51
|
Integrated microarray meta-analysis identifies miRNA-27a as an oncogene in ovarian cancer by inhibiting FOXO1. Life Sci 2018; 210:263-270. [PMID: 30138596 DOI: 10.1016/j.lfs.2018.08.043] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/10/2018] [Accepted: 08/19/2018] [Indexed: 02/05/2023]
Abstract
AIMS Survival of ovarian cancer patients is generally poor, partly because most of them are already at an advanced stage when diagnosed. The purpose of this study was to screen prognostic miRNAs for ovarian cancer, and to explore the underlying mechanisms. MAIN METHODS Integrated meta-analysis of miRNA microarrays retrieved from public repositories was employed to identify clinically significant miRNAs involved in ovarian cancer. Targets of candidate miRNA were predicted using four online databases, and validated with dual luciferase assay. Loss and gain of function were performed to investigate the role of miR27a in the growth of ovarian cancer cell lines. KEY FINDINGS Based on cross-validation results in multiple datasets, we recognized hsa-miR-27a as an oncogenic molecular and a prognostic factor for ovarian cancer patients. Dual luciferase assay indicated tumor suppressor FOXO1 was a direct target of miR-27a. In addition, hsa-miR-27a could stimulate SKOV3 and A2780 cell proliferation and migration by regulating the expression of FOXO1. SIGNIFICANCE In summary, our results indicate that miR-27a can promote progression of ovarian cancer by mediating FOXO1. To our knowledge, this is the first study focusing on the role of miR-27a/FOXO1 axis using the microarray meta-analysis in ovarian cancer. Furthermore, inhibiting miR-27a expression may be a new strategy for the treatment of ovarian cancer.
Collapse
|
52
|
miR-27a in serum acts as biomarker for prostate cancer detection and promotes cell proliferation by targeting Sprouty2. Oncol Lett 2018; 16:5291-5298. [PMID: 30250598 DOI: 10.3892/ol.2018.9274] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/01/2017] [Indexed: 12/23/2022] Open
Abstract
Prostate cancer (PCa) exhibits a high incidence among men, but there is no effective and non-invasive biomarker for the diagnosis of PCa, and the pathogenesis of PCa remains unclear. The present study identified that miR-27a was significantly overexpressed in the tumor tissues and sera of patients with PCa. In addition, high serum levels of miR-27a were correlated with poor survival in patients with PCa. Receiver-operating characteristic curves analysis demonstrated that the serum levels of miR-27a exhibited a high area under the curve value. Furthermore, miR-27a mimics or inhibitors significantly promoted or repressed the proliferation of PCa cells, respectively. In addition, it was identified that the expression of Sprouty2 (SPRY2) was inversely correlated with the expression of miR-27a in PCa tissues. The knockdown or overexpression of SPRY2 promoted or suppressed the proliferation of PCa cells, respectively, and the overexpression of SPRY2 inhibited the increased proliferation and cell cycle distribution of PCa cells mediated by miR-27a mimics. Taken together, these data indicated that the serum levels of miR-27a may be a novel and non-invasive biomarker for the diagnosis and prognosis of patients with PCa, and miR-27a/SPRY2 may be a therapeutic target for the treatment of PCa.
Collapse
|
53
|
Wu J, Wang Y, Shang L, Qi L, Song M. Five Common Functional Polymorphisms inmicroRNAsand Susceptibility to Breast Cancer: An Updated Meta-Analysis. Genet Test Mol Biomarkers 2018; 22:350-358. [PMID: 29782194 DOI: 10.1089/gtmb.2017.0270] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Jie Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Yusi Wang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Lihua Shang
- Department of Medical Oncology, Third Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lichun Qi
- Department of Cardiovascular Medicine, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mowei Song
- Department of Cardiovascular Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
54
|
Significance of prohibitin domain family in tumorigenesis and its implication in cancer diagnosis and treatment. Cell Death Dis 2018; 9:580. [PMID: 29784973 PMCID: PMC5962566 DOI: 10.1038/s41419-018-0661-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/27/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Abstract
Prohibitin (PHB) was originally isolated and characterized as an anti-proliferative gene in rat liver. The evolutionarily conserved PHB gene encodes two human protein isoforms with molecular weights of ~33 kDa, PHB1 and PHB2. PHB1 and PHB2 belong to the prohibitin domain family, and both are widely distributed in different cellular compartments such as the mitochondria, nucleus, and cell membrane. Most studies have confirmed differential expression of PHB1 and PHB2 in cancers compared to corresponding normal tissues. Furthermore, studies verified that PHB1 and PHB2 are involved in the biological processes of tumorigenesis, including cancer cell proliferation, apoptosis, and metastasis. Two small molecule inhibitors, Rocaglamide (RocA) and fluorizoline, derived from medicinal plants, were demonstrated to interact directly with PHB1 and thus inhibit the interaction of PHB with Raf-1, impeding Raf-1/ERK signaling cascades and significantly suppressing cancer cell metastasis. In addition, a short peptide ERAP and a natural product xanthohumol were shown to target PHB2 directly and prohibit cancer progression in estrogen-dependent cancers. As more efficient biomarkers and targets are urgently needed for cancer diagnosis and treatment, here we summarize the functional role of prohibitin domain family proteins, focusing on PHB1 and PHB2 in tumorigenesis and cancer development, with the expectation that targeting the prohibitin domain family will offer more clues for cancer therapy.
Collapse
|
55
|
Bryzgunova OE, Konoshenko MY, Laktionov PP. MicroRNA-guided gene expression in prostate cancer: Literature and database overview. J Gene Med 2018; 20:e3016. [DOI: 10.1002/jgm.3016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/15/2018] [Accepted: 03/17/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Olga E. Bryzgunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia and ‘E. Meshalkin National Medical Research Center’ of the Ministry of Health of the Russian Federation; Novosibirsk Russia
| | - Maria Yu Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia and ‘E. Meshalkin National Medical Research Center’ of the Ministry of Health of the Russian Federation; Novosibirsk Russia
| | - Pavel P. Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, Russia and ‘E. Meshalkin National Medical Research Center’ of the Ministry of Health of the Russian Federation; Novosibirsk Russia
| |
Collapse
|
56
|
Wang M, Sun J, Xu B, Chrusciel M, Gao J, Bazert M, Stelmaszewska J, Xu Y, Zhang H, Pawelczyk L, Sun F, Tsang SY, Rahman N, Wolczynski S, Li X. Functional Characterization of MicroRNA-27a-3p Expression in Human Polycystic Ovary Syndrome. Endocrinology 2018; 159:297-309. [PMID: 29029022 DOI: 10.1210/en.2017-00219] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 09/14/2017] [Indexed: 11/19/2022]
Abstract
The goal of this study was to characterize the function of microRNA-27a-3p (miR-27a-3p) in polycystic ovary syndrome (PCOS). miR-27a-3p expression was analyzed in excised granulosa cells (GCs) from 21 patients with PCOS and 12 normal patients undergoing in vitro fertilization cycle treatments and in 17 nontreated cuneiform ovarian resection PCOS samples and 13 control ovarian samples from patients without PCOS. We found that the expression of miR-27a-3p was significantly increased in both excised GCs and the ovaries of patients with PCOS compared with the controls. Insulin treatment of the human granulosa-like tumor cell line (KGN) resulted in decreased downregulated expression of miR-27a-3p, and this effect appeared to be mediated by signal transducer and activator of transcription STAT1 and STAT3. The overexpression of miR-27a-3p in KGN cells inhibited SMAD5, which in turn decreased cell proliferation and promoted cell apoptosis. After KGN cells were stimulated with insulin for 48 hours, there was increased expression of SMAD5 protein and decreased apoptosis. Additionally, knockdown/overexpression of SMAD5 in KGN cells reduced/increased cell number and promoted/inhibited cell apoptosis. Insulin-stimulated primary GCs isolated from patients with PCOS, in contrast to normal GCs or KGN cells, did not exhibit decreased miR-27a-3p expression. The differences in the expression levels in KGN cells and human PCOS GCs are likely explained by increased miR-27a-3p expression in the GCs caused by insulin resistance in PCOS. Taken together, our data provided evidence for a functional role of miR-27a-3p in the GCs' dysfunction that occurs in patients with PCOS.
Collapse
Affiliation(s)
- Mingming Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jing Sun
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Bo Xu
- Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Marcin Chrusciel
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku, Finland
| | - Jun Gao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Maciej Bazert
- Department of Infertility and Reproductive Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Joanna Stelmaszewska
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Yunyun Xu
- Department of General Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hongwen Zhang
- Department of General Surgery, 306th Hospital of People's Liberation Army of China, Beijing, China
| | - Leszek Pawelczyk
- Department of Infertility and Reproductive Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Fei Sun
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Suk Ying Tsang
- School of Life Science and State Key Laboratory of Agro-Biotechnology, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Nafis Rahman
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku, Finland
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Slawomir Wolczynski
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Xiangdong Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
57
|
Prostate-specific PTen deletion in mice activates inflammatory microRNA expression pathways in the epithelium early in hyperplasia development. Oncogenesis 2017; 6:400. [PMID: 29284790 PMCID: PMC5865543 DOI: 10.1038/s41389-017-0007-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/07/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023] Open
Abstract
PTen loss is one of the most frequent events in prostate cancer both at the initiation stage and during late stage metastatic development. The mouse model of prostate-specific probasin-mediated Pten deletion leads to prostate intraepithelial neoplasia (PIN) leading to adenocarcinoma. Using this model, we analysed the miR and mRNA transcriptome profile of Pten−/− PIN versus wild type age-matched prostate tissues and analysed the effects of Pten loss on miR expression in the early neoplastic process. At the PIN stage, Pten loss significantly changed the expression of over 20 miRNAs and over 4000 genes. The observed miR expression indicated a strong immunological cohort, which is seen in many human and mouse cancers and is thought to derive from infiltrating B and T immune cells. However, upon in situ hybridisation, these immunologically related miRs did not correlate with immune cell location, and emanated from the prostate epithelium itself and not from the associated immune cells present. Growing Pten−/− prostate cells in culture showed that the overexpressed miRNAs seen in Pten−/− were directly in response to the overactive PI3 kinase pathway and were in part responsible in reducing target gene expression levels. Inhibition of PI3 kinase downstream regulators, or re-introducing wild type PtencDNA reduced miR overexpression resulting in increased miR target gene expression. MiR inhibitors also showed this pattern, and synergised with an mTORC1 inhibitor. Overall, Pten deletion in the prostate epithelium activated a cohort of inflammation-related miRs usually associated with immune responses from B and T cells. These oncomiRs may then accelerate carcinogenesis.
Collapse
|
58
|
Feng Y, Duan F, Song C, Zhao X, Dai L, Cui S. Systematic evaluation of cancer risk associated with rs2292832 in miR‑149 and rs895819 in miR‑27a: a comprehensive and updated meta‑analysis. Oncotarget 2017; 7:22368-84. [PMID: 26993779 PMCID: PMC5008366 DOI: 10.18632/oncotarget.8082] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/24/2016] [Indexed: 01/23/2023] Open
Abstract
The aim of this study is to provide a precise quantification for the association between miR-149 T > C (rs2292832) and miR-27a A > G (rs895819) and the risk of cancer. We conducted a systematic literature review and evaluated the quality of included studies based on Newcastle-Ottawa Scale (NOS). Pooled odds ratios (ORs) and corresponding 95% confidence intervals (95% CIs) were calculated to assess the strengths of the associations. We identified 40 studies for pooled analyses. Overall, the results demonstrated that the rs2292832 polymorphism was subtly decrease the risk of breast cancer (CT + CC vs TT: OR = 0.83, 95% CI: 0.70-0.98, P = 0.03; CC vs CT + TT: OR = 0.80, 95% CI: 0.68-0.93, P = 0.00), and the rs895819 polymorphism wasassociated with significantly increased cancer risk in the Asian population (AG + GG vs AA: OR = 1.24, 95% CI: 1.03-1.50, P = 0.02) and in colorectal cancer subgroup (GG vs AA: OR = 1.45, 95% CI: 1.10-1.92, P = 0.00; AG + GG vs AA: OR = 1.35, 95% CI: 1.15-1.58, P = 0.00; GG vs AG + AA: OR = 1.36, 95% CI: 1.04-1.77, P = 0.02). In addition, a subtly decreased risk was observed in the Caucasian population and in breast cancer subgroup. In conclusion, the rs2292832 polymorphism was significantly associated with increased breast cancer risk, and the rs895819 polymorphism contributes to the susceptibility of colorectal and breast cancer.
Collapse
Affiliation(s)
- Yajing Feng
- Department of Infection Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R.China
| | - Fujiao Duan
- Department of Hospital Infection Management, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, Henan, P.R.China
| | - Chunhua Song
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, P.R.China
| | - Xia Zhao
- Department of Hospital Infection Management, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, Henan, P.R.China
| | - Liping Dai
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, P.R.China
| | - Shuli Cui
- College of Professional Study, Northeastern University, Boston, 02215 Massachusetts, USA
| |
Collapse
|
59
|
Wang M, Liu M, Sun J, Jia L, Ma S, Gao J, Xu Y, Zhang H, Tsang SY, Li X. MicroRNA-27a-3p affects estradiol and androgen imbalance by targeting Creb1 in the granulosa cells in mouse polycytic ovary syndrome model. Reprod Biol 2017; 17:295-304. [PMID: 29089199 DOI: 10.1016/j.repbio.2017.09.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 01/01/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine abnormality in women characterized by a menstrual disturbance with chronic anovulation and hyperandrogenism, polycystic ovaries, and insulin resistance. MicroRNAs (miRNAs) are important fine-tune regulators involved in various physiological and pathological processes, but their actions are not fully understood. In this study, we observed the increased expression of miR-27a-3p in the ovaries of mice with PCOS and explored its functions in primary mouse granulosa cells (mGCs) and the mouse granulosa-like tumor cell line, KK-1, using several approaches. QPCR results showed that miR-27a-3p expression was significantly higher in mGCs at the preantral follicle (PrF) stage. Using flow cytometry and hormone analysis, we found that overexpression of miR-27a-3p promoted apoptosis and inhibited estradiol (E2) production in KK-1 cells. Moreover using a luciferase assay and Western blotting analysis, we verified that the gene of cyclic AMP response element (CRE)-binding protein 1 (Creb1) was a potential target of miR-27a-3p, which in effect hindered the expression of its downstream factor cytochrome P450 family 19 subfamily A polypeptide 1 (Cyp19a1). With the decrease of aromatase activity, testosterone (T) is reduced to dihydrotestosterone (DHT) and this exerts its effect of upregulation of the miR-27a-3p expression. The imbalance of androgen and E2 levels affected by miR-27a-3p and its function of promoting GC apoptosis could be involved in the pathophysiology of PCOS. Our results indicate that miR-27a-3p in PCOS GCs may play an important role in ovarian follicular development and provide new insights into GC dysfunction in PCOS.
Collapse
Affiliation(s)
- Mingming Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Mei Liu
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing Sun
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Lina Jia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shuoqian Ma
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jun Gao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yunyun Xu
- Department of General Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Hongwen Zhang
- Department of General Surgery, 306th Hospital of PLA, Beijing 100101, China
| | - Suk Ying Tsang
- School of Life Science and State Key Laboratory of Agro-Biotechnology, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Xiangdong Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
60
|
Rao Z, He Z, He Y, Guo Z, Kong D, Liu J. MicroRNA‑512‑3p is upregulated, and promotes proliferation and cell cycle progression, in prostate cancer cells. Mol Med Rep 2017; 17:586-593. [PMID: 29115469 DOI: 10.3892/mmr.2017.7844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 07/07/2017] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed cancer in males worldwide. MicroRNAs (miRNAs/miRs) are small non‑coding RNAs that participate in the regulation of various biological processes by regulating post‑transcriptional gene expression. However, whether dysregulation of miRNA expression may be associated with the carcinogenesis of PCa remains to be elucidated. The present study identified differentially expressed miRNAs in PCa by analyzing two publicly available gene expression datasets, GSE14857 and GSE21036. The results demonstrated that miR‑512‑3p was significantly upregulated in PCa. Furthermore, the present study explored the molecular functions of miR‑512‑3p in PCa, and demonstrated that overexpression of miR‑512‑3p promoted PCa cell proliferation and reduced G1 phase cell cycle arrest in PCa. These results indicated that miR‑512‑3p may act as an oncogene in PCa. To the best of our knowledge, this is the first study revealed the molecular functions of miR‑512‑3p in PCa. To obtain valuable insights into the potential mechanisms of miR‑512‑3p, bioinformatics analyses were performed to identify the targets of miR‑512‑3p. Kyoto Encyclopedia of Genes and Genomes pathway and Gene Ontology category analyses revealed that miR‑512‑3p may be associated with the mitogen‑activated protein kinase signaling pathway and numerous biological processes, including cell adhesion, cell proliferation, cell cycle and apoptosis. These results suggested that miR‑512‑3p may be considered a potential diagnostic and therapeutic target of PCa.
Collapse
Affiliation(s)
- Zhigang Rao
- Department of Urology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437000, P.R. China
| | - Ziqi He
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Yi He
- Department of Obstetrics and Gynecology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437000, P.R. China
| | - Zonghua Guo
- Department of Urology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437000, P.R. China
| | - Dongbo Kong
- Department of Urology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437000, P.R. China
| | - Jufang Liu
- Department of Obstetrics and Gynecology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437000, P.R. China
| |
Collapse
|
61
|
Crosstalk between the Androgen Receptor and PPAR Gamma Signaling Pathways in the Prostate. PPAR Res 2017; 2017:9456020. [PMID: 29181019 PMCID: PMC5664321 DOI: 10.1155/2017/9456020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/29/2017] [Accepted: 09/14/2017] [Indexed: 01/07/2023] Open
Abstract
Nuclear receptors are a superfamily of ligand-activated transcription factors that play critical roles in the regulation of normal biological processes and several disease states. Of the nuclear receptors expressed within the prostate, the androgen receptor (AR) promotes the differentiation of prostatic epithelial cells and stimulates production of enzymes needed for liquefaction of semen. Multiple forms of AR also promote the growth of both early and late stage prostate cancers. As a result, drugs that target the AR signaling pathway are routinely used to treat patients with advanced forms of prostate cancer. Data also suggest that a second member of the nuclear receptor superfamily, the peroxisome proliferator activated receptor gamma (PPARγ), is a tumor suppressor that regulates growth of normal prostate and prostate cancers. Recent studies indicate there is a bidirectional interaction between AR and PPARγ, with each receptor influencing the expression and/or activity of the other within prostatic tissues. In this review, we examine how AR and PPARγ each regulate the growth and development of normal prostatic epithelial cells and prostate cancers. We also discuss interactions between the AR and PPARγ signaling pathways and how those interactions may influence prostate biology.
Collapse
|
62
|
Massillo C, Dalton GN, Farré PL, De Luca P, De Siervi A. Implications of microRNA dysregulation in the development of prostate cancer. Reproduction 2017; 154:R81-R97. [DOI: 10.1530/rep-17-0322] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/25/2017] [Accepted: 07/10/2017] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are non-coding small RNAs that target mRNA to reduce protein expression. They play fundamental roles in several diseases, including prostate cancer (PCa). A single miRNA can target hundreds of mRNAs and coordinately regulate them, which implicates them in nearly every biological pathway. Hence, miRNAs modulate proliferation, cell cycle, apoptosis, adhesion, migration, invasion and metastasis, most of them constituting crucial hallmarks of cancer. Due to these properties, miRNAs emerged as promising tools for diagnostic, prognosis and management of cancer patients. Moreover, they come out as potential targets for cancer treatment, and several efforts are being made to progress in the field of miRNA-based cancer therapy. In this review, we will summarize the recent information about miRNAs in PCa. We will recapitulate all the miRNAs involved in the androgen pathway and the biology of PCa, focusing in PCa initiation and progression. In particular, we will describe the miRNAs associated with cell proliferation, cell cycle and apoptosis in PCa, as well as invasion, adhesion and metastatic miRNAs. We will revise the recent progress made understanding the role of circulating miRNAs identified in PCa that might be useful for PCa patient stratification. Another key aspect to be discussed in this review is miRNAs’ role in PCa therapy, including the miRNAs delivery.
Collapse
|
63
|
Kanwal R, Plaga AR, Liu X, Shukla GC, Gupta S. MicroRNAs in prostate cancer: Functional role as biomarkers. Cancer Lett 2017; 407:9-20. [DOI: 10.1016/j.canlet.2017.08.011] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/03/2017] [Accepted: 08/06/2017] [Indexed: 12/19/2022]
|
64
|
Koushyar S, Economides G, Zaat S, Jiang W, Bevan CL, Dart DA. The prohibitin-repressive interaction with E2F1 is rapidly inhibited by androgen signalling in prostate cancer cells. Oncogenesis 2017; 6:e333. [PMID: 28504694 PMCID: PMC5523065 DOI: 10.1038/oncsis.2017.32] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/16/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022] Open
Abstract
Prohibitin (PHB) is a tumour suppressor molecule with pleiotropic activities across several cellular compartments including mitochondria, cell membrane and the nucleus. PHB and the steroid-activated androgen receptor (AR) have an interplay where AR downregulates PHB, and PHB represses AR. Additionally, their cellular locations and chromatin interactions are in dynamic opposition. We investigated the mechanisms of cell cycle inhibition by PHB and how this is modulated by AR in prostate cancer. Using a prostate cancer cell line overexpressing PHB, we analysed the gene expression changes associated with PHB-mediated cell cycle arrest. Over 1000 gene expression changes were found to be significant and gene ontology analysis confirmed PHB-mediated repression of genes essential for DNA replication and synthesis, for example, MCMs and TK1, via an E2F1 regulated pathway—agreeing with its G1/S cell cycle arrest activity. PHB is known to inhibit E2F1-mediated transcription, and the PHB:E2F1 interaction was seen in LNCaP nuclear extracts, which was then reduced by androgen treatment. Upon two-dimensional western blot analysis, the PHB protein itself showed androgen-mediated charge differentiation (only in AR-positive cells), indicating a potential dephosphorylation event. Kinexus phosphoprotein array analysis indicated that Src kinase was the main interacting intracellular signalling hub in androgen-treated LNCaP cells, and that Src inhibition could reduce this AR-mediated charge differentiation. PHB charge change may be associated with rapid dissociation from chromatin and E2F1, allowing the cell cycle to proceed. The AR and androgens may deactivate the repressive functions of PHB upon E2F1 leading to cell cycle progression, and indicates a role for AR in DNA replication licensing.
Collapse
Affiliation(s)
- S Koushyar
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| | - G Economides
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| | - S Zaat
- Androgen Signalling Laboratory, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - W Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| | - C L Bevan
- Androgen Signalling Laboratory, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - D A Dart
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
65
|
Malla B, Zaugg K, Vassella E, Aebersold DM, Dal Pra A. Exosomes and Exosomal MicroRNAs in Prostate Cancer Radiation Therapy. Int J Radiat Oncol Biol Phys 2017; 98:982-995. [PMID: 28721912 DOI: 10.1016/j.ijrobp.2017.03.031] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 12/11/2022]
Abstract
Despite current risk stratification systems using traditional clinicopathologic factors, many localized and locally advanced prostate cancers fail radical treatment (ie, radical prostatectomy, radiation therapy with or without androgen deprivation therapy). Therefore, a pressing need exists for enhanced methods of disease stratification through novel prognostic and predictive tools that can reliably be applied in clinical practice. Exosomes are 50- to 150-nm small vesicles released by cancer cells that reflect the genetic and nongenetic materials of parent cancer cells. Cancer cells can contain distinct sets of microRNA profiles, the expression of which can change owing to stress such as radiation therapy. These alterations or distinctions in contents allow exosomes to be used as prognostic and/or predictive biomarkers and to monitor the treatment response. Additionally, microRNAs have been shown to influence multiple processes in prostate tumorigenesis, including cell proliferation, induction of apoptosis, migration, oncogene inhibition, and radioresistance. Thus, comparative exosomal microRNA profiling at different levels could help portray tumor aggressiveness and response to radiation therapy. Although technical challenges persist in exosome isolation and characterization, recent improvements in microRNA profiling have evolved toward in-depth analyses of the exosomal cargo and its functions. We have reviewed the role of exosomes and exosomal microRNAs in biologic processes of prostate cancer progression and radiation therapy response, with a particular focus on the development of clinical assays for treatment personalization.
Collapse
Affiliation(s)
- Bijaya Malla
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Kathrin Zaugg
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Erik Vassella
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Alan Dal Pra
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland.
| |
Collapse
|
66
|
Fletcher CE, Godfrey JD, Shibakawa A, Bushell M, Bevan CL. A novel role for GSK3β as a modulator of Drosha microprocessor activity and MicroRNA biogenesis. Nucleic Acids Res 2017; 45:2809-2828. [PMID: 27907888 PMCID: PMC5389555 DOI: 10.1093/nar/gkw938] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 09/13/2016] [Accepted: 10/19/2016] [Indexed: 01/13/2023] Open
Abstract
Regulation of microRNA (miR) biogenesis is complex and stringently controlled. Here, we identify the kinase GSK3β as an important modulator of miR biogenesis at Microprocessor level. Repression of GSK3β activity reduces Drosha activity toward pri-miRs, leading to accumulation of unprocessed pri-miRs and reduction of pre-miRs and mature miRs without altering levels or cellular localisation of miR biogenesis proteins. Conversely, GSK3β activation increases Drosha activity and mature miR accumulation. GSK3β achieves this through promoting Drosha:cofactor and Drosha:pri-miR interactions: it binds to DGCR8 and p72 in the Microprocessor, an effect dependent upon presence of RNA. Indeed, GSK3β itself can immunoprecipitate pri-miRs, suggesting possible RNA-binding capacity. Kinase assays identify the mechanism for GSK3β-enhanced Drosha activity, which requires GSK3β nuclear localisation, as phosphorylation of Drosha at S300 and/or S302; confirmed by enhanced Drosha activity and association with cofactors, and increased abundance of mature miRs in the presence of phospho-mimic Drosha. Functional implications of GSK3β-enhanced miR biogenesis are illustrated by increased levels of GSK3β-upregulated miR targets following GSK3β inhibition. These data, the first to link GSK3β with the miR cascade in humans, highlight a novel pro-biogenesis role for GSK3β in increasing miR biogenesis as a component of the Microprocessor complex with wide-ranging functional consequences.
Collapse
Affiliation(s)
- Claire E Fletcher
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Jack D Godfrey
- Medical Research Council Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Akifumi Shibakawa
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Martin Bushell
- Medical Research Council Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Charlotte L Bevan
- Imperial Centre for Translational and Experimental Medicine, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
67
|
Liu P, Xu Y, Zhang W, Li Y, Tang L, Chen W, Xu J, Sun Q, Guan X. Prohibitin promotes androgen receptor activation in ER-positive breast cancer. Cell Cycle 2017; 16:776-784. [PMID: 28272969 DOI: 10.1080/15384101.2017.1295193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Prohibitin (PHB) is an evolutionarily conserved protein with multiple functions in both normal and cancer cells. Androgen receptor (AR) was reported to act as a different role in the ER-positive and ER-negative breast cancer. However, little is known about the role of PHB and whether PHB could regulate AR expression in the ER-positive breast cancer. Here, we determined the expression and clinical outcomes of PHB in breast cancer samples using 121 breast cancer tissues and published databases, and investigated the role of PHB in breast cancer cell growth, apoptosis and cell cycle arrest in the ER-positive breast cancer cells. We obtained the expression of PHB is significantly low in breast cancer samples, and low PHB expression positively correlated with poor prognosis of breast cancer. We detected that PHB could inhibit breast cancer cell proliferation, change cell cycle distribution and promote cell apoptosis in the ER-positive breast cancer cells. Moreover, we found PHB could significantly increase AR expression in both mRNA and protein levels in the ER-positive breast cancer cells. Additionally, a significant positive correlation between PHB and AR expression was identified in the 121 breast cancer tissues. PHB and AR expression are associated with prognosis in the ER-positive breast cancer patients. Our results indicate that PHB promotes AR activation in ER-positive breast cancer, making PHB and AR potential molecular targets for ER-positive breast cancer therapy.
Collapse
Affiliation(s)
- Pengying Liu
- a Department of Medical Oncology , Jinling Hospital, School of Medicine, Southern Medical University , Guangzhou , China
| | - Yumei Xu
- a Department of Medical Oncology , Jinling Hospital, School of Medicine, Southern Medical University , Guangzhou , China
| | - Wenwen Zhang
- b Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| | - Yan Li
- a Department of Medical Oncology , Jinling Hospital, School of Medicine, Southern Medical University , Guangzhou , China
| | - Lin Tang
- b Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| | - Weiwei Chen
- b Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| | - Jing Xu
- b Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| | - Qian Sun
- b Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| | - Xiaoxiang Guan
- a Department of Medical Oncology , Jinling Hospital, School of Medicine, Southern Medical University , Guangzhou , China.,b Department of Medical Oncology , Jinling Hospital, Medical School of Nanjing University , Nanjing , China
| |
Collapse
|
68
|
Regulation of TFPIα expression by miR-27a/b-3p in human endothelial cells under normal conditions and in response to androgens. Sci Rep 2017; 7:43500. [PMID: 28240250 PMCID: PMC5327489 DOI: 10.1038/srep43500] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/26/2017] [Indexed: 12/29/2022] Open
Abstract
The increased risk of cardiovascular events in older men is multifactorial, but the significant reduction of testosterone levels has been involved. As this hormone regulates the expression of TFPI by unknown mechanisms, we aimed to evaluate the role of miRNAs in the regulation of TFPIα expression under normal conditions and in response to androgens. In silico studies allowed the selection of 4 miRNAs as potential TFPIα regulators. Only miR-27a/b-3p significantly reduced TFPIα expression in two endothelial cell lines. Luciferase assays demonstrated a direct interaction between miR-27a/b-3p and TFPI 3′UTR. Ex vivo analysis of TFPI and miRNA levels in 74 HUVEC samples from healthy subjects, showed a significant and inverse correlation between TFPI and miR-27a-3p. Moreover, anticoagulant activity of TFPIα from cells supernatants decreased ~30% with miR-27a/b-3p and increased ~50% with anti-miR-27a/b-3p. Interestingly, treatment of EA.hy926 with a physiological dose of dihydrotestosterone (30 nM) significantly increased (~40%) TFPIα expression with a parallel decreased (~50%) of miR-27a/b-3p expression. In concordance, increased levels of miR-27a/b-3p normalized the up-regulation induced by testosterone. Our results suggest that testosterone is a hinge in miR-27/TFPIα regulation axis. Future studies are needed to investigate whether testosterone variations are involved in a miR-27/TFPIα dysregulation that could increase the cardiovascular risk.
Collapse
|
69
|
Epigenomic Regulation of Androgen Receptor Signaling: Potential Role in Prostate Cancer Therapy. Cancers (Basel) 2017; 9:cancers9010009. [PMID: 28275218 PMCID: PMC5295780 DOI: 10.3390/cancers9010009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/02/2017] [Accepted: 01/11/2017] [Indexed: 12/18/2022] Open
Abstract
Androgen receptor (AR) signaling remains the major oncogenic pathway in prostate cancer (PCa). Androgen-deprivation therapy (ADT) is the principle treatment for locally advanced and metastatic disease. However, a significant number of patients acquire treatment resistance leading to castration resistant prostate cancer (CRPC). Epigenetics, the study of heritable and reversible changes in gene expression without alterations in DNA sequences, is a crucial regulatory step in AR signaling. We and others, recently described the technological advance Chem-seq, a method to identify the interaction between a drug and the genome. This has permitted better understanding of the underlying regulatory mechanisms of AR during carcinogenesis and revealed the importance of epigenetic modifiers. In screening for new epigenomic modifiying drugs, we identified SD-70, and found that this demethylase inhibitor is effective in CRPC cells in combination with current therapies. The aim of this review is to explore the role of epigenetic modifications as biomarkers for detection, prognosis, and risk evaluation of PCa. Furthermore, we also provide an update of the recent findings on the epigenetic key processes (DNA methylation, chromatin modifications and alterations in noncoding RNA profiles) involved in AR expression and their possible role as therapeutic targets.
Collapse
|
70
|
Ceder Y, Bjartell A, Culig Z, Rubin MA, Tomlins S, Visakorpi T. The Molecular Evolution of Castration-resistant Prostate Cancer. Eur Urol Focus 2016; 2:506-513. [PMID: 28723516 DOI: 10.1016/j.euf.2016.11.012] [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: 11/17/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 12/28/2022]
Abstract
CONTEXT Androgen deprivation therapy (ADT) is the backbone of treatment for advanced prostate cancer. However, castration-resistant prostate cancer (CRPC) nearly invariably develops through a range of different molecular mechanisms accompanied by progression to a more aggressive phenotype. OBJECTIVE To understand the key molecular mechanisms leading to CRPC and the functional implications of this progression. Understanding molecular evolutionary mechanisms in CRPC is essential for the development of novel curative therapeutic approaches. EVIDENCE ACQUISITION A systematic literature search to identify relevant original articles was conducted using PubMed. Findings verified in independent studies and supported by in vivo data were prioritised. From the eligible collection, 50 papers were selected. EVIDENCE SYNTHESIS The majority of CRPC tumours harbour alterations in the androgen receptor (AR) at the DNA, RNA, and/or protein level, and/or other alterations involving the AR signalling pathway, so this central molecule is the focus of this review. To survive and resume growth despite low levels of circulating androgens, prostate cancer cells can also adapt androgen synthesis or induce alternative pathways. CONCLUSIONS Despite more efficient ADT strategies, most evidence points to persistent AR signalling as a major mechanism of progression to CRPC. Resistance due to transdifferentiation or AR independence is also emerging as a mechanism of resistance. The diversity of potential resistance mechanisms supports the need for combination treatment and serial monitoring for adaptive treatment strategies. PATIENT SUMMARY In this review, we summarise how prostate cancer cells evade androgen deprivation therapy and become more aggressive. Defining the molecular mechanisms will be critical for the development of new treatment approaches and hence improved survival.
Collapse
Affiliation(s)
- Yvonne Ceder
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, Lund, Sweden.
| | - Anders Bjartell
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Malmö, Sweden
| | - Zoran Culig
- Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Mark A Rubin
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medicine and Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Scott Tomlins
- Michigan Center for Translational Pathology, Department of Pathology, Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology, University of Tampere and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
71
|
Shukla KK, Misra S, Pareek P, Mishra V, Singhal B, Sharma P. Recent scenario of microRNA as diagnostic and prognostic biomarkers of prostate cancer. Urol Oncol 2016; 35:92-101. [PMID: 27890424 DOI: 10.1016/j.urolonc.2016.10.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 10/24/2016] [Accepted: 10/26/2016] [Indexed: 12/11/2022]
Abstract
Prostate cancer (CaP) is a leading cause of cancer death and displays a broad range of clinical behavior from relatively indolent to aggressive metastatic disease. Due to the alteration and incomplete characterization of the CaP genomic markers, the quest for novel cellular metabolic regulatory molecules like micro RNA (miRNA) as a biomarker could be considered for the prognosis and treatment of CaP in future. In this article, we review the existing literature pertaining to CaP. Study provides a comprehensive miRNA profile expressed in CaP. Beside the miRNA expressed in the tumor tissue, circulating miRNAs have been found highly stable and are both detectable and quantifiable in a range of accessible bio fluids; therefore, miRNA has the potential to be useful diagnostic, prognostic and predictive biomarker. Along with being an important molecule in modulation of CaP progression, the miRNA have certain limitations such as lack of stable expression of multiple target genes and often disrupt entire signaling networks of cellular metabolic pathways. We conclude that: The alteration of miRNA and their role played in cellular regulatory networks would be the next target of basic research in CaP. The miRNAs identified may be validated and modeled to understand their role in CaP, using bioinformatics. There is an immediate unmet need in the translational approach of identified miRNAs. The characterization of miRNAs involved in CaP is still incomplete: adequate validation studies are required to corroborate current results.
Collapse
Affiliation(s)
- Kamla Kant Shukla
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India.
| | - Sanjeev Misra
- Department of Surgical Oncology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Puneet Pareek
- Department of Radiation Oncology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Vivek Mishra
- Department of Biotechnology, IFTM University, Moradabad, Uttar Pradesh, India
| | - Barkha Singhal
- Department of Biology, Texas Woman׳s University, Denton, TX, USA
| | - Parveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
72
|
Wan X, Huang W, Yang S, Zhang Y, Zhang P, Kong Z, Li T, Wu H, Jing F, Li Y. Androgen-induced miR-27A acted as a tumor suppressor by targeting MAP2K4 and mediated prostate cancer progression. Int J Biochem Cell Biol 2016; 79:249-260. [PMID: 27594411 DOI: 10.1016/j.biocel.2016.08.043] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/18/2016] [Accepted: 08/29/2016] [Indexed: 11/17/2022]
Abstract
Prostate cancer (PCa) is the most commonly diagnosed and secondly leading cause of cancer death among males. But the precise mechanism of prostate cancer progression, including microRNAs (miRNAs) functioning in it, is still needs further study. We found miR-27a to be down-regulated in prostate cancer, and we investigated the mechanism and role of miRNA-27a in prostate cancer. MiR-27a, a transcriptional target of AR, was an androgen-induced miRNA in LNCaP cells. In castration-resistant prostate cancer (CRPC) cells, we for the first time reported that miR-27a was downregulated by PI3K signaling. MiR-27a functioned as a tumor suppressor in prostate cancer. Over-expression of miR-27a decreased prostate cancer cell proliferation and migration, and induced prostate cancer cell cycle arrest and apoptosis. MAP2K4, miR-27a's direct target gene, functioned as an oncogene in prostate cancer by reducing G1-S phase arrest and inhibiting cell apoptosis of prostate cancer cells. In conclusion, miR-27a functions as a tumor suppressor by suppressing MAP2K4 which acts as an oncogene in prostate cancer cell lines; we also provided a new mechanism of castration-resistant prostate cancer mediated by miR-27a that downregulation of miR-27a caused by aberrant AR signaling and PI3K/Akt signaling after androgen deprivation therapy (ADT) would promote the progression of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Xuechao Wan
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai 200433, PR China
| | - Wenhua Huang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai 200433, PR China
| | - Shu Yang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai 200433, PR China
| | - Yalong Zhang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai 200433, PR China
| | - Pu Zhang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai 200433, PR China
| | - Zhe Kong
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai 200433, PR China
| | - Tao Li
- Shanghai Institute of Planned Parenthood Research Hospital, WHO Collaborating Center for Research in Human Reproduction, Shanghai 200433, PR China
| | - Hai Wu
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai 200433, PR China
| | - Fengxiang Jing
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Science, Shanghai 200433, PR China.
| | - Yao Li
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai 200433, PR China.
| |
Collapse
|
73
|
Antiproliferative effect of a synthetic aptamer mimicking androgen response elements in the LNCaP cell line. Cancer Gene Ther 2016; 23:254-7. [PMID: 27364573 DOI: 10.1038/cgt.2016.26] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/26/2016] [Accepted: 04/26/2016] [Indexed: 02/07/2023]
Abstract
Prostate cancer usually develops to a hormone-refractory state that is irresponsive to conventional therapeutic approaches. Therefore, new methods for treating aggressive prostate cancer are under development. Because of the importance of androgen receptors (ARs) in the development of the hormone-refractory state and AR mechanism of action, this study was designed. A single-stranded DNA as an aptamer was designed that could mimic the hormone response element (HRE). The LNCaP cells as an AR-rich model were divided into three sets of triplicate groups: the test group was transfected with Aptamer Mimicking HRE (AMH), Mock received only transfection reagents (mock) and a negative control. All three sets received 0, 10 and 100 nM of dehydroepiandrosterone (DHEA) separately. Data analysis showed hormone dependency of LNCaP cells in the negative control group upon treatment with 10 and 100 nM DHEA (compared with cells left untreated (P=0.001)). Transfection of AMH resulted in significant reduction of proliferation in the test group when compared with the negative control group with 10 (P=0.001) or 100 nM DHEA (P=0.02). AMH can form a hairpin structure at 37 °C and mimic the genomic HRE. Hence, it is capable of effectively competing with genomic HRE and interrupting the androgen signaling pathway in a prostate cancer cell line (LNCaP).
Collapse
|
74
|
Ding M, Lin B, Li T, Liu Y, Li Y, Zhou X, Miao M, Gu J, Pan H, Yang F, Li T, Liu XY, Li R. A dual yet opposite growth-regulating function of miR-204 and its target XRN1 in prostate adenocarcinoma cells and neuroendocrine-like prostate cancer cells. Oncotarget 2016; 6:7686-700. [PMID: 25797256 PMCID: PMC4480709 DOI: 10.18632/oncotarget.3480] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 02/03/2015] [Indexed: 12/01/2022] Open
Abstract
Androgen deprivation therapy in prostate cancer (PCa) causes neuroendocrine differentiation (NED) of prostatic adenocarcinomas (PAC) cells, leading to recurrence of PCa. Androgen-responsive genes involved in PCa progression including NED remain largely unknown. Here we demonstrated the importance of androgen receptor (AR)-microRNA-204 (miR-204)-XRN1 axis in PCa cell lines and the rat ventral prostate. Androgens downregulate miR-204, resulting in induction of XRN1 (5′-3′ exoribonuclease 1), which we identified as a miR-204 target. miR-204 acts as a tumor suppressor in two PAC cell lines (LNCaP and 22Rv1) and as an oncomiR in two neuroendocrine-like prostate cancer (NEPC) cell lines (PC-3 and CL1). Importantly, overexpression of miR-204 and knockdown of XRN1 inhibited AR expression in PCa cells. Repression of miR-34a, a known AR-targeting miRNA, contributes AR expression by XRN1. Thus we revealed the AR-miR-204-XRN1-miR-34a positive feedback loop and a dual function of miR-204/XRN1 axis in prostate cancer.
Collapse
Affiliation(s)
- Miao Ding
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.,WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Biaoyang Lin
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Urology, University of Washington, Seattle, WA, USA
| | - Tao Li
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanyuan Liu
- WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Yuhua Li
- WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Xiaoyu Zhou
- WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Maohua Miao
- WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Jinfa Gu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Hongjie Pan
- WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Fen Yang
- WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Tianqi Li
- WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Xin Yuan Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Runsheng Li
- WHO Collaborating Center for Research in Human Reproduction, Shanghai, China.,Key Laboratory of Contraceptive Drugs and Devices of NPFPC, Shanghai Institute of Planned Parenthood Research, Shanghai, China.,The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.,Institute of Reproduction and Development, Fudan University, Shanghai, China
| |
Collapse
|
75
|
Targeting oncomiRNAs and mimicking tumor suppressor miRNAs: Νew trends in the development of miRNA therapeutic strategies in oncology (Review). Int J Oncol 2016; 49:5-32. [PMID: 27175518 PMCID: PMC4902075 DOI: 10.3892/ijo.2016.3503] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 04/29/2016] [Indexed: 12/16/2022] Open
Abstract
MicroRNA (miRNA or miR) therapeutics in cancer are based on targeting or mimicking miRNAs involved in cancer onset, progression, angiogenesis, epithelial-mesenchymal transition and metastasis. Several studies conclusively have demonstrated that miRNAs are deeply involved in tumor onset and progression, either behaving as tumor-promoting miRNAs (oncomiRNAs and metastamiRNAs) or as tumor suppressor miRNAs. This review focuses on the most promising examples potentially leading to the development of anticancer, miRNA-based therapeutic protocols. The inhibition of miRNA activity can be readily achieved by the use of miRNA inhibitors and oligomers, including RNA, DNA and DNA analogues (miRNA antisense therapy), small molecule inhibitors, miRNA sponges or through miRNA masking. On the contrary, the enhancement of miRNA function (miRNA replacement therapy) can be achieved by the use of modified miRNA mimetics, such as plasmid or lentiviral vectors carrying miRNA sequences. Combination strategies have been recently developed based on the observation that i) the combined administration of different antagomiR molecules induces greater antitumor effects and ii) some anti-miR molecules can sensitize drug-resistant tumor cell lines to therapeutic drugs. In this review, we discuss two additional issues: i) the combination of miRNA replacement therapy with drug administration and ii) the combination of antagomiR and miRNA replacement therapy. One of the solid results emerging from different independent studies is that miRNA replacement therapy can enhance the antitumor effects of the antitumor drugs. The second important conclusion of the reviewed studies is that the combination of anti-miRNA and miRNA replacement strategies may lead to excellent results, in terms of antitumor effects.
Collapse
|
76
|
Li Y, Li J, Sun X, Chen J, Sun X, Zheng J, Chen R. MicroRNA-27a functions as a tumor suppressor in renal cell carcinoma by targeting epidermal growth factor receptor. Oncol Lett 2016; 11:4217-4223. [PMID: 27313769 DOI: 10.3892/ol.2016.4500] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 01/13/2016] [Indexed: 01/27/2023] Open
Abstract
Numerous studies have suggested that microRNAs (miRNAs) are vital in the development of various types of human cancers, including renal cell carcinoma (RCC), and the regulation of tumor progression and invasion. However, the effect of miRNA-27a (miR-27a) on the tumorigenesis of RCC is unclear. The aim of the present study was to investigate the function of miR-27a and identify its possible target genes in RCC cells. In the present study, cell proliferation, migration and invasion and the percentage of apoptotic cells were detected by methylthiazol tetrazolium assays, Annexin V analysis, wound-healing assays and Transwell invasion assays. Western blot analysis was performed to validate the protein expression level and assess whether the epidermal growth factor receptor (EGFR) was a target gene of miR-27a. A tumor xenograft animal model was used to detect the role of miR-27a on RCC cell growth in vivo. The present study demonstrated that miR-27a significantly suppressed human RCC 786-O cell proliferation and induced cell apoptosis. Restoration of miR-27 also resulted in 786-O cell migration and invasion inhibition. Furthermore, upregulated miR-27a attenuated RCC tumor growth in the tumor xenograft animal model. The present results suggested that miR-27a functions as a tumor suppressor in RCC. The western blot analysis assay revealed that EGFR was a novel target of miR-27a. The growth suppression of RCC cells was attributed partly to the downregulation of the cell cycle by ERFR inhibition. The present findings may aid in the understanding of the molecular mechanism of miR-27a in the tumorigenesis of RCC, and may provide novel diagnostic and therapeutic options for RCC.
Collapse
Affiliation(s)
- Yueyan Li
- Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jie Li
- Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiaolei Sun
- Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jiacun Chen
- Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiaoqing Sun
- Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Junnian Zheng
- Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Renfu Chen
- Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
77
|
Zhang R, Zhu L, Zhang L, Xu A, Li Z, Xu Y, He P, Wu M, Wei F, Wang C. PTEN enhances G2/M arrest in etoposide-treated MCF‑7 cells through activation of the ATM pathway. Oncol Rep 2016; 35:2707-14. [PMID: 26986476 DOI: 10.3892/or.2016.4674] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/27/2015] [Indexed: 11/06/2022] Open
Abstract
As an effective tumor suppressor, phosphatase and tensin homolog (PTEN) has attracted the increased attention of scientists. Recent studies have shown that PTEN plays unique roles in the DNA damage response (DDR) and can interact with the Chk1 pathway. However, little is known about how PTEN contributes to DDR through the ATM-Chk2 pathway. It is well-known that etoposide induces G2/M arrest in a variety of cell lines, including MCF-7 cells. The DNA damage-induced G2/M arrest results from the activation of protein kinase ataxia telangiectasia mutated (ATM), followed by the activation of Chk2 that subsequently inactivates CDC25C, resulting in G2/M arrest. In the present study, we assessed the contribution of PTEN to the etoposide-induced G2/M cell cycle arrest. PTEN was knocked down in MCF-7 cells by specific shRNA, and the effects of PTEN on the ATM-Chk2 pathway were investigated through various approaches. The results showed that knockdown of PTEN strongly antagonized ATM activation in response to etoposide treatment, and thereby reduced the phosphorylation level of ATM substrates, including H2AX, P53 and Chk2. Furthermore, depletion of PTEN reduced the etoposide-induced phosphorylation of CDC25C and strikingly compromised etoposide-induced G2/M arrest in the MCF-7 cells. Altogether, we demonstrated that PTEN plays a unique role in etoposide-induced G2/M arrest by facilitating the activation of the ATM pathway, and PTEN was required for the proper activation of checkpoints in response to DNA damage in MCF-7 cells.
Collapse
Affiliation(s)
- Ruopeng Zhang
- Department of Obstetrics and Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Affiliated to Southern Medical University, Longgang, Shenzhen, Guangdong 518028, P.R. China
| | - Li Zhu
- Department of Reproductive Medicine, Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Lirong Zhang
- Department of Reproductive Medicine, Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Anli Xu
- Department of Reproductive Medicine, Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Zhengwei Li
- Clinical Medicine College of Dali University, Dali, Yunnan 671000, P.R. China
| | - Yijuan Xu
- Clinical Medicine College of Dali University, Dali, Yunnan 671000, P.R. China
| | - Pei He
- Clinical Medicine College of Dali University, Dali, Yunnan 671000, P.R. China
| | - Maoqing Wu
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Fengxiang Wei
- The Genetics Laboratory, Shenzhen Longgang District Maternity and Child Healthcare Hospital, Longgang, Shenzhen, Guangdong 518028, P.R. China
| | - Chenhong Wang
- Department of Obstetrics and Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Affiliated to Southern Medical University, Longgang, Shenzhen, Guangdong 518028, P.R. China
| |
Collapse
|
78
|
Abstract
Human eukaryotic prohibitin (prohibitin-1 and prohibitin-2) is a membrane protein with different cellular localizations. It is involved in multiple cellular functions, including energy metabolism, proliferation, apoptosis, and senescence. The subcellular localization of prohibitin may determine its functions. Membrane prohibitin regulate the cellular signaling of membrane transport, nuclear prohibitin control transcription activation and the cell cycle, and mitochondrial prohibitin complex stabilize the mitochondrial genome and modulate mitochondrial dynamics, mitochondrial morphology, mitochondrial biogenesis, and the mitochondrial intrinsic apoptotic pathway. Moreover, prohibitin can translocates into the nucleus or the mitochondria under apoptotic signals and the subcellular shuttling of prohibitin is necessary for apoptosis process. Apoptosis is the process of programmed cell death that is important for the maintenance of normal physiological functions. Consequently, any alteration in the content, post-transcriptional modification (i.e. phosphorylation) or the nuclear or mitochondrial translocation of prohibitin may influence cell fate. Understanding the mechanisms of the expression and regulation of prohibitin may be useful for future research. This review provides an overview of the multifaceted and essential roles played by prohibitin in the regulation of cell survival and apoptosis.
Collapse
Affiliation(s)
- Ya-Ting Peng
- Department of Respiratory Medicine, Respiratory Disease Research Institute, Second XiangYa Hospital of Central South University, Changsha, 410011, People's Republic of China
| | | | | | | |
Collapse
|
79
|
Ceder Y. Non-coding RNAs in Prostate Cancer: From Discovery to Clinical Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 886:155-170. [PMID: 26659491 DOI: 10.1007/978-94-017-7417-8_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Prostate cancer is a heterogeneous disease for which the molecular mechanisms are still not fully elucidated. Prostate cancer research has traditionally focused on genomic and epigenetic alterations affecting the proteome, but over the last decade non-coding RNAs, especially microRNAs, have been recognized to play a key role in prostate cancer progression. A considerable number of individual microRNAs have been found to be deregulated in prostate cancer and their biological significance elucidated in functional studies. This review will delineate the current advances regarding the involvement of microRNAs and their targets in prostate cancer biology as well as their potential usage in the clinical management of the disease. The main focus will be on microRNAs contributing to initiation and progression of prostate cancer, including androgen signalling, cellular plasticity, stem cells biology and metastatic processes. To conclude, implications on potential future microRNA-based therapeutics based on the recent advances regarding the interplay between microRNAs and their targets are discussed.
Collapse
Affiliation(s)
- Yvonne Ceder
- Translational Cancer Research, Lund University, Medicon Village, Building 404:A3, 223 81, Lund, Sweden.
| |
Collapse
|
80
|
Koushyar S, Jiang WG, Dart DA. Unveiling the potential of prohibitin in cancer. Cancer Lett 2015; 369:316-22. [PMID: 26450374 DOI: 10.1016/j.canlet.2015.09.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/16/2015] [Accepted: 09/19/2015] [Indexed: 12/18/2022]
Abstract
Recently, research has shed new light on the role of Prohibitin (PHB) in cancer pathogenesis across an array of cancer types. Important mechanisms for PHB have been unveiled in several cancers, especially with regard to the androgen independent state of prostate cancer (PC) and oestrogen dependent breast cancer. However, PHB is often overlooked due to its complex but subtle roles within the cell. Having gathered both historical and current research exploring PHB's role in different cancer types including prostate and breast, here we aim to pair this information with its molecular properties in the hope of translating this information into a clinical perspective, thus discussing its possible use in future cancer therapy.
Collapse
Affiliation(s)
- Sarah Koushyar
- Cardiff China Medical Research Collaborative (CCMRC), Cardiff University, School of Medicine, Henry Welcome Building, Heath Park, Cardiff CF14 4XN, UK.
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative (CCMRC), Cardiff University, School of Medicine, Henry Welcome Building, Heath Park, Cardiff CF14 4XN, UK
| | - D Alwyn Dart
- Cardiff China Medical Research Collaborative (CCMRC), Cardiff University, School of Medicine, Henry Welcome Building, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
81
|
Abente EJ, Subramanian M, Ramachandran V, Najafi-Shoushtari SH. MicroRNAs in obesity-associated disorders. Arch Biochem Biophys 2015; 589:108-19. [PMID: 26416722 DOI: 10.1016/j.abb.2015.09.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 02/08/2023]
Abstract
The emergence of a worldwide obesity epidemic has dramatically increased the prevalence of insulin resistance and metabolic syndrome, predisposing individuals to a greater risk for the development of non-alcoholic fatty liver disease, type II diabetes and atherosclerotic cardiovascular diseases. Current available pharmacological interventions combined with diet and exercise-based managements are still poorly effective for weight management, likely in part due to an incomplete understanding of regulatory mechanisms and pathways contributing to the systemic metabolic abnormalities under disturbed energy homeostasis. MicroRNAs, small non-coding RNAs that regulate posttranscriptional gene expression, have been increasingly described to influence shifts in metabolic pathways under various obesity-related disease settings. Here we review recent discoveries of the mechanistic role that microRNAs play in regulating metabolic functions in liver and adipose tissues involved in obesity associated disorders, and briefly discusses the potential candidates that are being pursued as viable therapeutic targets.
Collapse
Affiliation(s)
- Eugenio J Abente
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York 10021, NY, USA; Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Murugan Subramanian
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York 10021, NY, USA; Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Vimal Ramachandran
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York 10021, NY, USA; Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - S Hani Najafi-Shoushtari
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York 10021, NY, USA; Weill Cornell Medical College-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar.
| |
Collapse
|
82
|
Romero-Cordoba SL, Salido-Guadarrama I, Rodriguez-Dorantes M, Hidalgo-Miranda A. miRNA biogenesis: biological impact in the development of cancer. Cancer Biol Ther 2015; 15:1444-55. [PMID: 25482951 PMCID: PMC4622859 DOI: 10.4161/15384047.2014.955442] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
microRNAs (miRNAs) are non coding RNAs with different biological functions and pathological implications. Given their role as post-transcriptional gene expression regulators, they are involved in several important physiological processes like development, cell differentiation and cell signaling. miRNAs act as modulators of gene expression programs in different diseases, particularly in cancer, where they act through the repression of genes which are critical for carcinogenesis. The expression level of mature miRNAs is the result of a fine mechanism of biogenesis, carried out by different enzymatic complexes that exert their function at transcriptional and post-transcriptional levels. In this review, we will focus our discussion on the alterations in the miRNA biogenesis machinery, and its impact on the establishment and development of cancer programs.
Collapse
Key Words
- Ago2, Argonaute 2 protein
- Ars2, Arsenic Resistance protein 2
- DGCR8, DiGeorge syndrome Critical Region 8 protein
- EMT, epithelial–mesenchymal transition
- KSRP, KH-type splicing regulatory protein
- MK2, MAPK-activated protein kinase 2
- PABP, poly(A)-binding protein
- PACT, kinase R–activating protein
- PRC2, Polycomb repressor complex
- RISC, RNA-induced silencing complex
- TRBP, TAR RNA binding protein
- TUT4, terminal uridine transferase-4
- XPO5, exportin 5
- cancer
- cellular signaling
- circRNA, circular RNA
- hnRNPs, heterogeneous nuclear ribonucleoproteins
- miRNA biogenesis
- miRNAs, microRNAs
Collapse
|
83
|
Cathcart P, Lucchesi W, Ottaviani S, De Giorgio A, Krell J, Stebbing J, Castellano L. Noncoding RNAs and the control of signalling via nuclear receptor regulation in health and disease. Best Pract Res Clin Endocrinol Metab 2015; 29:529-43. [PMID: 26303081 DOI: 10.1016/j.beem.2015.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nuclear receptors belong to a superfamily of proteins that play central roles in human biology, orchestrating a large variety of biological functions in both health and disease. Understanding the interactions and regulatory pathways of NRs will allow development of potential therapeutic interventions for a multitude of disease processes. Non-coding RNAs have recently been discovered to have significant interactions with NR signalling pathways via a variety of biological connections. This review summarises the known interactions between ncRNAs and the NR superfamily in health, embryogenesis and a plethora of human diseases.
Collapse
Affiliation(s)
- Paul Cathcart
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Walter Lucchesi
- School of Pharmacy, University of Reading, Whiteknights Reading Berks RG6 6AP, UK
| | - Silvia Ottaviani
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Alex De Giorgio
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Jonathan Krell
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Leandro Castellano
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK.
| |
Collapse
|
84
|
Assessment of association between genetic variants in microRNA genes hsa-miR-499, hsa-miR-196a2 and hsa-miR-27a and prostate cancer risk in Serbian population. Exp Mol Pathol 2015; 99:145-50. [PMID: 26112096 DOI: 10.1016/j.yexmp.2015.06.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/18/2015] [Indexed: 12/30/2022]
Abstract
Due to their potentially functional significance, genetic variants within microRNA genes have been recognized as candidates for cancer-related genetic biomarkers. Among the most extensively studied so far are rs3746444, rs11614913 and rs895819. Nevertheless, only few previous studies in Asian population analyzed the association of rs3746444 and rs11614913 with prostate cancer (PCa) risk, while rs895819 was not evaluated in relation to this issue. The aim of this study was to assess the possible association between these genetic variants and PCa risk and progression in Serbian population. 355 samples of peripheral blood were obtained from the patients with PCa and 353 samples from patients with benign prostatic hyperplasia (BPH). 312 volunteers derived from general population who gave samples of buccal swabs were included in the control group. Genotyping of rs3746444, rs11614913 and rs895819 was performed by using PCR-RFLP method, HRM analysis and allele-specific PCR, respectively. Allelic and genotypic associations were evaluated by unconditional linear (for serum PSA level in PCa patients) and logistic regression method with adjustment for age. Minor allele C of rs895819 was found to be associated with the increased risk of developing PCa under dominant (P=0.035; OR=1.38, 95%CI 1.02-1.86) and overdominant (P=0.04; OR=1.37, 95%CI 1.01-1.85) genetic model. Same genetic variant was found to be associated with the clinical stage of localized PCa, as well as with the presence of distant metastases. Allele G of rs3746444 was also shown to be associated with the decreased risk of PCa progression. According to our data, rs3746444 qualifies for a genetic variant potentially associated with PCa aggressiveness in Serbian population. Furthermore, our study provided the first evidence of association between rs895819 and PCa risk, as well as for its genetic association with the presence of distant metastases among PCa patients.
Collapse
|
85
|
MicroRNA-27a Contributes to Rhabdomyosarcoma Cell Proliferation by Suppressing RARA and RXRA. PLoS One 2015; 10:e0125171. [PMID: 25915942 PMCID: PMC4410939 DOI: 10.1371/journal.pone.0125171] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 03/21/2015] [Indexed: 12/21/2022] Open
Abstract
Background Rhabdomyosarcomas (RMS) are rare but very aggressive childhood tumors that arise as a consequence of a regulatory disruption in the growth and differentiation pathways of myogenic precursor cells. According to morphological criteria, there are two major RMS subtypes: embryonal RMS (ERMS) and alveolar RMS (ARMS) with the latter showing greater aggressiveness and metastatic potential with respect to the former. Efforts to unravel the complex molecular mechanisms underlying RMS pathogenesis and progression have revealed that microRNAs (miRNAs) play a key role in tumorigenesis. Methodology/Principal Findings The expression profiles of 8 different RMS cell lines were analyzed to investigate the involvement of miRNAs in RMS. The miRNA population from each cell line was compared to a reference sample consisting of a balanced pool of total RNA extracted from those 8 cell lines. Sixteen miRNAs whose expression discriminates between translocation-positive ARMS and negative RMS were identified. Attention was focused on the role of miR-27a that is up-regulated in the more aggressive RMS cell lines (translocation-positive ARMS) in which it probably acts as an oncogene. MiR-27a overexpressing cells showed a significant increase in their proliferation rate that was paralleled by a decrease in the number of cells in the G1 phase of the cell cycle. It was possible to demonstrate that miR-27a is implicated in cell cycle control by targeting the retinoic acid alpha receptor (RARA) and retinoic X receptor alpha (RXRA). Conclusions Study results have demonstrated that miRNA expression signature profiling can be used to classify different RMS subtypes and suggest that miR-27a may have a therapeutic potential in RMS by modulating the expression of retinoic acid receptors.
Collapse
|
86
|
Zheng R, Jiang Y, Wang X. [The establishment of Raji drug-resistant cell line and analyses of prohibitin and miR-27a expression]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:326-30. [PMID: 25916297 PMCID: PMC7342615 DOI: 10.3760/cma.j.issn.0253-2727.2015.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To establish Raji adriamycin (ADR)-resistant cell lines and analysis the expression of mitochondria Prohibitin (PHB) and microRNA-27a (miR-27a), as well as discuss its clinical significance. METHODS Built ADR-resistant Raji cells, detected their resistant index and drug-resistant spectrum and stability, observed their morphology and growth characteristics in general; evaluated the expression of phb mRNA and miR-27a in ADR-resistant cells (Raji/A) and sensitive cells (Raji/S) via real-time quantitative polymerase chain reaction (RT-PCR). RESULTS The ADR-resistant Raji cell lines were built; expression of PHB1 mRNA in Raji/A was evidently higher than in Raji/S (P<0.05), the expressing difference of PHB2 mRNA in Raji/A and Raji/S was statistically meaningless (P>0.05), the expression of miR-27a in Raji/A was much higher than in Raji/S (P<0.05). CONCLUSION By building the experimental model of Raji ADR-resistant cell lines, high expression level of PHB1 and miR-27a were detected in the cell lines, indicating that PHB1 and miR-27a may be associated with ADR-resistance of Raji cells.
Collapse
Affiliation(s)
- Rongli Zheng
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Ji'nan 250021, China
| | - Yujie Jiang
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Ji'nan 250021, China
| | - Xin Wang
- Department of Hematology, Provincial Hospital Affiliated to Shandong University, Ji'nan 250021, China
| |
Collapse
|
87
|
Ayub SG, Kaul D, Ayub T. Microdissecting the role of microRNAs in the pathogenesis of prostate cancer. Cancer Genet 2015; 208:289-302. [PMID: 26004033 DOI: 10.1016/j.cancergen.2015.02.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 02/16/2015] [Accepted: 02/21/2015] [Indexed: 12/18/2022]
Abstract
MicroRNAs (miRNAs) are naturally occurring, small, non-coding RNA molecules that post-transcriptionally regulate the expression of a large number of genes involved in various biological processes, either through mRNA degradation or through translation inhibition. Since the discovery of miRNAs, a vast amount of research has implicated the deregulated expression of miRNAs in different malignancies, including prostate cancer (PCa). Different miRNA expression profiles are reportedly associated with the development, progression, and emergence of castration-resistant PCa (CRPC), suggesting their use in the diagnosis, prognosis, and development of anti-cancer treatment models directed against this disease. However, before their exploitation in terms of therapeutics, a thorough understanding and in-depth mechanistic studies of these miRNAs and the gene networks they orchestrate are necessary for ascertaining their definitive role in the development and progression of PCa. This review attempts to extensively summarize the current knowledge of aberrantly expressed miRNAs and their mode of action in PCa, while highlighting the existing discrepancies and future research warranted.
Collapse
Affiliation(s)
- Shiekh Gazalla Ayub
- Department of Experimental Medicine and Biotechnology, Post-Graduate Institute of Medical Sciences and Research, Chandigarh, India.
| | - Deepak Kaul
- Department of Experimental Medicine and Biotechnology, Post-Graduate Institute of Medical Sciences and Research, Chandigarh, India
| | - Taha Ayub
- Department of Social and Preventive Medicine, Government Medical College, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
88
|
Role of MicroRNAs in Prostate Cancer Pathogenesis. Clin Genitourin Cancer 2015; 13:261-270. [PMID: 25733057 DOI: 10.1016/j.clgc.2015.01.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/09/2015] [Accepted: 01/16/2015] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa) remains the most commonly diagnosed malignant tumor in men, and is the second highest cause of cancer mortality after lung tumors in the United States. Accumulating research indicates that microRNAs (miRNAs) are increasingly being implicated in PCa. miRNAs are conserved small noncoding RNAs that control gene expression posttranscriptionally. Recent profiling research suggests that miRNAs are aberrantly expressed in PCa, and these have been implicated in the regulation of apoptosis, cell cycle, epithelial to mesenchymal transition, PCa stem cells, and androgen receptor pathway. All of these might provide the basis for new approaches for PCa. Here, we review current findings regarding miRNA research in PCa to provide a strong basis for future study aimed at promising contributions of miRNA in PCa.
Collapse
|
89
|
Li WQ, Yu HY, Zhong NZ, Hou LJ, Li YM, He J, Liu HM, Xia CY, Lu YC. miR‑27a suppresses the clonogenic growth and migration of human glioblastoma multiforme cells by targeting BTG2. Int J Oncol 2015; 46:1601-8. [PMID: 25626081 DOI: 10.3892/ijo.2015.2843] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 11/28/2014] [Indexed: 11/06/2022] Open
Abstract
miR-27a and BTG2 are implicated in gliomagenesis and glioma progression. However, hitherto, a link between miR-27a and BTG2 in glioma has not been reported. In the present study, we investigated the effects of miR-27a on the proliferation and invasiveness of glioblastoma cells in vitro and in a mouse xenograft model and further studied the relation between miR‑27a expression and its target gene BTG2, which was identified by computation prediction algorithms. Our MTT and clonogenic assays showed that miR-27a overexpression significantly increased the clonogenic growth of glioblastoma U87MG and U251MG cells. The Transwell assays further revealed that miR-27a overexpression markedly increased the number of migrated U87MG and U251MG cells. TargetScan and other prediction algorithms identified BTG2 as a target gene of miR-27a, which was confirmed by EGFP reporter and immunoblotting assays showing an inverse relation between miR-27a expression and endogenous BTG2 expression. BTG2 overexpression also increased the proliferation and invasiveness of glioblastoma cells and BTG2 functioned downstream of miR-27a in modulating the proliferation and migration of glioblastoma cells. In conclusion, miR-27a modulates human glioblastoma growth and invasion by targeting BTG2.
Collapse
Affiliation(s)
- Wei-Qing Li
- Department of Pathology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Hong-Yu Yu
- Department of Pathology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Nan-Zhe Zhong
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Li-Jun Hou
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yi-Ming Li
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Jin He
- Department of Pathology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Hui-Min Liu
- Department of Pathology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Chun-Yan Xia
- Department of Pathology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yi-Cheng Lu
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
90
|
Jackson BL, Grabowska A, Ratan HL. MicroRNA in prostate cancer: functional importance and potential as circulating biomarkers. BMC Cancer 2014; 14:930. [PMID: 25496077 PMCID: PMC4295407 DOI: 10.1186/1471-2407-14-930] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 12/01/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND This non-systematic review article aims to summarise the progress made in understanding the functional consequences of microRNA (miRNA) dysregulation in prostate cancer development, and the identification of potential miRNA targets as serum biomarkers for diagnosis or disease stratification. RESULTS A number of miRNAs have been shown to influence key cellular processes involved in prostate tumourigenesis, including apoptosis-avoidance, cell proliferation and migration and the androgen signalling pathway. An overlapping group of miRNAs have shown differential expression in the serum of patients with prostate cancer of varying stages compared with unaffected individuals. The majority of studies thus far however, involve small numbers of patients and have shown variable and occasionally conflicting results CONCLUSION MiRNAs show promise as potential circulating biomarkers in prostate cancer, but larger prospective studies are required to validate particular targets and better define their clinical utility.
Collapse
Affiliation(s)
- Benjamin L Jackson
- />Unit of Cancer Biology, University of Nottingham, Queens Medical Centre, Derby Road, Nottingham, NG7 2UH England
| | - Anna Grabowska
- />Unit of Cancer Biology, University of Nottingham, Queens Medical Centre, Derby Road, Nottingham, NG7 2UH England
| | - Hari L Ratan
- />Unit of Cancer Biology, University of Nottingham, Queens Medical Centre, Derby Road, Nottingham, NG7 2UH England
- />Department of Urology, Nottingham City Hospital, Hucknall Road, Nottingham, NG5 1PB England
| |
Collapse
|
91
|
Jiang Y, Duan Y, Zhou H. MicroRNA-27a directly targets KRAS to inhibit cell proliferation in esophageal squamous cell carcinoma. Oncol Lett 2014; 9:471-477. [PMID: 25436011 PMCID: PMC4246996 DOI: 10.3892/ol.2014.2701] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 09/22/2014] [Indexed: 01/27/2023] Open
Abstract
MicroRNAs (miRNAs) are a type of small non-coding RNA that negatively regulate gene expression levels by binding to the 3′-untranslated region of specific target mRNAs. To investigate the role of miR-27a in esophageal squamous cell carcinoma (ESCC), TargetScan software was used to predict the target gene of miR-27a. Kirsten rat sarcoma viral oncogene homolog (KRAS), which has been implicated as a regulator of cell proliferation, differentiation and transformation, was identified as a potential target gene of miR-27a and, thus, was the focus of the present study. Luciferase activity in cells transfected with miR-27a mimics was 48% lower when compared with that of the miRNA-negative control. Furthermore, expression levels of the K-ras protein were reduced by ≤50% in cells cotransfected with an expression vector containing miR-27a and miR-27a binding sequences, when compared with the control. The expression level of miR-27a was significantly lower in ESCC cell lines and tissues when compared with healthy esophageal epithelial cells and tissues. However, the expression level of the target gene, KRAS was upregulated and ESCC cell proliferation was significantly inhibited following miR-27a mimic or small interfering K-ras transfection. In conclusion, the present study demonstrated that the expression level of miR-27a was low in ESCC and that miR-27a directly targets the KRAS gene, resulting in inhibited cell proliferation in esophageal cancer.
Collapse
Affiliation(s)
- Yuzhi Jiang
- Department of Radiology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Yuting Duan
- Department of Radiology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Haibin Zhou
- Department of Thoracic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
92
|
Ottaviani S, de Giorgio A, Harding V, Stebbing J, Castellano L. Noncoding RNAs and the control of hormonal signaling via nuclear receptor regulation. J Mol Endocrinol 2014; 53:R61-70. [PMID: 25062739 DOI: 10.1530/jme-14-0134] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Despite its identification over 100 years ago, new discoveries continue to add to the complexity of the regulation of the endocrine system. Today the nuclear receptors (NRs) that play such a pivotal role in the extensive communication networks of hormones and gene expression remain an area of intense research. By orchestrating core processes, from metabolism to organismal development, the gene expression programs they control are dependent on their cellular context, their own levels, and those of numerous co-regulatory proteins. A previously unknown component of these networks, noncoding RNAs (ncRNAs) are now recognized as potent regulators of NR signaling, influencing receptor and co-factor levels and functions while being reciprocally regulated by the NRs themselves. This review explores the regulation enacted by microRNAs and long ncRNAs on NR function, using representative examples to show the varied roles of ncRNAs, in turn producing significant effects on the NR functional network in health and disease.
Collapse
Affiliation(s)
- Silvia Ottaviani
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Alexander de Giorgio
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Victoria Harding
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Justin Stebbing
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| | - Leandro Castellano
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, London W12 0NN, UK
| |
Collapse
|
93
|
Fletcher CE, Dart DA, Bevan CL. Interplay between steroid signalling and microRNAs: implications for hormone-dependent cancers. Endocr Relat Cancer 2014; 21:R409-29. [PMID: 25062737 DOI: 10.1530/erc-14-0208] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hormones are key drivers of cancer development. To date, interest has largely been focussed on the classical model of hormonal gene regulation, but there is increasing evidence for a role of hormone signalling pathways in post-translational regulation of gene expression. In particular, a complex and dynamic network of bi-directional interactions with microRNAs (miRs) at all stages of biogenesis and during target gene repression is emerging. miRs, which act mainly by negatively regulating gene expression through association with 3'-UTRs of mRNA species, are increasingly understood to be important in development, normal physiology and pathogenesis. Given recent demonstrations of altered miR profiles in a diverse range of cancers, their ability to function as oncogenes or tumour suppressors, and hormonal regulation of miRs, understanding mechanisms by which miRs are generated and regulated is vitally important. miRs are transcribed by RNA polymerase II and then processed in the nucleus by the Drosha-containing Microprocessor complex and in the cytoplasm by Dicer, before mature miRs are incorporated into the RNA-induced silencing complex. It is increasingly evident that multiple cellular signalling pathways converge upon the miR biogenesis cascade, adding further layers of regulatory complexity to modulate miR maturation. This review summarises recent advances in identification of novel components and regulators of the Microprocessor and Dicer complexes, with particular emphasis on the role of hormone signalling pathways in regulating their activity. Understanding hormone regulation of miR production and how this is perturbed in cancer are critical for the development of miR-based therapeutics and biomarkers.
Collapse
Affiliation(s)
- Claire E Fletcher
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London W12 0NN, UKCardiff University School of MedicineCardiff University Peking University Cancer Institute, Cardiff CF14 4XN, UK
| | - D Alwyn Dart
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London W12 0NN, UKCardiff University School of MedicineCardiff University Peking University Cancer Institute, Cardiff CF14 4XN, UK Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London W12 0NN, UKCardiff University School of MedicineCardiff University Peking University Cancer Institute, Cardiff CF14 4XN, UK
| | - Charlotte L Bevan
- Department of Surgery and CancerImperial College London, Imperial Centre for Translational and Experimental Medicine, Du Cane Road, London W12 0NN, UKCardiff University School of MedicineCardiff University Peking University Cancer Institute, Cardiff CF14 4XN, UK
| |
Collapse
|
94
|
Kroiss A, Vincent S, Decaussin-Petrucci M, Meugnier E, Viallet J, Ruffion A, Chalmel F, Samarut J, Allioli N. Androgen-regulated microRNA-135a decreases prostate cancer cell migration and invasion through downregulating ROCK1 and ROCK2. Oncogene 2014; 34:2846-55. [PMID: 25065599 DOI: 10.1038/onc.2014.222] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 06/12/2014] [Accepted: 06/15/2014] [Indexed: 12/20/2022]
Abstract
Androgen signaling, via the androgen receptor (AR), is crucial in mediating prostate cancer (PCa) initiation and progression. Identifying new downstream effectors of the androgens/AR pathway will allow a better understanding of these mechanisms and could reveal novel biomarkers and/or therapeutic agents to improve the rate of patient survival. We compared the microRNA expression profiles in androgen-sensitive LNCaP cells stimulated or not with 1 nM R1881 by performing a high-throughput reverse transcriptase-quantitative PCR and found that miR-135a was upregulated. After androgen stimulation, we showed that AR directly activates the transcription of miR-135a2 gene by binding to an androgen response element in the promoter region. Our findings identify miR-135a as a novel effector in androgens/AR signaling. Using xenograft experiments in chick embryos and adult male mice, we showed that miR-135a overexpression decreases in vivo invasion abilities of prostate PC-3 cells. Through in vitro wound-healing migration and invasion assays, we demonstrated that this effect is mediated through downregulating ROCK1 and ROCK2 expression, two genes that we characterized as miR-135a direct target genes. In human surgical samples from prostatectomy, we observed that miR-135a expression was lower in tumoral compared with paired adjacent normal tissues, mainly in tumors classified with a high Gleason score (⩾8). Moreover, miR-135a expression is lower in invasive tumors, showing extraprostatic extension, as compared with intraprostatic localized tumors. In tumor relative to normal glands, we also showed a more frequently higher ROCK1 protein expression determined using a semi-quantitative immunohistochemistry analysis. Therefore, in tumor cells, the lower miR-135a expression could lead to a higher ROCK1 protein expression, which could explain their invasion abilities. The highlighted relationship between miR-135a expression level and the degree of disease aggressiveness suggests that miR-135a may be considered as a prognostic marker in human PCa.
Collapse
Affiliation(s)
- A Kroiss
- Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Institut de Génomique Fonctionnelle de Lyon (IGFL), Lyon, France
| | - S Vincent
- Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Institut de Génomique Fonctionnelle de Lyon (IGFL), Lyon, France
| | - M Decaussin-Petrucci
- 1] Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Institut de Génomique Fonctionnelle de Lyon (IGFL), Lyon, France [2] Faculté de Médecine et de Maïeutique Lyon-Sud-Charles Mérieux, Université Claude Bernard Lyon 1, Pierre-Bénite, France [3] Service d'Anatomie et de Cytologie Pathologiques, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - E Meugnier
- Laboratoire CarMen, INSERM U1060, INRA1362, Université Claude Bernard Lyon 1, Faculté de Médecine et de Maïeutique Lyon-Sud-Charles Mérieux, Oullins, France
| | - J Viallet
- In Ovo, Institut Albert Bonniot, CRI INSERM/UJF U823, Université de Grenoble, La Tronche Cedex, France
| | - A Ruffion
- 1] Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Institut de Génomique Fonctionnelle de Lyon (IGFL), Lyon, France [2] Faculté de Médecine et de Maïeutique Lyon-Sud-Charles Mérieux, Université Claude Bernard Lyon 1, Pierre-Bénite, France [3] Service d'Urologie, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - F Chalmel
- GERHM, INSERM U1085-Irset, Université Rennes 1, Rennes, France
| | - J Samarut
- 1] Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Institut de Génomique Fonctionnelle de Lyon (IGFL), Lyon, France [2] Faculté de Médecine et de Maïeutique Lyon-Sud-Charles Mérieux, Université Claude Bernard Lyon 1, Pierre-Bénite, France [3] Service de Biochimie Biologie Moleculaire Sud, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - N Allioli
- 1] Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Institut de Génomique Fonctionnelle de Lyon (IGFL), Lyon, France [2] Institut des Sciences Pharmaceutiques et Biologiques (ISPB)-Faculté de Pharmacie de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
95
|
Teng Y, Litchfield LM, Ivanova MM, Prough RA, Clark BJ, Klinge CM. Dehydroepiandrosterone-induces miR-21 transcription in HepG2 cells through estrogen receptor β and androgen receptor. Mol Cell Endocrinol 2014; 392:23-36. [PMID: 24845419 PMCID: PMC4074919 DOI: 10.1016/j.mce.2014.05.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 05/02/2014] [Accepted: 05/09/2014] [Indexed: 12/15/2022]
Abstract
Although oncomiR miR-21 is highly expressed in liver and overexpressed in hepatocellular carcinoma (HCC), its regulation is uncharacterized. We examined the effect of physiologically relevant nanomolar concentrations of dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEA-S) on miR-21 expression in HepG2 human hepatoma cells. 10nM DHEA and DHEA-S increase pri-miR-21 transcription in HepG2 cells. Dietary DHEA increased miR-21 in vivo in mouse liver. siRNA and inhibitor studies suggest that DHEA-S requires desulfation for activity and that DHEA-induced pri-miR-21 transcription involves metabolism to androgen and estrogen receptor (AR and ER) ligands. Activation of ERβ and AR by DHEA metabolites androst-5-ene-3,17-dione (ADIONE), androst-5-ene-3β,17β-diol (ADIOL), dihydrotestosterone (DHT), and 5α-androstane-3β,17β-diol (3β-Adiol) increased miR-21 transcription. DHEA-induced miR-21 increased cell proliferation and decreased Pdcd4 protein, a bona fide miR-21. Estradiol (E2) inhibited miR-21 expression via ERα. DHEA increased ERβ and AR recruitment to the miR-21 promoter within the VMP1/TMEM49 gene, with possible significance in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yun Teng
- Department of Biochemistry & Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Lacey M Litchfield
- Department of Biochemistry & Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Margarita M Ivanova
- Department of Biochemistry & Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Russell A Prough
- Department of Biochemistry & Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Barbara J Clark
- Department of Biochemistry & Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Carolyn M Klinge
- Department of Biochemistry & Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| |
Collapse
|
96
|
ChunJiao S, Huan C, ChaoYang X, GuoMei R. Uncovering the roles of miRNAs and their relationship with androgen receptor in prostate cancer. IUBMB Life 2014; 66:379-86. [PMID: 24979663 PMCID: PMC4140600 DOI: 10.1002/iub.1281] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 06/02/2014] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCa) is the second most commonly occurring malignant tumor in Europe and America. Normal and neoplastic growth of prostate gland are dependent on androgen receptor (AR) expression and function. PCa is driven by androgen and its receptor, and they continue to be the key drivers of castration-resistant prostate cancer (CRPC). CRPC is the terminal stage of PCa and seriously jeopardizes the patient's quality of life and lifespan. miRNAs are small noncoding RNAs, 18–25 nt in length that destabilize mRNA or repress protein synthesis by interacting with the 3′-untranslated regions (3'-UTR) of target mRNAs. miRNAs can regulate AR or be regulated by AR and then affect various signaling pathways related to cellular functions and tumor processes. In this review, we focus on the relationship between miRNAs and AR in PCa and elucidate their roles in the induction of malignant changes in PCa. © 2014 The Authors IUBMB Life published by Wiley Periodicals, Inc. on behalf of International Union of Biochemistry and Molecular Biology, 66(6):379–386, 2014
Collapse
Affiliation(s)
- Song ChunJiao
- Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang Province, China
| | | | | | | |
Collapse
|
97
|
Sita-Lumsden A, Fletcher CE, Dart DA, Brooke GN, Waxman J, Bevan CL. Circulating nucleic acids as biomarkers of prostate cancer. Biomark Med 2014; 7:867-77. [PMID: 24266819 DOI: 10.2217/bmm.13.104] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer, the most common cancer of western men, requires new biomarkers, especially given that the benefits of PSA testing remain uncertain. Nucleic acids can now be accurately and sensitively detected in human blood. Over the last decade, investigations into utility of circulating cell-free miRNA, DNA and mRNA as novel biomarkers have expanded exponentially. In the near future, they may be routinely used to accurately diagnose cancers, stratify indolent from aggressive disease and inform treatment decisions. However, advancement of such tests into clinical settings is hampered by technical problems with assay specificity and sensitivity, and small study sizes. This review highlights the different forms of circulating nucleic acids and those that show the most potential as viable biomarkers for prostate cancer.
Collapse
Affiliation(s)
- Ailsa Sita-Lumsden
- Department of Surgery & Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, London W12 0NN, UK
| | | | | | | | | | | |
Collapse
|
98
|
Seven M, Karatas OF, Duz MB, Ozen M. The role of miRNAs in cancer: from pathogenesis to therapeutic implications. Future Oncol 2014; 10:1027-48. [DOI: 10.2217/fon.13.259] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
ABSTRACT: Cancer is still one of the dominating causes of deaths worldwide, although there have been important enhancements for detection and diagnosis of cancer recently. miRNAs are shown to participate in carcinogenesis of several types of tumors and their aberrant expression of miRNAs has been detected in cell lines, xenografts and clinical samples. miRNAs are thought to target and modulate the expression of more than 60% of human genes, which makes the expressional regulation by miRNAs the most abundant post-transcriptional regulation mode. Here, we have reviewed the most current literature to shed a light on the functions of miRNAs on human carcinogenesis. Possible roles of miRNAs in oncogenesis through both genetic and epigenetic changes occurring during cancer initiation, progression, invasion or metastasis are summarized.
Collapse
Affiliation(s)
- Mehmet Seven
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey
| | - Omer Faruk Karatas
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey
- Molecular Biology & Genetics Department, Erzurum Technical University, Erzurum, Turkey
| | - Mehmet Bugrahan Duz
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey
| | - Mustafa Ozen
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey
- Department of Pathology & Immunology Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
99
|
Farooqi AA, Hou MF, Chen CC, Wang CL, Chang HW. Androgen receptor and gene network: Micromechanics reassemble the signaling machinery of TMPRSS2-ERG positive prostate cancer cells. Cancer Cell Int 2014; 14:34. [PMID: 24739220 PMCID: PMC4002202 DOI: 10.1186/1475-2867-14-34] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 04/08/2014] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is a gland tumor in the male reproductive system. It is a multifaceted and genomically complex disease. Transmembrane protease, serine 2 and v-ets erythroblastosis virus E26 homolog (TMPRSS2-ERG) gene fusions are the common molecular signature of prostate cancer. Although tremendous advances have been made in unraveling various facets of TMPRSS2-ERG-positive prostate cancer, many research findings must be sequentially collected and re-interpreted. It is important to understand the activation or repression of target genes and proteins in response to various stimuli and the assembly in signal transduction in TMPRSS2-ERG fusion-positive prostate cancer cells. Accordingly, we divide this multi-component review ofprostate cancer cells into several segments: 1) The role of TMPRSS2-ERG fusion in genomic instability and methylated regulation in prostate cancer and normal cells; 2) Signal transduction cascades in TMPRSS2-ERG fusion-positive prostate cancer; 3) Overexpressed genes in TMPRSS2-ERG fusion-positive prostate cancer cells; 4) miRNA mediated regulation of the androgen receptor (AR) and its associated protein network; 5) Quantitative control of ERG in prostate cancer cells; 6) TMPRSS2-ERG encoded protein targeting; In conclusion, we provide a detailed understanding of TMPRSS2-ERG fusion related information in prostate cancer development to provide a rationale for exploring TMPRSS2-ERG fusion-mediated molecular network machinery.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, 35 Km Ferozepur Road, Lahore, Pakistan
| | - Ming-Feng Hou
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan ; Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Chien-Chi Chen
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | - Chun-Lin Wang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | - Hsueh-Wei Chang
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan ; Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan ; Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
100
|
Yu JJ, Wu YX, Zhao FJ, Xia SJ. miR-96 promotes cell proliferation and clonogenicity by down-regulating of FOXO1 in prostate cancer cells. Med Oncol 2014; 31:910. [PMID: 24633705 DOI: 10.1007/s12032-014-0910-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 03/04/2014] [Indexed: 12/11/2022]
Abstract
The present study aimed to investigate the biological functions of miR-96 in the processes of proliferation and clonogenicity in the prostate cancer cells. miR-96 was identified to be markedly up-regulated in prostate cancer cell and cancer tissues compared with normal prostate cell and normal prostate tissues by microarray method and RT-PCR analysis. Down-regulation of miR-96 expression reduced the proliferation and colony formation ability of PC3 prostate cancer cells, while over-expression of miR-96 induced proliferation and colony formation ability of LNCaP prostate cancer cells. Forkhead box protein O1 (FOXO1) is key tumor suppressors and has been shown to play key roles in the regulation of diverse cellular processes, including cell proliferation, differentiation, cell cycle progression and apoptosis. The expression level of FOXO1 was strikingly up-regulated in PC3 cells after transfected with miR-96 inhibitor, and FOXO1 expression was down-regulated in LNCaP cells after transfected with miR-96 mimics. miR-96 may play a vital role in promoting cell proliferation in human prostate cancer cells. Inhibition of miR-96 caused expression increase of tumor suppressor gene FOXO1, thus manipulating miR-96 expression may be a promising approach in treatment of prostate cancer.
Collapse
Affiliation(s)
- Jun-Jie Yu
- Department of Urology, First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, People's Republic of China
| | | | | | | |
Collapse
|