201
|
Neault M, Couteau F, Bonneau É, De Guire V, Mallette FA. Molecular Regulation of Cellular Senescence by MicroRNAs: Implications in Cancer and Age-Related Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:27-98. [DOI: 10.1016/bs.ircmb.2017.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
202
|
Xiao JH, Hao QY, Wang K, Paul J, Wang YX. Emerging Role of MicroRNAs and Long Noncoding RNAs in Healthy and Diseased Lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:343-359. [DOI: 10.1007/978-3-319-63245-2_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
203
|
|
204
|
Barbato S, Solaini G, Fabbri M. MicroRNAs in Oncogenesis and Tumor Suppression. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 333:229-268. [PMID: 28729026 DOI: 10.1016/bs.ircmb.2017.05.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
MicroRNAs (MiRNAs) have emerged in the last 15 years as central players in the biology of cancer. Increasing lines of evidence have supported their regulatory role in the expression of both oncogenes and tumor-suppressor genes, progressively clarifying which genes are modulated by specific MiRNAs dysregulated in cancer. Intriguingly, a "target-specific" understanding of MiRNA function in oncology has been replaced by a more "pathway-specific" vision of their involvement in cancer biology. This work provides a state-of-the-art knowledge of the role of MiRNAs in the most frequently altered signaling pathways in cancer cells and provides an updated overview on some of the most relevant findings trying to decode the complex molecular mechanisms of cancer.
Collapse
Affiliation(s)
- Simona Barbato
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna, Italy
| | - Giancarlo Solaini
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna, Italy
| | - Muller Fabbri
- Children's Center for Cancer and Blood Diseases and The Saban Research Institute, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
205
|
Li CH, Xiao Z, Tong JHM, To KF, Fang X, Cheng ASL, Chen Y. EZH2 coupled with HOTAIR to silence MicroRNA-34a by the induction of heterochromatin formation in human pancreatic ductal adenocarcinoma. Int J Cancer 2017; 140:120-129. [DOI: 10.1002/ijc.30414] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Chi-Han Li
- School of Biomedical Sciences, Faculty of Medicine; The Chinese University of Hong Kong; Shatin NT Hong Kong
| | - Zhangang Xiao
- School of Biomedical Sciences, Faculty of Medicine; The Chinese University of Hong Kong; Shatin NT Hong Kong
| | - Joanna Hung-Man Tong
- Department of Anatomical and Cellular Pathology; Prince of Wales Hospital, The Chinese University of Hong Kong; Shatin Hong Kong
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology; Prince of Wales Hospital, The Chinese University of Hong Kong; Shatin Hong Kong
| | - Xiangdong Fang
- CAS Key Laboratory of Genome Sciences and Information; Beijing Institute of Genomics, Chinese Academy of Sciences; Beijing China
| | - Alfred SL Cheng
- School of Biomedical Sciences, Faculty of Medicine; The Chinese University of Hong Kong; Shatin NT Hong Kong
- State Key Laboratory of Digestive Diseases; The Chinese University of Hong Kong; Shatin NT Hong Kong
| | - Yangchao Chen
- School of Biomedical Sciences, Faculty of Medicine; The Chinese University of Hong Kong; Shatin NT Hong Kong
- State Key Laboratory of Digestive Diseases; The Chinese University of Hong Kong; Shatin NT Hong Kong
- Shenzhen Research Institute, The Chinese University of Hong Kong; Shenzhen China
| |
Collapse
|
206
|
Krause CJ, Popp O, Thirunarayanan N, Dittmar G, Lipp M, Müller G. MicroRNA-34a promotes genomic instability by a broad suppression of genome maintenance mechanisms downstream of the oncogene KSHV-vGPCR. Oncotarget 2016; 7:10414-32. [PMID: 26871287 PMCID: PMC4891129 DOI: 10.18632/oncotarget.7248] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 01/17/2016] [Indexed: 12/18/2022] Open
Abstract
The Kaposi's sarcoma-associated herpesvirus (KSHV)-encoded chemokine receptor vGPCR acts as an oncogene in Kaposi's sarcomagenesis. Until now, the molecular mechanisms by which the vGPCR contributes to tumor development remain incompletely understood. Here, we show that the KSHV-vGPCR contributes to tumor progression through microRNA (miR)-34a-mediated induction of genomic instability. Large-scale analyses on the DNA, gene and protein level of cell lines derived from a mouse model of vGPCR-driven tumorigenesis revealed that a vGPCR–induced upregulation of miR-34a resulted in a broad suppression of genome maintenance genes. A knockdown of either the vGPCR or miR-34a largely restored the expression of these genes and confirmed miR-34a as a downstream effector of the KSHV-vGPCR that compromises genome maintenance mechanisms. This novel, protumorigenic role of miR-34a questions the use of miR-34a mimetics in cancer therapy as they could impair genome stability.
Collapse
Affiliation(s)
- Claudia J Krause
- Molecular Tumor Genetics and Immunogenetics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Oliver Popp
- Mass Spectrometry Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Nanthakumar Thirunarayanan
- Molecular Tumor Genetics and Immunogenetics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gunnar Dittmar
- Mass Spectrometry Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Martin Lipp
- Molecular Tumor Genetics and Immunogenetics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gerd Müller
- Molecular Tumor Genetics and Immunogenetics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
207
|
Yacoub RA, Fawzy IO, Assal RA, Hosny KA, Zekri ARN, Esmat G, El Tayebi HM, Abdelaziz AI. miR-34a: Multiple Opposing Targets and One Destiny in Hepatocellular Carcinoma. J Clin Transl Hepatol 2016; 4:300-305. [PMID: 28097098 PMCID: PMC5225149 DOI: 10.14218/jcth.2016.00031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/29/2016] [Accepted: 12/22/2016] [Indexed: 01/26/2023] Open
Abstract
Background and Aims: The role of miR-34a in hepatocellular carcinoma (HCC) is controversial and several unresolved issues remain, including its expression pattern and relevance to tumor etiology, tumor stage and prognosis, and finally, its impact on apoptosis. Methods: miR-34a expression was assessed in hepatitis C virus (HCV)-induced non-metastatic HCC tissues by RT-Q-PCR. Huh-7 cells were transfected with miR-34a mimics and the impact of miR-34a was examined on 84 pro-apoptotic/anti-apoptotic genes using PCR array; its net effect was tested on cell viability via MTT assay. Results: miR-34a expression was up-regulated in HCC tissues. Moreover, miR-34a induced a large set of pro-apoptotic/anti-apoptotic genes, with a net result of triggering apoptosis and repressing cell viability. Conclusions: HCC-related differential expression of miR-34a could be etiology-based or stage-specific, and low expression of miR-34a may predict poor prognosis. This study's findings also emphasize the role of miR-34a in apoptosis.
Collapse
Affiliation(s)
- Radwa Alaa Yacoub
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Injie Omar Fawzy
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Reem Amr Assal
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Karim Adel Hosny
- Department of General Surgery, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Abdel-Rahman Nabawy Zekri
- Virology and Immunology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Gamal Esmat
- Department of Endemic Medicine and Hepatology, Cairo University, Cairo, Egypt
| | - Hend Mohamed El Tayebi
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ahmed Ihab Abdelaziz
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
- School of Medicine, New Giza University, Cairo, Egypt
| |
Collapse
|
208
|
Nassar FJ, Nasr R, Talhouk R. MicroRNAs as biomarkers for early breast cancer diagnosis, prognosis and therapy prediction. Pharmacol Ther 2016; 172:34-49. [PMID: 27916656 DOI: 10.1016/j.pharmthera.2016.11.012] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Breast cancer is a major health problem that affects one in eight women worldwide. As such, detecting breast cancer at an early stage anticipates better disease outcome and prolonged patient survival. Extensive research has shown that microRNA (miRNA) are dysregulated at all stages of breast cancer. miRNA are a class of small noncoding RNA molecules that can modulate gene expression and are easily accessible and quantifiable. This review highlights miRNA as diagnostic, prognostic and therapy predictive biomarkers for early breast cancer with an emphasis on the latter. It also examines the challenges that lie ahead in their use as biomarkers. Noteworthy, this review addresses miRNAs reported in patients with early breast cancer prior to chemotherapy, radiotherapy, surgical procedures or distant metastasis (unless indicated otherwise). In this context, miRNA that are mentioned in this review were significantly modulated using more than one statistical test and/or validated by at least two studies. A standardized protocol for miRNA assessment is proposed starting from sample collection to data analysis that ensures comparative analysis of data and reproducibility of results.
Collapse
Affiliation(s)
- Farah J Nassar
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Rihab Nasr
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Rabih Talhouk
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
209
|
Affiliation(s)
- Junguo Ma
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Xiaoyu Li
- College of Life Science, Henan Normal University, Xinxiang, China
| |
Collapse
|
210
|
Barbagallo D, Condorelli A, Ragusa M, Salito L, Sammito M, Banelli B, Caltabiano R, Barbagallo G, Zappalà A, Battaglia R, Cirnigliaro M, Lanzafame S, Vasquez E, Parenti R, Cicirata F, Di Pietro C, Romani M, Purrello M. Dysregulated miR-671-5p / CDR1-AS / CDR1 / VSNL1 axis is involved in glioblastoma multiforme. Oncotarget 2016; 7:4746-59. [PMID: 26683098 PMCID: PMC4826240 DOI: 10.18632/oncotarget.6621] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 11/14/2015] [Indexed: 01/04/2023] Open
Abstract
MiR-671-5p is encoded by a gene localized at 7q36.1, a region amplified in human glioblastoma multiforme (GBM), the most malignant brain cancer. To investigate whether expression of miR-671-5p were altered in GBM, we analyzed biopsies from a cohort of forty-five GBM patients and from five GBM cell lines. Our data show significant overexpression of miR-671-5p in both biopsies and cell lines. By exploiting specific miRNA mimics and inhibitors, we demonstrated that miR-671-5p overexpression significantly increases migration and to a less extent proliferation rates of GBM cells. Through a combined in silico and in vitro approach, we identified CDR1-AS, CDR1, VSNL1 as downstream miR-671-5p targets in GBM. Expression of these genes significantly decreased both in GBM biopsies and cell lines and negatively correlated with that of miR-671-5p. Based on our data, we propose that the axis miR-671-5p / CDR1-AS / CDR1 / VSNL1 is functionally altered in GBM cells and is involved in the modification of their biopathological profile.
Collapse
Affiliation(s)
- Davide Barbagallo
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| | - Angelo Condorelli
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| | - Marco Ragusa
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| | - Loredana Salito
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| | - Mariangela Sammito
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| | - Barbara Banelli
- UOS Epigenetica dei Tumori, IRCCS A.O.U. San Martino-IST, Genova, Italy, EU
| | - Rosario Caltabiano
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate G.F. Ingrassia, Università di Catania, Catania, Italy, EU
| | - Giuseppe Barbagallo
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate G.F. Ingrassia, Università di Catania, Catania, Italy, EU
| | - Agata Zappalà
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Fisiologia, Università di Catania, Catania, Italy, EU
| | - Rosalia Battaglia
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| | - Matilde Cirnigliaro
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| | - Salvatore Lanzafame
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate G.F. Ingrassia, Università di Catania, Catania, Italy, EU
| | - Enrico Vasquez
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate G.F. Ingrassia, Università di Catania, Catania, Italy, EU
| | - Rosalba Parenti
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Fisiologia, Università di Catania, Catania, Italy, EU
| | - Federico Cicirata
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Fisiologia, Università di Catania, Catania, Italy, EU
| | - Cinzia Di Pietro
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| | - Massimo Romani
- UOS Epigenetica dei Tumori, IRCCS A.O.U. San Martino-IST, Genova, Italy, EU
| | - Michele Purrello
- Dipartimento di Scienze Biomediche e Biotecnologiche, Sezione di Biologia e Genetica G Sichel, Unità di BioMedicina Molecolare, Genomica e dei Sistemi Complessi, Università di Catania, Catania, Italy, EU
| |
Collapse
|
211
|
Majidinia M, Yousefi B. DNA damage response regulation by microRNAs as a therapeutic target in cancer. DNA Repair (Amst) 2016; 47:1-11. [DOI: 10.1016/j.dnarep.2016.09.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/12/2022]
|
212
|
Xiang W, Lin H, Wang Q, Chen W, Liu Z, Chen H, Zhang H, Chen W. miR‑34a suppresses proliferation and induces apoptosis of human lens epithelial cells by targeting E2F3. Mol Med Rep 2016; 14:5049-5056. [PMID: 27840975 PMCID: PMC5355663 DOI: 10.3892/mmr.2016.5901] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 10/06/2016] [Indexed: 12/29/2022] Open
Abstract
microRNA (miRNA) is abnormally expressed in numerous diseases, and it was intimately associated with cell proliferation and apoptosis. However, the mechanism by which miRNAs control cataractogenesis remains unclear. In the current study, it was demonstrated that miR-34a was highly expressed in the cataractous lens by stem-loop reverse transcription-quantitative polymerase chain reaction. Trying to investigate the role of miR-34a in human lens epithelial cells, miR-34a mimics were transfected into SRA01/04 cells, and this suppressed proliferation and induced apoptosis. Subsequently, E2F3 was confirmed as a direct target of miR-34a. Downregulation of E2F3 by small interfering (si) RNA siE2F3 resulted in proliferation inhibition and apoptosis of SRA01/04 cells. Furthermore, it was demonstrated that miR-34a and siE2F3 downregulated E2F3 expression at a protein level. In summary, the current study demonstrated that miR-34a suppressed the proliferation and induced apoptosis of SRA01/04 cells by downregulating E2F3. These observations provide novel insights with potential therapeutic applications for the treatment of cataracts.
Collapse
Affiliation(s)
- Wu Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Haotian Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Qilin Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Wan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Zhaochuan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Hui Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat‑Sen University, Guangzhou, Guangdong 510074, P.R. China
| | - Weirong Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
213
|
Sirt1 expression is associated with CD31 expression in blood cells from patients with chronic obstructive pulmonary disease. Respir Res 2016; 17:139. [PMID: 27784320 PMCID: PMC5081972 DOI: 10.1186/s12931-016-0452-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/14/2016] [Indexed: 12/31/2022] Open
Abstract
Background Cigarette smoke induced oxidative stress has been shown to reduce silent information regulator 1 (Sirt1) levels in lung tissue from smokers and patients with COPD patients. Sirt1 is known to inhibit endothelial senescence and may play a protective role in vascular cells. Endothelial progenitor cells (EPCs) are mobilized into circulation under various pathophysiological conditions, and are thought to play an important role in tissue repair in chronic obstructive lung disease (COPD). Therefore, Sirt1 and EPC-associated mRNAs were measured in blood samples from patients with COPD and from cultured CD34+ progenitor cells to examine whether these genes are associated with COPD development. Methods This study included 358 patients with a smoking history of more than 10 pack-years. RNA was extracted from blood samples and from CD34+ progenitor cells treated with cigarette smoke extract (CSE), followed by assessment of CD31, CD34, Sirt1 mRNA, miR-34a, and miR-126-3p expression by real-time RT-PCR. Results The expression of CD31, CD34, Sirt1 mRNAs, and miR-126-3p decreased and that of miR-34a increased in moderate COPD compared with that in control smokers. However, no significant differences in these genes were observed in blood cells from patients with severe COPD compared with those in control smokers. CSE significantly decreased Sirt1 and increased miR-34a expression in cultured progenitor cells. Conclusion Sirt1 expression in blood cells from patients with COPD could be a biomarker for disease stability in patients with moderate COPD. MiR-34a may participate in apoptosis and/or senescence of EPCs in smokers. Decreased expression of CD31, CD34, and miR-126-3p potentially represents decreased numbers of EPCs in blood cell from patients with COPD.
Collapse
|
214
|
Fan N, Wang J. MicroRNA 34a contributes to virus-mediated apoptosis through binding to its target gene Bax in influenza A virus infection. Biomed Pharmacother 2016; 83:1464-1470. [DOI: 10.1016/j.biopha.2016.08.049] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/18/2016] [Accepted: 08/18/2016] [Indexed: 12/11/2022] Open
|
215
|
Mohammadi A, Mansoori B, Baradaran B. The role of microRNAs in colorectal cancer. Biomed Pharmacother 2016; 84:705-713. [PMID: 27701052 DOI: 10.1016/j.biopha.2016.09.099] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/25/2016] [Accepted: 09/26/2016] [Indexed: 01/30/2023] Open
Abstract
Colorectal cancer (CRC) is still the third most common cancer in the world. Mechanism of CRC tumorigenesis has been widely studied at the molecular levels, and has been recently entered the area of microRNAs. MicroRNAs are small 19 to 22 nucleotides of RNA that engage in the regulation of cell differentiation, apoptosis, and cell cycle progression. MicroRNAs are similar to small interfering RNA (siRNA), that post-transcriptionally regulate gene expression and control various cellular mechanisms. They are important factors in the carcinogenesis of CRC, one of the most important factors includes microRNA. MicroRNAs have been linked to CRC development, and these molecules have been recently studied as new potential biomarkers in diagnosis and treatment of CRC. Specific microRNA expression patterns help distinguish CRC from other colon related disease, and may be used as a prognostication factor in patients after treatment with different chemotherapy drugs. More over the newest molecular therapy via tumor suppressor micro RNA replacement can be new insight in molecular therapy of CRC. This review summarizes the potential roles of microRNAs as potential biomarkers for CRC diagnosis, and treatment.
Collapse
Affiliation(s)
- Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
216
|
Abstract
High-throughput sequencing of cancer genomes is increasingly becoming an essential tool of clinical oncology that facilitates target identification and targeted therapy within the context of precision medicine. The cumulative profiles of somatic mutations in cancer yielded by comprehensive molecular studies also constitute a fingerprint of historical exposures to exogenous and endogenous mutagens, providing insight into cancer evolution and etiology. Mutational signatures that were first established by inspection of the TP53 gene somatic landscape have now been confirmed and expanded by comprehensive sequencing studies. Further, the degree of granularity achieved by deep sequencing allows detection of low-abundance mutations with clinical relevance. In tumors, they represent the emergence of small aggressive clones; in normal tissues, they signal a mutagenic exposure related to cancer risk; and, in blood, they may soon become effective surveillance tools for diagnostic purposes and for monitoring of cancer prognosis and recurrence.
Collapse
Affiliation(s)
- Ana I Robles
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Jin Jen
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, and Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
217
|
Rupaimoole R, Slack FJ. A role for miR-34 in colon cancer stem cell homeostasis. Stem Cell Investig 2016; 3:42. [PMID: 27668249 DOI: 10.21037/sci.2016.08.04] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/09/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Rajesha Rupaimoole
- Department of Pathology and Institute of RNA Medicine, Beth Israel Deaconess Medical Center Cancer Center, Harvard Medical School, Boston, MA 02215, USA
| | - Frank J Slack
- Department of Pathology and Institute of RNA Medicine, Beth Israel Deaconess Medical Center Cancer Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
218
|
Liu L, Liu L, Shi J, Tan M, Xiong J, Li X, Hu Q, Yi Z, Mao D. MicroRNA-34b mediates hippocampal astrocyte apoptosis in a rat model of recurrent seizures. BMC Neurosci 2016; 17:56. [PMID: 27514646 PMCID: PMC4981991 DOI: 10.1186/s12868-016-0291-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 07/21/2016] [Indexed: 01/03/2023] Open
Abstract
Background Recurrent convulsions can cause irreversible astrocyte death, impede neuron regeneration, and further aggravate brain damage. MicroRNAs have been revealed as players in the progression of numerous diseases including cancer and Alzheimer’s disease. Particularly, microRNA has been found linked to seizure-induced neuronal death. In this study, a rat model of recurrent convulsions induced by flurothyl treatments was utilised to assess the alterations of microRNA expressions in hippocampus tissues. We also applied an in vitro model in which primary astrocytes were exposed to kainic acid to verify the targets of miR-34b-5p identified in the animal model. Results We discovered that miR-34b-5p, a member of the miR-34 family, increased significantly in flurothyl-treated rat hippocampus tissue. More surprisingly, this upregulation occurred concurrently with accumulating astrocyte apoptosis, indicating the involvement of miR-34b-5p in seizures caused astrocyte apoptosis. Results from the in vitro experiments further demonstrated that miR-34b-5p directly targeted Bcl-2 mRNA, translationally repressed Bcl-2 protein, and thus modulated cell apoptosis by influencing Bcl-2, Bax, and Caspase-3. Conclusion Our findings prove microRNAs play a role in mediating recurrent convulsions-induced astrocyte death and further indicate that miR-34b-5p could acts as a regulator for astrocyte apoptosis induced by recurrent seizures. Electronic supplementary material The online version of this article (doi:10.1186/s12868-016-0291-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | - Jiayun Shi
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | - Menglin Tan
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | - Jie Xiong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | - Xingfang Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | - Qingpeng Hu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | - Zhuwen Yi
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China
| | - Ding'an Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 86 Renmin Middle Rd, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
219
|
Romeo SG, Conti A, Polito F, Tomasello C, Barresi V, La Torre DL, Cucinotta M, Angileri FF, Bartolotta M, Di Giorgio RM, Aguennouz M. miRNA regulation of Sirtuin-1 expression in human astrocytoma. Oncol Lett 2016; 12:2992-2998. [PMID: 27698888 DOI: 10.3892/ol.2016.4960] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 06/08/2016] [Indexed: 12/19/2022] Open
Abstract
Sirtuins are a family of 7 histone deacetylases largely involved in the regulation of cell proliferation, survival and death. The role of sirtuins in tumorigenesis and cancer progression has been previously studied in certain cancer types. Few studies have investigated sirtuin expression in gliomas, with controversial results. The aim of the present study was to investigate the expression of sirtuin-1 (Sirt-1) in diffuse astrocytoma [low grade astrocytoma (LGA)], anaplastic astrocytoma (AA) and glioblastoma multiforme (GBM) and in primary glioma cell lines: PLGAC (primary LGA cells); PAAC (primary AA cells); and PGBMC (primary GBM cells). Tumor samples were obtained from patients who underwent craniotomy for microsurgical tumor resection at the Neurosurgery Unit of the University of Messina between 2011 and 2014. Sirt-1 expression was qualitatively analyzed in 30 human glial tumor samples and 5 non-neoplastic brain tissue (NBT) specimens using immunohistochemistry and western blotting techniques. Sirt-1 expression was quantitatively analyzed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). In addition, Sirt-1 expression in primary cell lines was investigated by immunoblotting and RT-qPCR. Sirt-1 expression was downregulated in gliomas compared to NBTs. Sirt-1 levels also varied among different tumor grades, with more evident downregulation in high-grade (P<0.001) than low-grade tumors (P<0.01). These data were confirmed in cell lines, with the exception of upregulation of protein level in the highest malignancy grade cell lines. The present results suggest a role for miRNA-34a, miRNA-132 and miRNA-217 in the epigenetic control of Sirt-1 during gliomagenesis and progression, and demonstrate the different implications of Sirt-1 in human tissues and cell lines. Furthermore, the present results reveal that Sirt-1 may be an intrinsic regulator of tumor progression and the regulation of Sirt-1 involves complex molecular pathways. However, the biological functions of Sirt-1 in gliomagenesis require additional investigation.
Collapse
Affiliation(s)
| | - Alfredo Conti
- Department of Neurosciences, University of Messina, I-98125 Messina, Italy
| | - Francesca Polito
- Department of Neurosciences, University of Messina, I-98125 Messina, Italy
| | - Chiara Tomasello
- Department of Neurosciences, University of Messina, I-98125 Messina, Italy
| | - Valeria Barresi
- Department of Human Pathology, University of Messina, I-98125 Messina, Italy
| | | | - Maria Cucinotta
- Department of Neurosciences, University of Messina, I-98125 Messina, Italy
| | | | - Marcello Bartolotta
- Department of Human Pathology, University of Messina, I-98125 Messina, Italy
| | | | - M'Hammed Aguennouz
- Department of Neurosciences, University of Messina, I-98125 Messina, Italy
| |
Collapse
|
220
|
Abstract
Effective breast cancer management and decreasing breast cancer fatalities is contingent upon reliable diagnostic procedures and treatment modalities, including those based on ionizing radiation. On the one hand, ionizing radiation is widely used for cancer diagnostics and therapy, on the other hand it is genotoxic cancer-causing agent. Here we discuss recent studies on the effects of low (diagnostic) and high (treatment) doses of ionizing radiation on healthy breast cells, breast cancer cells, and cancer cells resistant to common drug therapies.
Collapse
Affiliation(s)
- Lidia Luzhna
- a Department of Biological Sciences , University of Lethbridge , Lethbridge , AB , Canada
| | - Olga Kovalchuk
- a Department of Biological Sciences , University of Lethbridge , Lethbridge , AB , Canada
| |
Collapse
|
221
|
Cellular Reprogramming Using Defined Factors and MicroRNAs. Stem Cells Int 2016; 2016:7530942. [PMID: 27382371 PMCID: PMC4921148 DOI: 10.1155/2016/7530942] [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] [Received: 12/11/2015] [Revised: 03/08/2016] [Accepted: 04/10/2016] [Indexed: 01/07/2023] Open
Abstract
Development of human bodies, organs, and tissues contains numerous steps of cellular differentiation including an initial zygote, embryonic stem (ES) cells, three germ layers, and multiple expertized lineages of cells. Induced pluripotent stem (iPS) cells have been recently developed using defined reprogramming factors such as Nanog, Klf5, Oct3/4 (Pou5f1), Sox2, and Myc. This outstanding innovation is largely changing life science and medicine. Methods of direct reprogramming of cells into myocytes, neurons, chondrocytes, and osteoblasts have been further developed using modified combination of factors such as N-myc, L-myc, Sox9, and microRNAs in defined cell/tissue culture conditions. Mesenchymal stem cells (MSCs) and dental pulp stem cells (DPSCs) are also emerging multipotent stem cells with particular microRNA expression signatures. It was shown that miRNA-720 had a role in cellular reprogramming through targeting the pluripotency factor Nanog and induction of DNA methyltransferases (DNMTs). This review reports histories, topics, and idea of cellular reprogramming.
Collapse
|
222
|
Hemmatzadeh M, Mohammadi H, Karimi M, Musavishenas MH, Baradaran B. Differential role of microRNAs in the pathogenesis and treatment of Esophageal cancer. Biomed Pharmacother 2016; 82:509-19. [PMID: 27470391 DOI: 10.1016/j.biopha.2016.05.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Esophageal cancer (EC) is the most invasive disease associated with inclusive poor prognosis. EC usually is found as either adenocarcinoma (EAC) or squamous cell carcinomas (ESCC). ESCC forms in squamous cells and highly occurs in the upper third of the esophagus. EAC appears in glandular cells and ordinarily develops in the lower one third of the esophagus near the stomach. Barrett's esophagus (BE) is a metaplastic precursor of EAC. There is a persistent need for improving our understanding of the molecular basis of this disease. MicroRNAs (miRNAs) demonstrate an uncovered class of small, non-coding RNAs that can negatively regulate the protein coding gene, and are associated with approximately all known physiological and pathological processes, especially cancer. MiRNAs can affect cancer pathogenesis, playing a crucial role as either oncogenes or tumor suppressors. The recent emergence of observations on the role of miRNAs in cancer and their functions has induced many investigations to examine their relevance to esophageal cancer. In esophageal cancer, miRNA dysregulation plays a crucial role in cancer prognosis and in patients' responsiveness to neo-adjuvant and adjuvant therapies. In this review, the oncogenic, tumor suppressive, and drug resistance related roles of miRNAs, and their involvement in the pathogenesis and treatment of esophageal cancer were summarized.
Collapse
Affiliation(s)
- Maryam Hemmatzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Tabriz University of Medical Sciences, International Branch (Aras), Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Tabriz University of Medical Sciences, International Branch (Aras), Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Musavishenas
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Tabriz University of Medical Sciences, International Branch (Aras), Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
223
|
Herrera-Pérez Z, Gretz N, Dweep H. A Comprehensive Review on the Genetic Regulation of Cisplatin-induced Nephrotoxicity. Curr Genomics 2016; 17:279-93. [PMID: 27252593 PMCID: PMC4869013 DOI: 10.2174/1389202917666160202220555] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 09/10/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022] Open
Abstract
Cisplatin (CDDP) is a well-known antineoplastic drug which has been extensively utilized over the last decades in the treatment of numerous kinds of tumors. However, CDDP induces a wide range of toxicities in a dose-dependent manner, among which nephrotoxicity is of particular importance. Still, the mechanism of CDDP-induced renal damage is not completely understood; moreover, the knowledge about the role of microRNAs (miRNAs) in the nephrotoxic response is still unknown. miRNAs are known to interact with the representative members of a diverse range of regulatory pathways (including postnatal development, proliferation, inflammation and fibrosis) and pathological conditions, including kidney diseases: polycystic kidney diseases (PKDs), diabetic nephropathy (DN), kidney cancer, and drug-induced kidney injury. In this review, we shed light on the following important aspects: (i) information on genes/proteins and their interactions with previously known pathways engaged with CDDP-induced nephrotoxicity, (ii) information on newly discovered biomarkers, especially, miRNAs for detecting CDDP-induced nephrotoxicity and (iii) information to improve our understanding on CDDP. This information will not only help the researchers belonging to nephrotoxicity field, but also supply an indisputable help for oncologists to better understand and manage the side effects induced by CDDP during cancer treatment. Moreover, we provide up-to-date information about different in vivo and in vitro models that have been utilized over the last decades to study CDDP-induced renal injury. Taken together, this review offers a comprehensive network on genes, miRNAs, pathways and animal models which will serve as a useful resource to understand the molecular mechanism of CDDP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Zeneida Herrera-Pérez
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Harsh Dweep
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
224
|
Ganesan M, Natarajan SK, Zhang J, Mott JL, Poluektova LI, McVicker BL, Kharbanda KK, Tuma DJ, Osna NA. Role of apoptotic hepatocytes in HCV dissemination: regulation by acetaldehyde. Am J Physiol Gastrointest Liver Physiol 2016; 310:G930-G940. [PMID: 27056722 PMCID: PMC6842882 DOI: 10.1152/ajpgi.00021.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/31/2016] [Indexed: 02/08/2023]
Abstract
Alcohol consumption exacerbates hepatitis C virus (HCV) pathogenesis and promotes disease progression, although the mechanisms are not quite clear. We have previously observed that acetaldehyde (Ach) continuously produced by the acetaldehyde-generating system (AGS), temporarily enhanced HCV RNA levels, followed by a decrease to normal or lower levels, which corresponded to apoptosis induction. Here, we studied whether Ach-induced apoptosis caused depletion of HCV-infected cells and what role apoptotic bodies (AB) play in HCV-alcohol crosstalk. In liver cells exposed to AGS, we observed the induction of miR-122 and miR-34a. As miR-34a has been associated with apoptotic signaling and miR-122 with HCV replication, these findings may suggest that cells with intensive viral replication undergo apoptosis. Furthermore, when AGS-induced apoptosis was blocked by a pan-caspase inhibitor, the expression of HCV RNA was not changed. AB from HCV-infected cells contained HCV core protein and the assembled HCV particle that infect intact hepatocytes, thereby promoting the spread of infection. In addition, AB are captured by macrophages to switch their cytokine profile to the proinflammatory one. Macrophages exposed to HCV(+) AB expressed more IL-1β, IL-18, IL-6, and IL-10 mRNAs compared with those exposed to HCV(-) AB. The generation of AB from AGS-treated HCV-infected cells even enhanced the induction of aforementioned cytokines. We conclude that HCV and alcohol metabolites trigger the formation of AB containing HCV particles. The consequent spread of HCV to neighboring hepatocytes via infected AB, as well as the induction of liver inflammation by AB-mediated macrophage activation potentially exacerbate the HCV infection course by alcohol and worsen disease progression.
Collapse
Affiliation(s)
- Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sathish Kumar Natarajan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jinjin Zhang
- School of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Justin L Mott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | | | - Benita L McVicker
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Dean J Tuma
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Natalia A Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska;
| |
Collapse
|
225
|
Ofek P, Calderón M, Mehrabadi FS, Krivitsky A, Ferber S, Tiram G, Yerushalmi N, Kredo-Russo S, Grossman R, Ram Z, Haag R, Satchi-Fainaro R. Restoring the oncosuppressor activity of microRNA-34a in glioblastoma using a polyglycerol-based polyplex. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2201-2214. [PMID: 27262933 PMCID: PMC5364374 DOI: 10.1016/j.nano.2016.05.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/04/2016] [Accepted: 05/22/2016] [Indexed: 12/19/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary neoplasm of the brain. Poor prognosis is mainly attributed to tumor heterogeneity, invasiveness, and drug resistance. microRNA-based therapeutics represent a promising approach due to their ability to inhibit multiple targets. In this work, we aim to restore the oncosuppressor activity of microRNA-34a (miR-34a) in GBM. We developed a cationic carrier system, dendritic polyglycerolamine (dPG-NH2), which remarkably improves miRNA stability, intracellular trafficking, and activity. dPG-NH2 carrying mature miR-34a targets C-MET, CDK6, Notch1 and BCL-2, consequently inhibiting cell cycle progression, proliferation and migration of GBM cells. Following complexation with dPG-NH2, miRNA is stable in plasma and able to cross the blood–brain barrier. We further show inhibition of tumor growth following treatment with dPG-NH2–miR-34a in a human glioblastoma mouse model. We hereby present a promising technology using dPG-NH2–miR-34a polyplex for brain-tumor treatment, with enhanced efficacy and no apparent signs of toxicity.
Collapse
Affiliation(s)
- Paula Ofek
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo Calderón
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Adva Krivitsky
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
| | - Shiran Ferber
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
| | | | | | - Rachel Grossman
- Department of Neurosurgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Zvi Ram
- Department of Neurosurgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
226
|
Abstract
It is now clear that functional p53 is critical to protect the genome from alterations that lead to tumorigenesis. However, with the myriad of cellular stresses and pathways linked to p53 activation, much remains unknown about how p53 maintains genome stability and the proteins involved. The current understanding of the multiple ways p53 contributes to genome stability and how two of its negative regulators, Mdm2 and Mdmx, induce genome instability will be described.
Collapse
Affiliation(s)
- Christine M Eischen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37212
| |
Collapse
|
227
|
Beckerman R, Yoh K, Mattia-Sansobrino M, Zupnick A, Laptenko O, Karni-Schmidt O, Ahn J, Byeon IJ, Keezer S, Prives C. Lysines in the tetramerization domain of p53 selectively modulate G1 arrest. Cell Cycle 2016; 15:1425-38. [PMID: 27210019 DOI: 10.1080/15384101.2016.1170270] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Functional in a tetrameric state, the protein product of the p53 tumor suppressor gene confers its tumor-suppressive activity by transactivating genes which promote cell-cycle arrest, senescence, or programmed cell death. How p53 distinguishes between these divergent outcomes is still a matter of considerable interest. Here we discuss the impact of 2 mutations in the tetramerization domain that confer unique properties onto p53. By changing lysines 351 and 357 to arginine, thereby blocking all post-translational modifications of these residues, DNA binding and transcriptional regulation by p53 remain virtually unchanged. On the other hand, by changing these lysines to glutamine (2KQ-p53), thereby neutralizing their positive charge and potentially mimicking acetylation, p53 is impaired in the induction of cell cycle arrest and yet can still effectively induce cell death. Surprisingly, when 2KQ-p53 is expressed at high levels in H1299 cells, it can bind to and transactivate numerous p53 target genes including p21, but not others such as miR-34a and cyclin G1 to the same extent as wild-type p53. Our findings show that strong induction of p21 is not sufficient to block H1299 cells in G1, and imply that modification of one or both of the lysines within the tetramerization domain may serve as a mechanism to shunt p53 from inducing cell cycle arrest.
Collapse
Affiliation(s)
| | - Kathryn Yoh
- a Department of Biological Sciences , Columbia University , New York , NY , USA
| | | | | | - Oleg Laptenko
- a Department of Biological Sciences , Columbia University , New York , NY , USA
| | - Orit Karni-Schmidt
- a Department of Biological Sciences , Columbia University , New York , NY , USA
| | - Jinwoo Ahn
- b Department of Structural Biology , University of Pittsburgh , Pittsburgh , PA , USA
| | - In-Ja Byeon
- b Department of Structural Biology , University of Pittsburgh , Pittsburgh , PA , USA
| | - Susan Keezer
- c Cell Signaling Technology, Inc. , Danvers , MA , USA
| | - Carol Prives
- a Department of Biological Sciences , Columbia University , New York , NY , USA
| |
Collapse
|
228
|
p53 directly activates cystatin D/CST5 to mediate mesenchymal-epithelial transition: a possible link to tumor suppression by vitamin D3. Oncotarget 2016; 6:15842-56. [PMID: 26158294 PMCID: PMC4599241 DOI: 10.18632/oncotarget.4683] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/10/2015] [Indexed: 12/11/2022] Open
Abstract
Cystatin D (CST5) encodes an inhibitor of cysteine proteases of the cathepsin family and is directly induced by the vitamin D receptor (VDR). Interestingly, vitamin D3 exerts tumor suppressive effects in a variety of tumor types. In colorectal cancer (CRC) cells CST5 was shown to mediate mesenchymal-epithelial transition (MET). Interestingly, vitamin D3 was shown to exert tumor suppressive effects in a variety of tumor types, including colorectal cancer (CRC). We recently performed an integrated genomic and proteomic screen to identify targets of the p53 tumor suppressor in CRC cells. Thereby, we identified CST5 as a putative p53 target gene. Here, we validated and characterized CST5 as a direct p53 target gene. After activation of a conditional p53 allele, CST5 was upregulated on mRNA and protein levels. Treatment with nutlin-3a or etoposide induced CST5 in a p53-dependent manner. These regulations were direct, since ectopic and endogenous p53 occupied a conserved binding site in the CST5 promoter region. In addition, treatment with calcitriol, the active vitamin D3 metabolite, and simultaneous activation of p53 resulted in enhanced CST5 induction and increased repression of SNAIL, an epithelial-mesenchymal transition (EMT) inducing transcription factor. Furthermore, CST5 inactivation decreased p53-induced mesenchymal-epithelial transition (MET) as evidenced by decreased inhibition of SNAIL and of migration by p53. Furthermore, CST5 expression was directly repressed by SNAIL. In summary, these results imply CST5 as an important mediator of tumor suppression by p53 in colorectal cancer. In addition, they suggest that a combined treatment activating p53 and the vitamin D3 pathway may function via induction of CST5.
Collapse
|
229
|
Wu CW, Peng ML, Yeh KT, Tsai YY, Chiang CC, Cheng YW. Inactivation of p53 in pterygium influence miR-200a expression resulting in ZEB1/ZEB2 up-regulation and EMT processing. Exp Eye Res 2016; 146:206-211. [DOI: 10.1016/j.exer.2016.03.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/10/2016] [Accepted: 03/13/2016] [Indexed: 01/07/2023]
|
230
|
Abstract
Tumor suppresser gene TP53 is one of the most frequently deleted
or mutated genes in gastrointestinal cancers. As a transcription factor, p53
regulates a number of important protein coding genes to control cell cycle, cell
death, DNA damage/repair, stemness, differentiation and other key cellular
functions. In addition, p53 is also able to activate the expression of a number
of small non-coding microRNAs (miRNAs) through direct binding to the promoter
region of these miRNAs. Many miRNAs have been identified to be potential tumor
suppressors by regulating key effecter target mRNAs. Our understanding of the
regulatory network of p53 has recently expanded to include long non-coding RNAs
(lncRNAs). Like miRNA, lncRNAs have been found to play important roles in cancer
biology. With our increased understanding of the important functions of these
non-coding RNAs and their relationship with p53, we are gaining exciting new
insights into the biology and function of cells in response to various growth
environment changes. In this review we summarize the current understanding of
the ever expanding involvement of non-coding RNAs in the p53 regulatory network
and its implications for our understanding of gastrointestinal cancer.
Collapse
Affiliation(s)
- Andrew Fesler
- Translational Research Laboratory, Department of Pathology, Stony Brook University, Stony Brook, USA
| | - Ning Zhang
- Department of Pharmacy, Dalian Medical University, Dalian, China
| | - Jingfang Ju
- Translational Research Laboratory, Department of Pathology, Stony Brook University, Stony Brook, USA
| |
Collapse
|
231
|
MicroRNA: a connecting road between apoptosis and cholesterol metabolism. Tumour Biol 2016; 37:8529-54. [PMID: 27105614 DOI: 10.1007/s13277-016-4988-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/10/2016] [Indexed: 12/15/2022] Open
Abstract
Resistance to apoptosis leads to tumorigenesis and failure of anti-cancer therapy. Recent studies also highlight abrogated lipid/cholesterol metabolism as one of the root causes of cancer that can lead to metastatic transformations. Cancer cells are dependent on tremendous supply of cellular cholesterol for the formation of new membranes and continuation of cell signaling. Cholesterol homeostasis network tightly regulates this metabolic need of cancer cells on cholesterol and other lipids. Genetic landscape is also shared between apoptosis and cholesterol metabolism. MicroRNAs (miRNAs) are the new fine tuners of signaling pathways and cellular processes and are known for their ability to post-transcriptionally repress gene expression in a targeted manner. This review summarizes the current knowledge about the cross talk between apoptosis and cholesterol metabolism via miRNAs. In addition, we also emphasize herein recent therapeutic modulations of specific miRNAs and their promising potential for the treatment of deadly diseases including cancer and cholesterol related pathologies. Understanding of the impact of miRNA-based regulation of apoptosis and metabolic processes is still at its dawn and needs further research for the development of future miRNA-based therapies. As both these physiological processes affect cellular homeostasis, we believe that this comprehensive summary of miRNAs modulating both apoptosis and cholesterol metabolism will open uncharted territory for scientific exploration and will provide the foundation for discovering novel drug targets for cancer and metabolic diseases.
Collapse
|
232
|
Ward CM, Li B, Pace BS. Original Research: Stable expression of miR-34a mediates fetal hemoglobin induction in K562 cells. Exp Biol Med (Maywood) 2016; 241:719-29. [PMID: 26940952 PMCID: PMC4950382 DOI: 10.1177/1535370216636725] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sickle cell anemia is a common genetic disorder caused by a point mutation in the sixth codon of the β-globin gene affecting people of African descent worldwide. A wide variety of clinical phenotypes ranging from mild to severe symptoms and complications occur due to hemoglobin S polymerization, red blood cell sickling, and vaso-occlusion. Research efforts are ongoing to develop strategies of fetal hemoglobin (HbF; α2γ2) induction to inhibit sickle hemoglobin polymerization and improve clinical outcomes. Insights have been gained from investigating mutations in the β-globin locus or transcription factors involved in the mechanisms of hemoglobin switching. Recent efforts to expand molecular targets that modulate γ-globin expression involve microRNAs that work through posttranscriptional gene regulation. Therefore, the goal of our study was to identify novel microRNA genes involved in fetal hemoglobin expression. Using in silico analysis, we identified a miR-34a binding site in the γ-globin mRNA which was tested for functional relevance. Stable expression of the shMIMIC miR-34a lentivirus vector increased fetal hemoglobin levels in single cell K562 clones consistent with silencing of a γ-globin gene repressor. Furthermore, miR-34a promoted cell differentiation supported by increased expression of KLF1, glycophorin A, and the erythropoietin receptor. Western blot analysis of known negative regulators of γ-globin including YY1, histone deacetylase 1, and STAT3, which are regulated by miR-34a showed no change in YY1 and histone deacetylase 1 levels; however, total- and phosphorylated-STAT3 levels were decreased in single cell miR-34a K562 clones. These data support a mechanism of fetal hemoglobin activation by miR-34a involving STAT3 gene silencing.
Collapse
Affiliation(s)
- Christina M Ward
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| | - Biaoru Li
- Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| | - Betty S Pace
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
233
|
Abstract
B-cell chronic lymphocytic leukemia (CLL) is the most common adult human leukemia. Although, the molecular alterations leading to CLL onset and progression are still under investigation (specifically, the interplay and exact role of oncogenes and tumor suppressors in CLL pathogenesis). MicroRNAs are small non-coding RNAs that regulate gene expression and are expressed in a tissue specific manner. Deregulation of microRNAs can alter expression levels of genes involved in the development and/or progression of tumors. In CLL, microRNAs can function as oncogenes or tumor suppressors. Here, we review the most recent findings on the role of microRNAs in the onset/progression of CLL, and how this knowledge can be used to identify new biomarkers and targets to treat this leukemia.
Collapse
Affiliation(s)
- Veronica Balatti
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Mario Acunzo
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Yuri Pekarky
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Carlo M Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
234
|
Chen J. The Cell-Cycle Arrest and Apoptotic Functions of p53 in Tumor Initiation and Progression. Cold Spring Harb Perspect Med 2016; 6:a026104. [PMID: 26931810 DOI: 10.1101/cshperspect.a026104] [Citation(s) in RCA: 788] [Impact Index Per Article: 87.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
P53 is a transcription factor highly inducible by many stress signals such as DNA damage, oncogene activation, and nutrient deprivation. Cell-cycle arrest and apoptosis are the most prominent outcomes of p53 activation. Many studies showed that p53 cell-cycle and apoptosis functions are important for preventing tumor development. p53 also regulates many cellular processes including metabolism, antioxidant response, and DNA repair. Emerging evidence suggests that these noncanonical p53 activities may also have potent antitumor effects within certain context. This review focuses on the cell-cycle arrest and apoptosis functions of p53, their roles in tumor suppression, and the regulation of cell fate decision after p53 activation.
Collapse
Affiliation(s)
- Jiandong Chen
- Molecular Oncology Department, Moffitt Cancer Center, Tampa, Florida 33612
| |
Collapse
|
235
|
Flubendazole, FDA-approved anthelmintic, targets breast cancer stem-like cells. Oncotarget 2016; 6:6326-40. [PMID: 25811972 PMCID: PMC4467440 DOI: 10.18632/oncotarget.3436] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 01/05/2015] [Indexed: 01/07/2023] Open
Abstract
Cancer stem-like cell (CS-like cell) is considered to be responsible for recurrence and drug resistance events in breast cancer, which makes it a potential target for novel cancer therapeutic strategy. The FDA approved flubendazole, has been widely used in the treatment of intestinal parasites. Here, we demonstrated a novel effect of flubendazole on breast CS-like cells. Flubendazole inhibited breast cancer cells proliferation in dose- and time-dependent manner and delayed tumor growth in xenograft models by intraperitoneal injection. Importantly, flubendazole reduced CD44high/CD24low subpopulation and suppressed the formation of mammosphere and the expression of self-renewal related genes including c-myc, oct4, sox2, nanog and cyclinD1. Moreover, we found that flubendazole induced cell differentiation and inhibited cell migration. Consistently, flubendazole reduced mesenchymal markers (β-catenin, N-cadherin and Vimentin) expression and induced epithelial and differentiation marker (Keratin 18) expression in breast cancer cells. Mechanism study revealed that flubendazole arrested cell cycle at G2/M phase and induced monopolar spindle formation through inhibiting tubulin polymerization. Furthermore, flubendazole enhanced cytotoxic activity of conventional therapeutic drugs fluorouracil and doxorubicin against breast cancer cells. In conclusion, our findings uncovered a remarkable effect of flubendazole on suppressing breast CS-like cells, indicating a novel utilization of flubendazole in breast cancer therapy.
Collapse
|
236
|
Schmid G, Notaro S, Reimer D, Abdel-Azim S, Duggan-Peer M, Holly J, Fiegl H, Rössler J, Wiedemair A, Concin N, Altevogt P, Marth C, Zeimet AG. Expression and promotor hypermethylation of miR-34a in the various histological subtypes of ovarian cancer. BMC Cancer 2016; 16:102. [PMID: 26879132 PMCID: PMC4754861 DOI: 10.1186/s12885-016-2135-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 02/07/2016] [Indexed: 01/07/2023] Open
Abstract
Background An increasing body of evidence shows that miR-34 family has tumor suppressive properties mediating apoptosis, cell cycle arrest and senescence. In ovarian cancer, miR34 family members were found to be under expressed. Particularly miR-34a has been revealed to be a direct transcriptional target of p53 which is frequently mutated in epithelial ovarian carcinomas especially in high grade serous cancer. Moreover, methylation of miR-34a CpG Islands was found to down-regulate miR-34a expression. The aim of this study was to investigate the clinical relevance of mir34a as well as its promoter methylation in a subset of 133 ovarian cancers with a special focus on the p53 mutation status, the dualistic type I and type II ovarian cancer model and the different histotypes. Methods One hundred thirty-three epithelial ovarian cancers and 8 samples of healthy ovarian surface epithelium were retrospectively analysed for miR-34a expression with quantitative real-time reverse transcription PCR (qRT-PCR). Gene-specific DNA methylation was evaluated with MethyLight technique. Results Significantly lower miR-34a expression was found in ovarian cancers than in healthy ovarian epithelium (p = 0.002). The expression of miR-34a was found lower in type II than in type I cancers (p = 0.037), in p53 mutated as compared to p53 wild type cancers (p = 0.003) and in high grade compared to in low grade cancers (p = 0.028). In multivariate COX regression model low expressing miR-34a cancers exhibited a reduced PFS (p = 0.039) and OS (p = 0.018). In serous cancers low miR-34a levels showed a worse OS confirmed also in multivariate analysis (p = 0.022). miR-34a promoter methylation was found higher in type II cancers than in type I (p = 0.006). mir34a expression and promoter methylation showed an inverse correlation in cancer samples (p = 0.05). Conclusion We demonstrated a clinical independent role of miR-34a in epithelial ovarian cancers. Moreover, we corroborated the correlation between miR-34a expression and its promoter methylation in a large set of ovarian cancers. The inverse association between miR-34a expression and grading, p53 mutation status and dualistic tumor type classification, together with its prognostic relevance may underline the tumor-suppressive character of miR-34a in ovarian cancer.
Collapse
Affiliation(s)
- Gabriel Schmid
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Sara Notaro
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria. .,Department of Gynecology and Obstetrics, University of Brescia, Piazza Spedali Civili 1, 25123, Brescia, Italy.
| | - Daniel Reimer
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Samira Abdel-Azim
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Michaela Duggan-Peer
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Jessica Holly
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Heidi Fiegl
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Julia Rössler
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Annemarie Wiedemair
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Nicole Concin
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Christian Marth
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Alain Gustave Zeimet
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
237
|
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
|
238
|
Cabrita MA, Vanzyl EJ, Hamill JD, Pan E, Marcellus KA, Tolls VJ, Alonzi RC, Pastic A, Rambo TME, Sayed H, McKay BC. A Temperature Sensitive Variant of p53 Drives p53-Dependent MicroRNA Expression without Evidence of Widespread Post-Transcriptional Gene Silencing. PLoS One 2016; 11:e0148529. [PMID: 26840126 PMCID: PMC4739602 DOI: 10.1371/journal.pone.0148529] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/19/2016] [Indexed: 01/07/2023] Open
Abstract
The p53 tumour suppressor is a transcription factor that can regulate the expression of numerous genes including many encoding proteins and microRNAs (miRNAs). The predominant outcomes of a typical p53 response are the initiation of apoptotic cascades and the activation of cell cycle checkpoints. HT29-tsp53 cells express a temperature sensitive variant of p53 and in the absence of exogenous DNA damage, these cells preferentially undergo G1 phase cell cycle arrest at the permissive temperature that correlates with increased expression of the cyclin-dependent kinase inhibitor p21WAF1. Recent evidence also suggests that a variety of miRNAs can induce G1 arrest by inhibiting the expression of proteins like CDK4 and CDK6. Here we used oligonucleotide microarrays to identify p53-regulated miRNAs that are induced in these cells undergoing G1 arrest. At the permissive temperature, the expression of several miRNAs was increased through a combination of either transcriptional or post-transcriptional regulation. In particular, miR-34a-5p, miR-143-3p and miR-145-5p were strongly induced and they reached levels comparable to that of reference miRNAs (miR-191 and miR-103). Importantly, miR-34a-5p and miR-145-5p are known to silence the Cdk4 and/or Cdk6 G1 cyclin-dependent kinases (cdks). Surprisingly, there was no p53-dependent decrease in the expression of either of these G1 cdks. To search for other potential targets of p53-regulated miRNAs, p53-downregulated mRNAs were identified through parallel microarray analysis of mRNA expression. Once again, there was no clear effect of p53 on the repression of mRNAs under these conditions despite a remarkable increase in p53-induced mRNA expression. Therefore, despite a strong p53 transcriptional response, there was no clear evidence that p53-responsive miRNA contributed to gene silencing. Taken together, the changes in cell cycle distribution in this cell line at the permissive temperature is likely attributable to transcriptional upregulation of the CDKN1A mRNA and p21WAF1 protein and not to the down regulation of CDK4 or CDK6 by p53-regulated miRNAs.
Collapse
Affiliation(s)
- Miguel A. Cabrita
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa ON
| | | | - Jeff D. Hamill
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa ON
| | - Elysia Pan
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa ON
| | | | | | - Rhea C. Alonzi
- Institute for Biochemistry, Carleton University, Ottawa, ON
| | - Alyssa Pastic
- Department of Biology, Carleton University, Ottawa, ON
| | | | - Hadil Sayed
- Department of Biology, Carleton University, Ottawa, ON
| | - Bruce C. McKay
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa ON
- Department of Biology, Carleton University, Ottawa, ON
- Institute for Biochemistry, Carleton University, Ottawa, ON
- * E-mail:
| |
Collapse
|
239
|
Zhou J, Ju WQ, Yuan XP, Zhu XF, Wang DP, He XS. miR-26a regulates mouse hepatocyte proliferation via directly targeting the 3' untranslated region of CCND2 and CCNE2. Hepatobiliary Pancreat Dis Int 2016; 15:65-72. [PMID: 26818545 DOI: 10.1016/s1499-3872(15)60383-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The deficiency of liver regeneration needs to be addressed in the fields of liver surgery, split liver transplantation and living donor liver transplantation. Researches of microRNAs would broaden our understandings on the mechanisms of various diseases. Our previous research confirmed that miR-26a regulated liver regeneration in mice; however, the relationship between miR-26a and its target, directly or indirectly, remains unclear. Therefore, the present study further investigated the mechanism of miR-26a in regulating mouse hepatocyte proliferation. METHODS An established mouse liver cell line, Nctc-1469, was transfected with Ad5-miR-26a-EGFP, Ad5-anti-miR-26a-EGFP or Ad5-EGFP vector. Cell proliferation was assessed by MTS, cell apoptosis and cell cycle by flow cytometry, and gene expression by Western blotting and quantitative real-time PCR. Dual-luciferase reporter assays were used to test targets of miR-26a. RESULTS Compared with the Ad5-EGFP group, Ad5-anti-miR-26a-EGFP down-regulated miR-26a and increased proliferation of hepatocytes, with more cells entering the G1 phase of cell cycle (82.70%+/-1.45% vs 75.80%+/-3.92%), and decreased apoptosis (5.50%+/-0.35% vs 6.73%+/-0.42%). CCND2 and CCNE2 were the direct targeted genes of miR-26a. miR-26a down-regulation up-regulated CCND2 and CCNE2 expressions and down-regulated p53 expression in Nctc-1469 cells. On the contrary, miR-26a over-expression showed the opposite results. CONCLUSIONS miR-26a regulated mouse hepatocyte proliferation by directly targeting the 3' untranslated regions of cyclin D2/cyclin E2; miR-26a also regulated p53-mediated apoptosis. Our data suggested that miR-26a may be a promising regulator in liver regeneration.
Collapse
Affiliation(s)
- Jian Zhou
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | | | | | | | | | | |
Collapse
|
240
|
Pang J, Xiong H, Yang H, Ou Y, Xu Y, Huang Q, Lai L, Chen S, Zhang Z, Cai Y, Zheng Y. Circulating miR-34a levels correlate with age-related hearing loss in mice and humans. Exp Gerontol 2016; 76:58-67. [PMID: 26802970 DOI: 10.1016/j.exger.2016.01.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/23/2015] [Accepted: 01/19/2016] [Indexed: 12/21/2022]
Abstract
Age-related hearing loss (AHL) is a progressive neurodegenerative disease that is largely silent in its initial stages. There is no sensitive blood biomarker for diagnosis or early detection of AHL. MicroRNAs (miRNAs or miRs) are abundant and highly stable in blood, and have been recently described as powerful circulating biomarkers in a wide range of diseases. In the present study, we identified concordant increases in miR-34a levels in the cochlea, auditory cortex, and plasma of C57BL/6 mice during aging. These increases were accompanied by elevated hearing thresholds and greater loss of hair cells. Levels of miR-34a targets, silent information regulator 1 (SIRT1), B-cell lymphoma-2 (Bcl-2), and E2F transcription factor 3 (E2F3), in the cochlea, auditory cortex, and plasma decreased with aging inversely to miR-34a. Moreover, plasma miR-34a levels were significantly higher in patients with AHL compared with controls who had normal hearing and had a receiver-operating characteristic curve that distinguished AHL patients from controls. However, SIRT1, Bcl-2, and E2F3 showed no correlation with AHL in humans. In summary, circulating miR-34a level may potentially serve as a useful biomarker for early detection of AHL.
Collapse
Affiliation(s)
- Jiaqi Pang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Hao Xiong
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Haidi Yang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Yongkang Ou
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Yaodong Xu
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Qiuhong Huang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Lan Lai
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Suijun Chen
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Zhigang Zhang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Yuexin Cai
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | - Yiqing Zheng
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China.
| |
Collapse
|
241
|
Li D, Li C, Xu Y, Xu D, Li H, Gao L, Chen S, Fu L, Xu X, Liu Y, Zhang X, Zhang J, Ming H, Zheng L. Differential Expression of microRNAs in the Ovaries from Letrozole-Induced Rat Model of Polycystic Ovary Syndrome. DNA Cell Biol 2016; 35:177-83. [PMID: 26745201 DOI: 10.1089/dna.2015.3145] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex and heterogeneous endocrine disorder. To understand the pathogenesis of PCOS, we established rat models of PCOS induced by letrozole and employed deep sequencing to screen the differential expression of microRNAs (miRNAs) in PCOS rats and control rats. We observed vaginal smear and detected ovarian pathological alteration and hormone level changes in PCOS rats. Deep sequencing showed that a total of 129 miRNAs were differentially expressed in the ovaries from letrozole-induced rat model compared with the control, including 49 miRNAs upregulated and 80 miRNAs downregulated. Furthermore, the differential expression of miR-201-5p, miR-34b-5p, miR-141-3p, and miR-200a-3p were confirmed by real-time polymerase chain reaction. Bioinformatic analysis revealed that these four miRNAs were predicted to target a large set of genes with different functions. Pathway analysis supported that the miRNAs regulate oocyte meiosis, mitogen-activated protein kinase (MAPK) signaling, phosphoinositide 3-kinase/Akt (PI3K-Akt) signaling, Rap1 signaling, and Notch signaling. These data indicate that miRNAs are differentially expressed in rat PCOS model and the differentially expressed miRNA are involved in the etiology and pathophysiology of PCOS. Our findings will help identify miRNAs as novel diagnostic markers and therapeutic targets for PCOS.
Collapse
Affiliation(s)
- Dandan Li
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China .,2 Department of Gynecological Oncology, Tumor Hospital of Jilin Province , Changchun, People's Republic of China
| | - Chunjin Li
- 3 College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University , Changchun, People's Republic of China
| | - Ying Xu
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China
| | - Duo Xu
- 4 Department of Breast Oncology, Tumor Hospital of Jilin Province , Changchun, People's Republic of China
| | - Hongjiao Li
- 3 College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University , Changchun, People's Republic of China
| | - Liwei Gao
- 5 Department of Gynecology and Obstetrics, Zhengzhou Central Hospital Affiliated to Zhengzhou University , Zhengzhou, People's Republic of China
| | - Shuxiong Chen
- 3 College of Animal Sciences, Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University , Changchun, People's Republic of China
| | - Lulu Fu
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China
| | - Xin Xu
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China
| | - Yongzheng Liu
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China
| | - Xueying Zhang
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China
| | - Jingshun Zhang
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China
| | - Hao Ming
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China
| | - Lianwen Zheng
- 1 Reproductive Medical Center, The Second Hospital of Jilin University , Changchun, People's Republic of China
| |
Collapse
|
242
|
Donzelli S, Cioce M, Muti P, Strano S, Yarden Y, Blandino G. MicroRNAs: Non-coding fine tuners of receptor tyrosine kinase signalling in cancer. Semin Cell Dev Biol 2016; 50:133-42. [PMID: 26773212 DOI: 10.1016/j.semcdb.2015.12.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/23/2015] [Indexed: 12/19/2022]
Abstract
Emerging evidence point to a crucial role for non-coding RNAs in modulating homeostatic signaling under physiological and pathological conditions. MicroRNAs, the best-characterized non-coding RNAs to date, can exquisitely integrate spatial and temporal signals in complex networks, thereby confer specificity and sensitivity to tissue response to changes in the microenvironment. MicroRNAs appear as preferential partners for Receptor Tyrosine Kinases (RTKs) in mediating signaling under stress conditions. Stress signaling can be especially relevant to disease. Here we focus on the ability of microRNAs to mediate RTK signaling in cancer, by acting as both tumor suppressors and oncogenes. We will provide a few general examples of microRNAs modulating specific tumorigenic functions downstream of RTK signaling and integrate oncogenic signals from multiple RTKs. A special focus will be devoted to epidermal growth factor receptor (EGFR) signaling, a system offering relatively rich information. We will explore the role of selected microRNAs as bidirectional modulators of EGFR functions in cancer cells. In addition, we will present the emerging evidence for microRNAs being specifically modulated by oncogenic EGFR mutants and we will discuss how this impinges on EGFRmut driven chemoresistance, which fits into the tumor heterogeneity-driven cancer progression. Finally, we discuss how other non-coding RNA species are emerging as important modulators of cancer progression and why the scenario depicted herein is destined to become increasingly complex in the future.
Collapse
Affiliation(s)
- Sara Donzelli
- Translational Oncogenomics, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Mario Cioce
- Translational Oncogenomics, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Paola Muti
- Dept of Oncology, McMaster University, Hamilton, On L8V1C3, Canada
| | - Sabrina Strano
- Molecular Chemoprevention Units, Regina Elena National Cancer Institute, 00144 Rome, Italy; Dept of Oncology, McMaster University, Hamilton, On L8V1C3, Canada
| | - Yosef Yarden
- Dept of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Giovanni Blandino
- Translational Oncogenomics, Regina Elena National Cancer Institute, 00144 Rome, Italy; Dept of Oncology, McMaster University, Hamilton, On L8V1C3, Canada.
| |
Collapse
|
243
|
Ghose J, Bhattacharyya NP. Transcriptional regulation of microRNA-100, -146a, and -150 genes by p53 and NFκB p65/RelA in mouse striatal STHdh(Q7)/ Hdh(Q7) cells and human cervical carcinoma HeLa cells. RNA Biol 2016; 12:457-77. [PMID: 25757558 DOI: 10.1080/15476286.2015.1014288] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
MicroRNA (miRNA) genes generally share many features common to those of protein coding genes. Various transcription factors (TFs) and co-regulators are also known to regulate miRNA genes. Here we identify novel p53 and NFκB p65/RelA responsive miRNAs and demonstrate that these 2 TFs bind to the regulatory sequences of miR-100, -146a and -150 in both mouse striatal and human cervical carcinoma cells and regulate their expression. p53 represses the miRNAs while NFκB p65/RelA induces them. Further, we provide evidence that exogenous p53 inhibits NFκB p65/RelA activity by reducing its nuclear content and competing with it for CBP binding. This suggests for the existence of a functional cross-talk between the 2 TFs in regulating miRNA expression. Moreover, promoter occupancy assay reveals that exogenous p53 excludes NFκB p65/RelA from its binding site in the upstream sequence of miR-100 gene thereby causing its repression. Thus, our work identifies novel p53 and NFκB p65/RelA responsive miRNAs in human and mouse and uncovers possible mechanisms of co-regulation of miR-100. It is to be mentioned here that cross-talks between p53 and NFκB p65/RelA have been observed to define the outcome of several biological processes and that the pro-apoptotic effect of p53 and the pro-survival functions of NFκB can be largely mediated via the biological roles of the miRNAs these TFs regulate. Our observation with cell lines thus provides an important platform upon which further work is to be done to establish the biological significance of such co-regulation of miRNAs by p53 and NFκB p65/RelA.
Collapse
Key Words
- ChIP, Chromatin immunoprecipitation
- Co-IP, Co-immunoprecipitation
- NFκB p65/RelA
- NFκB, nuclear factor kappa-light-chain-enhancer of activated B cells
- RLU, Relative light unit
- RNA POL II, RNA Polymerase II
- RNA POL III, RNA Polymerase III
- RT-PCR, Reverse transcription polymerase chain reaction
- TF, Transcriptional factor
- TFBS
- Transcription factor binding site
- WB, Western blot
- miR-100
- miR-146a
- miR-150
- miRNA gene regulation
- miRNAs, microRNAs
- microRNA
- p53
- p53, tumor protein 53
- p65, RELA, RELA
- transcription factor
- v-rel avian reticuloendotheliosis viral oncogene homolog A
Collapse
Affiliation(s)
- Jayeeta Ghose
- a Crystallography and Molecular Biology Division; Saha Institute of Nuclear Physics ; Bidhannagar, Kolkata , India
| | | |
Collapse
|
244
|
Kaller M, Hermeking H. Interplay Between Transcription Factors and MicroRNAs Regulating Epithelial-Mesenchymal Transitions in Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 937:71-92. [DOI: 10.1007/978-3-319-42059-2_4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
245
|
Adams BD, Wali VB, Cheng CJ, Inukai S, Booth CJ, Agarwal S, Rimm DL, Győrffy B, Santarpia L, Pusztai L, Saltzman WM, Slack FJ. miR-34a Silences c-SRC to Attenuate Tumor Growth in Triple-Negative Breast Cancer. Cancer Res 2015; 76:927-39. [PMID: 26676753 DOI: 10.1158/0008-5472.can-15-2321] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/09/2015] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype with no clinically proven biologically targeted treatment options. The molecular heterogeneity of TNBC and lack of high frequency driver mutations other than TP53 have hindered the development of new and effective therapies that significantly improve patient outcomes. miRNAs, global regulators of survival and proliferation pathways important in tumor development and maintenance, are becoming promising therapeutic agents. We performed miRNA-profiling studies in different TNBC subtypes to identify miRNAs that significantly contribute to disease progression. We found that miR-34a was lost in TNBC, specifically within mesenchymal and mesenchymal stem cell-like subtypes, whereas expression of miR-34a targets was significantly enriched. Furthermore, restoration of miR-34a in cell lines representing these subtypes inhibited proliferation and invasion, activated senescence, and promoted sensitivity to dasatinib by targeting the proto-oncogene c-SRC. Notably, SRC depletion in TNBC cell lines phenocopied the effects of miR-34a reintroduction, whereas SRC overexpression rescued the antitumorigenic properties mediated by miR-34a. miR-34a levels also increased when cells were treated with c-SRC inhibitors, suggesting a negative feedback exists between miR-34a and c-SRC. Moreover, miR-34a administration significantly delayed tumor growth of subcutaneously and orthotopically implanted tumors in nude mice, and was accompanied by c-SRC downregulation. Finally, we found that miR-34a and SRC levels were inversely correlated in human tumor specimens. Together, our results demonstrate that miR-34a exerts potent antitumorigenic effects in vitro and in vivo and suggests that miR-34a replacement therapy, which is currently being tested in human clinical trials, represents a promising therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Brian D Adams
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut
| | - Vikram B Wali
- Yale Cancer Center Genetics and Genomics Program, Yale University School of Medicine, New Haven, Connecticut
| | - Christopher J Cheng
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut. Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Sachi Inukai
- Institute for RNA Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Carmen J Booth
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Seema Agarwal
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Balázs Győrffy
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary. MTA TTK Lendület Cancer Biomarker Research Group, Budapest, Hungary
| | - Libero Santarpia
- Humanitas Clinical and Research Institute, Rozzano, Milan, Italy
| | - Lajos Pusztai
- Yale Cancer Center Genetics and Genomics Program, Yale University School of Medicine, New Haven, Connecticut
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Frank J Slack
- Institute for RNA Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
246
|
Alemar B, Gregório C, Ashton-Prolla P. miRNAs As Diagnostic and Prognostic Biomarkers in Pancreatic Ductal Adenocarcinoma and Its Precursor Lesions: A Review. Biomark Insights 2015; 10:113-24. [PMID: 26688661 PMCID: PMC4677802 DOI: 10.4137/bmi.s27679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 08/30/2015] [Accepted: 09/06/2015] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a rare but lethal tumor, is difficult to diagnose without performing an invasive procedure. miRNAs are known to be deregulated in PDAC patients, and recent studies have shown that they can be used as diagnostic and prognostic of the disease. The detection of miRNAs in samples acquired through minimally or noninvasive procedures, such as serum, plasma, and saliva, can have a positive impact on the clinical management of these patients. This article is a comprehensive review of the major studies that have evaluated the expression of miRNAs as biomarkers in pancreatic cancer and its premalignant lesions.
Collapse
Affiliation(s)
- Bárbara Alemar
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cleandra Gregório
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patricia Ashton-Prolla
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
247
|
Adams BD, Parsons C, Slack FJ. The tumor-suppressive and potential therapeutic functions of miR-34a in epithelial carcinomas. Expert Opin Ther Targets 2015; 20:737-53. [PMID: 26652031 DOI: 10.1517/14728222.2016.1114102] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Many RNA species have been identified as important players in the development of chronic diseases including cancer. Certain classes of regulatory RNAs such as microRNAs (miRNAs) have been investigated in such detail that bona fide tumor suppressive and oncogenic miRNAs have been identified. Because of this, there has been a major effort to therapeutically target these small RNAs. One in particular, a liposomal formulation of miR-34a (MRX34), has entered Phase I trials. AREAS COVERED This review aims to summarize miRNA biology, its regulation within normal versus disease states and how it can be targeted therapeutically, with a particular emphasis on miR-34a. Understanding the complexity of a single miRNA will aid in the development of future RNA-based therapeutics for a broader range of chronic diseases. EXPERT OPINION The potential of miRNAs to be developed into anti-cancer therapeutics has become an increasingly important area of research. miR-34a is a tumor suppressive miRNA across many tumor types through its ability to inhibit cellular proliferation, invasion and tumor sphere formation. miR-34a also shows promise within certain in vivo solid tumor models. Finally, as miR-34a moves into clinical trials it will be important to determine if it can further sensitize tumors to certain chemotherapeutic agents.
Collapse
Affiliation(s)
- Brian D Adams
- a Department of Molecular, Cellular and Developmental Biology , Yale University , New Haven , CT , USA.,b Department of Pathology , BIDMC Cancer Center/Harvard Medical School , Boston , MA , USA
| | - Christine Parsons
- a Department of Molecular, Cellular and Developmental Biology , Yale University , New Haven , CT , USA
| | - Frank J Slack
- b Department of Pathology , BIDMC Cancer Center/Harvard Medical School , Boston , MA , USA
| |
Collapse
|
248
|
Chen X, Dong C, Law PTY, Chan MTV, Su Z, Wang S, Wu WKK, Xu H. MicroRNA-145 targets TRIM2 and exerts tumor-suppressing functions in epithelial ovarian cancer. Gynecol Oncol 2015; 139:513-9. [PMID: 26472353 DOI: 10.1016/j.ygyno.2015.10.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 10/05/2015] [Accepted: 10/10/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Epithelial ovarian cancer (EOC) is one of the most common cancers in women worldwide but relatively little is known about its molecular pathogenesis. MicroRNAs, which regulate gene expression post-transcriptionally, have emerged as key players in tumorigenesis. The present study aims to investigate the dysregulation of miR-145 in EOC. METHODS miRNA expression was assessed in EOC tissues and cell lines by quantitative reverse transcription (RT)-PCR. Xenograft mouse model was used for evaluation of the effect of miR-145 on tumor growth. Cell proliferation, colony formation assays, invasion assay, flow cytometry, Western blot and gene expression analysis were used for identification of the functional role of miR-145 in EOC cells. Luciferase reporter assay was used to confirm the interaction between miR-145 and its target mRNA 3'-UTR. RESULTS miR-145 expression was downregulated in EOC tissues and cell lines as compared with normal ovarian tissues. Transfection of miR-145 agomiR significantly inhibited the proliferation, colony forming ability, invasiveness and in vivo tumorigenicity of EOC cells. Transfection of agomiR-145 into EOC cells also markedly induced apoptosis. Furthermore, computational algorithm combined with luciferase reporter assays identified TRIM2 as the direct target of miR-145 in EOC cells. To this end, agomiR-145 downregulated TRIM2 to derepress Bim (a pro-apoptotic Bcl-2 family member degraded by TRIM2). CONCLUSIONS These data confirmed the tumor-suppressing function of miR-145 in EOC and identified TRIM2 as a new miR-145 target. In vivo delivery of agomiR-145 might be a feasible approach for miRNA-directed cancer therapy.
Collapse
Affiliation(s)
- Xiaobo Chen
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Changgui Dong
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai 200062, China.
| | - Priscilla T Y Law
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care and State Key Laboratory of Digestive Disease, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhaoliang Su
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Shengjun Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - William K K Wu
- Department of Anaesthesia and Intensive Care and State Key Laboratory of Digestive Disease, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Huaxi Xu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
249
|
Cortez MA, Ivan C, Valdecanas D, Wang X, Peltier HJ, Ye Y, Araujo L, Carbone DP, Shilo K, Giri DK, Kelnar K, Martin D, Komaki R, Gomez DR, Krishnan S, Calin GA, Bader AG, Welsh JW. PDL1 Regulation by p53 via miR-34. J Natl Cancer Inst 2015; 108:djv303. [PMID: 26577528 PMCID: PMC4862407 DOI: 10.1093/jnci/djv303] [Citation(s) in RCA: 491] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 09/25/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Although clinical studies have shown promise for targeting PD1/PDL1 signaling in non-small cell lung cancer (NSCLC), the regulation of PDL1 expression is poorly understood. Here, we show that PDL1 is regulated by p53 via miR-34. METHODS p53 wild-type and p53-deficient cell lines (p53(-/-) and p53(+/+) HCT116, p53-inducible H1299, and p53-knockdown H460) were used to determine if p53 regulates PDL1 via miR-34. PDL1 and miR-34a expression were analyzed in samples from patients with NSCLC and mutated p53 vs wild-type p53 tumors from The Cancer Genome Atlas for Lung Adenocarcinoma (TCGA LUAD). We confirmed that PDL1 is a direct target of miR-34 with western blotting and luciferase assays and used a p53(R172HΔ)g/+K-ras(LA1/+) syngeneic mouse model (n = 12) to deliver miR-34a-loaded liposomes (MRX34) plus radiotherapy (XRT) and assessed PDL1 expression and tumor-infiltrating lymphocytes (TILs). A two-sided t test was applied to compare the mean between different treatments. RESULTS We found that p53 regulates PDL1 via miR-34, which directly binds to the PDL1 3' untranslated region in models of NSCLC (fold-change luciferase activity to control group, mean for miR-34a = 0.50, SD = 0.2, P < .001; mean for miR-34b = 0.52, SD = 0.2, P = .006; and mean for miR-34c = 0.59, SD = 0.14, and P = .006). Therapeutic delivery of MRX34, currently the subject of a phase I clinical trial, promoted TILs (mean of CD8 expression percentage of control group = 22.5%, SD = 1.9%; mean of CD8 expression percentage of MRX34 = 30.1%, SD = 3.7%, P = .016, n = 4) and reduced CD8(+)PD1(+) cells in vivo (mean of CD8/PD1 expression percentage of control group = 40.2%, SD = 6.2%; mean of CD8/PD1 expression percentage of MRX34 = 20.3%, SD = 5.1%, P = .001, n = 4). Further, MRX34 plus XRT increased CD8(+) cell numbers more than either therapy alone (mean of CD8 expression percentage of MRX34 plus XRT to control group = 44.2%, SD = 8.7%, P = .004, n = 4). Finally, miR-34a delivery reduced the numbers of radiation-induced macrophages (mean of F4-80 expression percentage of control group = 52.4%, SD = 1.7%; mean of F4-80 expression percentage of MRX34 = 40.1%, SD = 3.5%, P = .008, n = 4) and T-regulatory cells. CONCLUSIONS We identified a novel mechanism by which tumor immune evasion is regulated by p53/miR-34/PDL1 axis. Our results suggest that delivery of miRNAs with standard therapies, such as XRT, may represent a novel therapeutic approach for lung cancer.
Collapse
Affiliation(s)
- Maria Angelica Cortez
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Cristina Ivan
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - David Valdecanas
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Xiaohong Wang
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Heidi J Peltier
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Yuping Ye
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Luiz Araujo
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - David P Carbone
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Konstantin Shilo
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Dipak K Giri
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Kevin Kelnar
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Desiree Martin
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Ritsuko Komaki
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Daniel R Gomez
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Sunil Krishnan
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - George A Calin
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - Andreas G Bader
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG)
| | - James W Welsh
- Departments of Experimental Radiation Oncology (MAC, DV, XW, YY), Experimental Therapeutics (CI, GAC), and Radiation Oncology (RK, DRG, SK, JWW), The University of Texas MD Anderson Cancer Center, Houston, TX; Mirna Therapeutics, Inc., Austin, TX (HJP, KK, DM, AGB); Ohio State University, Columbus, OH (LA, DPC, KS); Texas Veterinary Pathology Associates (Houston), Houston, TX (DKG).
| |
Collapse
|
250
|
Tong N, Chu H, Wang M, Xue Y, Du M, Lu L, Zhang H, Wang F, Fang Y, Li J, Wu D, Zhang Z, Sheng X. Pri-miR-34b/c rs4938723 polymorphism contributes to acute lymphoblastic leukemia susceptibility in Chinese children. Leuk Lymphoma 2015; 57:1436-41. [DOI: 10.3109/10428194.2015.1092528] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|