRC). The purpose of our study is to systematically assess the expression of miRNAs in matched cancer and normal tissue samples to identify promising diagnostic microRNA (miRNA) biomarkers for CRC. METHODS In our study, we examined by Real-Time PCR the expression levels of 96 mature miRNA in 32 CRC patients with differently expressed tumors versus normal colon tissues. Using enter and stepwise variable selection methods separately, conditional logistic regression was conducted to identify miRNAs associated with CRC. The classification performance of these indicators was assessed under the Fisher discriminant analysis. Receiver operating characteristic curve analyses were applied to obtain diagnostic utility of the differentially expressed miRNAs. RESULTS In this study, we confirmed 11 overexpressed miRNAs with no less than twofold difference, and 85 downexpressed miRNAs with up to 0.5-fold difference in CRC from 96 aberrantly expressed miRNAs being identified by real-time PCR. Conditional logistic regression results confirmed that miRNA-378 and miRNA-145 expression profile was statistically significant. The error diagnosis rate of these two miRNAs are 0.194 and 0.113, separeately, showing by discriminant analysis. CONCLUSIONS MiRNA-145 and miRNA-378* are potential biomarkers for early detection of CRC, which may help in diagnosing CRC in early period.
Collapse
Affiliation(s)
- Juan Peng
- Guilin Medical College, Huang Cheng North 2 Road 109, Qixing District, Guilin, Guangxi, China.
| | - Zhengyong Xie
- General Hospital of Guangzhou Military Command of PLA, Liuhua Road 111, Guangzhou, Guangdong, China.
| | - Liyang Cheng
- General Hospital of Guangzhou Military Command of PLA, Liuhua Road 111, Guangzhou, Guangdong, China.
| | - Yuxin Zhang
- General Hospital of Guangzhou Military Command of PLA, Liuhua Road 111, Guangzhou, Guangdong, China.
| | - Junyong Chen
- General Hospital of Guangzhou Military Command of PLA, Liuhua Road 111, Guangzhou, Guangdong, China.
| | - Hongping Yu
- Guilin Medical College, Huang Cheng North 2 Road 109, Qixing District, Guilin, Guangxi, China.
| | - Zehang Li
- General Hospital of Guangzhou Military Command of PLA, Liuhua Road 111, Guangzhou, Guangdong, China.
| | - Huixing Kang
- General Hospital of Guangzhou Military Command of PLA, Liuhua Road 111, Guangzhou, Guangdong, China.
| |
Collapse
|
43
|
Zhang S, Zhang H, Zhu J, Zhang X, Liu Y. MiR-522 contributes to cell proliferation of human glioblastoma cells by suppressing PHLPP1 expression. Biomed Pharmacother 2015; 70:164-9. [DOI: 10.1016/j.biopha.2015.01.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 01/04/2015] [Indexed: 02/07/2023] Open
|
44
|
Wang R, Ma Y, Yu D, Zhao J, Ma P. miR-377 functions as a tumor suppressor in human clear cell renal cell carcinoma by targeting ETS1. Biomed Pharmacother 2015; 70:64-71. [DOI: 10.1016/j.biopha.2015.01.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/04/2015] [Indexed: 12/12/2022] Open
|
45
|
Liu F, Ji F, Ji Y, Jiang Y, Sun X, Lu Y, Zhang L, Han Y, Liu X. Dissecting the mechanism of colorectal tumorigenesis based on RNA-sequencing data. Exp Mol Pathol 2015; 98:246-53. [PMID: 25576648 DOI: 10.1016/j.yexmp.2015.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/26/2014] [Accepted: 01/05/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This study aimed to identify the differentially expressed genes (DEGs), mutated genes and fusion genes in colorectal cancer. MATERIALS AND METHODS RNA-sequencing data (ID: SRP009386) from cancerous, paracancerous non-tumor and distant normal tissue from one Chinese patient with stage III colorectal cancer were downloaded from Sequence Read Archive. Quality control was checked using FastQC, followed by sequence alignment against the hg19 reference genome using TopHat v1.3.3. The expression levels were quantified using Cufflinks, followed by DEGs screening using NOISeq. Enrichment analysis was performed using DAVID. Transcription factors were screened using TRANSFA. Mutated loci were identified using SAMTools and VCFTools. Gene fusion events were detected by TopHat-fusion. RESULTS In total 2440, 1887 and 834 DEGs were respectively detected in cancerous vs. normal tissue, cancerous vs. paracancerous tissue and paracancerous vs. normal tissue. The up-regulated genes from cancerous and paracancerous tissue compared with normal tissue were enriched in "extracellular matrix receptor interaction" and "focal adhesion pathway" as well as some biological processes except for "negative regulation of programmed cell death" uniquely presenting in cancer. Dysregulated transcription factors including SOX4, BCL6, CEBPB and MSX2 were enriched in the unique biological process. Trp53 was identified with one mutated locus 7577142 (C → T) on chromosome 17. BCL6 also experienced missense mutation. Additionally, COL1A1-PPP2R2C and EXPH5-COL1A2 were observed fusion genes in cancer tissue. CONCLUSIONS The unique biological process in cancer tissue may be the cause for colorectal carcinogenesis. The screened transcription factors, mutated genes and fusion genes may contribute to the progression of colorectal cancer.
Collapse
Affiliation(s)
- Fuguo Liu
- Department of Gastroenterology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Fengzhi Ji
- Department of Gastroenterology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Yuling Ji
- Statistics Division, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Yueping Jiang
- Department of Gastroenterology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Xueguo Sun
- Department of Gastroenterology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Yanyan Lu
- Department of Gastroenterology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Lingyun Zhang
- Department of Gastroenterology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Yue Han
- Department of Gastroenterology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| | - Xishuang Liu
- Department of Gastroenterology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, Shandong Province, China.
| |
Collapse
|
46
|
Xu L, Li M, Wang M, Yan D, Feng G, An G. The expression of microRNA-375 in plasma and tissue is matched in human colorectal cancer. BMC Cancer 2014; 14:714. [PMID: 25255814 PMCID: PMC4181388 DOI: 10.1186/1471-2407-14-714] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 09/22/2014] [Indexed: 12/18/2022] Open
Abstract
Background MicroRNAs (miRNAs) offer great potential as cancer biomarkers. The importance of miRNAs profiling in tissue and body fluids in colorectal cancer (CRC) have been addressed respectively in many studies. The purpose of our study is to systematically assess the expression of miRNAs in cancer tissue and matched plasma samples and to evaluate their usefulness as minimally invasive diagnostic biomarkers for the detection of CRC. Methods The study was divided into two phases: firstly, qRT-PCR based TaqMan Low Density MiRNA Arrays (TLDAs) was used to screen the differentially expressed miRNAs in 6 plasma samples of CRC patients and 6 healthy controls. Secondly, marker validation by stem-loop reverse transcription real-time PCR using an independent set of paired cancer tissues (n = 88) and matched plasma samples (CRC, n = 88; control, n = 40). Correlation analysis was determined by Pearson’s test. Receiver operating characteristic curve analyses were applied to obtain diagnostic utility of the differentially expressed miRNAs. Target gene prediction and signal pathway analyses were used to predict the function of miRNAs. Results TLDAs identified 42 miRNAs, which were differentially expressed in patients and healthy individuals. Five of them (miR-375, miR-150, miR-206, miR-125b and miR-126*) were chosen to be validated in plasma and tissue samples. The results indicated that for plasma sample, miR-375 (p < 0.0001) and miR-206 (p = 0.0002) were dysregulated and could discriminate CRC patients from healthy controls. For tissue samples, miR-375 (p < 0.0001), miR-150 (p < 0.0001), miR-125b (p = 0.0065) and miR-126*(p = 0.0009) were down-regulated. miR-375 was significantly down-regulated and positively correlated in both tissue and plasma samples (r = 0.4663, p = 0.0007). Gene ontology and signal pathway analyses showed that most of the target genes that were regulated by miR-375 were involved in some critical pathways in the development and progression of cancer. Conclusions Our results indicate that the down-regulation of miR-375 in plasma and tissue is matched in CRC. Moreover, bioinformatics prediction revealed miR-375 association with some critical signal pathways in the development and progression of CRC. Therefore, plasma miR-375 holds great promise to be an alternative tissue biomarker for CRC detection. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-714) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | - Guosheng Feng
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | | |
Collapse
|
47
|
Li W, Zang W, Liu P, Wang Y, Du Y, Chen X, Deng M, Sun W, Wang L, Zhao G, Zhai B. MicroRNA-124 inhibits cellular proliferation and invasion by targeting Ets-1 in breast cancer. Tumour Biol 2014; 35:10897-904. [PMID: 25085587 DOI: 10.1007/s13277-014-2402-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/23/2014] [Indexed: 11/29/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that, by targeting certain messenger RNAs (mRNAs) for translational repression or cleavage, can regulate the expression of these genes. In addition, miRNAs may also function as oncogenes and tumor-suppressor genes, as the abnormal expression of miRNAs is associated with various human tumors. However, the effects of the expression of miR-124 in breast cancer remain unclear. The present study was conducted to study the expression of miR-124 in breast cancer, paying particular attention to miR-124's relation to the proliferation, invasion, and apoptosis in breast cancer cell MCF-7 and MDA-MB-231. Real-time quantitative RT-PCR (qRT-PCR) was performed to identify miR-124 that was down-regulated in breast cancer tissues. We also showed E26 transformation specific-1 (Ets-1) and miR-124 expression levels in breast cancer tissues that were associated with lymph node metastases. With transfected synthetic miR-124 agomir into MCF-7 and MDA-MB-231, a significant reduction (P < 0.05) in MCF-7 and MDA-MB-231 cell proliferation and colony forming potential was observed after treatment with miR-124. Apoptosis and migration rates were found to be significantly higher in two breast-derived cell lines transfected with a miR-124 agomir (P < 0.05). Luciferase reporter assay and Western blot were used to verify Ets-1 as a potential major target gene of miR-124, and the result showed that miR-124 can bind to putative binding sites within the Ets-1 mRNA 3' untranslated region (UTR) to reduce its expression. Based on these findings, we propose that miR-124 and Ets-1 may serve as a therapeutic agent in breast cancer.
Collapse
Affiliation(s)
- Wentao Li
- Department of Breast Surgery, The People's Hospital of Henan Province (The People's Hospital of Zhengzhou University), No. 7 Weiwu Road, Zhengzhou, 450003, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|