51
|
Barbazán J, Alonso-Alconada L, Elkhatib N, Geraldo S, Gurchenkov V, Glentis A, van Niel G, Palmulli R, Fernández B, Viaño P, Garcia-Caballero T, López-López R, Abal M, Vignjevic DM. Liver Metastasis Is Facilitated by the Adherence of Circulating Tumor Cells to Vascular Fibronectin Deposits. Cancer Res 2017; 77:3431-3441. [PMID: 28536280 DOI: 10.1158/0008-5472.can-16-1917] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/19/2016] [Accepted: 05/10/2017] [Indexed: 12/23/2022]
Abstract
The interaction between circulating tumor cells (CTC) and endothelial cells during extravasation is a critical process during metastatic colonization, but its mechanisms remain poorly characterized. Here we report that the luminal side of liver blood vessels contains fibronectin deposits that are enriched in mice bearing primary tumors and are also present in vessels from human livers affected with metastases. Cancer cells attached to endothelial fibronectin deposits via talin1, a major component of focal adhesions. Talin1 depletion impaired cancer cell adhesion to the endothelium and transendothelial migration, resulting in reduced liver metastasis formation in vivo Talin1 expression levels in patient CTC's correlated with prognosis and therapy response. Together, our findings uncover a new mechanism for liver metastasis formation involving an active contribution of hepatic vascular fibronectin and talin1 in cancer cells. Cancer Res; 77(13); 3431-41. ©2017 AACR.
Collapse
Affiliation(s)
- Jorge Barbazán
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France. .,Translational Medical Oncology; Health Research Institute of Santiago (IDIS); SERGAS, Santiago de Compostela, Spain
| | - Lorena Alonso-Alconada
- Translational Medical Oncology; Health Research Institute of Santiago (IDIS); SERGAS, Santiago de Compostela, Spain
| | - Nadia Elkhatib
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
| | - Sara Geraldo
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
| | - Vasily Gurchenkov
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
| | | | | | - Roberta Palmulli
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
| | - Beatriz Fernández
- Department of Pathology, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - Patricia Viaño
- Department of Pathology, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain.,IDIS, CHUS, Santiago de Compostela, Spain
| | - Tomas Garcia-Caballero
- Department of Morphological Sciences, School of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Rafael López-López
- Translational Medical Oncology; Health Research Institute of Santiago (IDIS); SERGAS, Santiago de Compostela, Spain
| | - Miguel Abal
- Translational Medical Oncology; Health Research Institute of Santiago (IDIS); SERGAS, Santiago de Compostela, Spain
| | | |
Collapse
|
52
|
Wang D, Chen G, Ren L. Preparation and Characterization of the Sulfobutylether-β-Cyclodextrin Inclusion Complex of Amiodarone Hydrochloride with Enhanced Oral Bioavailability in Fasted State. AAPS PharmSciTech 2017; 18:1526-1535. [PMID: 27757923 DOI: 10.1208/s12249-016-0646-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/03/2016] [Indexed: 11/30/2022] Open
Abstract
Amiodarone hydrochloride (AMD) is used in the treatment of a wide range of cardiac tachyarrhythmias, including both ventricular fibrillation (VF) and hemodynamically unstable ventricular tachycardia (VT). The objectives of this study were to improve the solubility and bioavailability in fasted state and to reduce the food effect of AMD by producing its inclusion complex with sulfobutylether-β-cyclodextrin (SBE-β-CD). The complex was prepared through a saturated water solution combined with the freeze-drying method and then characterized by Fourier transform infrared spectroscopy, proton nuclear magnetic resonance spectroscopy, and differential scanning calorimetry. The solubilities of AMD and its complex were 0.35 and 68.62 mg/mL, respectively, and the value of the inclusion complex was significantly improved by 196-fold compared with the solubility of free AMD. The dissolution of the AMD-SBE-β-CD inclusion complex in four different dissolution media was larger than that of the commercial product. The cumulative dissolution was more than 85% in water, pH 4.5 NaAc-HAC buffer, and pH 1.2 HCl aqueous solution. Moreover, the pharmacokinetic study found that the C max, AUC(0-t), and AUC(0-∞) of the AMI-SBE-β-CD inclusion complex had no significant difference in fasted and fed state, which indicated that the absorption of the AMI-SBE-β-CD inclusion complex in fasted state was increased and not affected by food.
Collapse
|
53
|
Zhang C, Mao HL, Cao Y. Nuclear accumulation of symplekin promotes cellular proliferation and dedifferentiation in an ERK1/2-dependent manner. Sci Rep 2017. [PMID: 28630428 PMCID: PMC5476558 DOI: 10.1038/s41598-017-04005-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Symplekin is a multifunctional protein that localizes to both tight junctions and the nucleus in polarized epithelial cells, with confirmed roles in mRNA maturation, transcriptional modulation and tight-junction assembly. However, the mechanisms governing its subcellular distribution and related functions remain unclear. In this study, we found that symplekin primarily localizes to the nuclei of cultured dedifferentiated colorectal cancer cells, and nuclear symplekin showed higher phosphorylation and binding affinity with YBX3 than its membrane fraction. Moreover, the accumulation of nuclear symplekin promoted cell proliferation and dedifferentiation as well as β-catenin transactivation in vitro. Nuclear symplekin acts as a transcriptional co-activator for the expression of many cell cycle-related genes. Furthermore, extracellular signal-regulated kinase (ERK) phosphorylated symplekin at T1257 to facilitate its nuclear accumulation upon epidermal growth factor (EGF) stimulation. Meanwhile, reduction of total symplekin also induced certain epithelial-mesenchymal transition features in HT-29 cells. Taken together, our results confirm the coordinated roles of symplekin in cell junctions and gene transcription, which are related to its subcellular localization. The significance of nuclear symplekin in tumorigenesis is also highlighted, and ERK-dependent phosphorylation represents a mechanism for its subcellular sorting.
Collapse
Affiliation(s)
- Chen Zhang
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Hai-Lei Mao
- Department of Anesthesiology and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Cao
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
54
|
Lv J, Sun B, Mai Z, Jiang M, Du J. CLDN-1 promoted the epithelial to migration and mesenchymal transition (EMT) in human bronchial epithelial cells via Notch pathway. Mol Cell Biochem 2017; 432:91-98. [PMID: 28316062 DOI: 10.1007/s11010-017-3000-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/04/2017] [Indexed: 01/11/2023]
Abstract
Claudin-1 (CLDN-1) is one of main tight junction components that play an important role in epithelial-mesenchymal transition (EMT). However, the effects of CLDN-1 on the migration and EMT induced by TGF-β1 in primary normal human bronchial epithelial (NHBE) and BEAS-2B cells have not been clear. The expression of CLDN-1 was quantified by Western blotting in NHBE and BEAS-2B cells. Cell migration and invasion were detected using transwell assays. The expression level of E-cadherin, N-cadherin, α-SMA, and Vimentin was evaluated by quantitative real-time PCR and Western blotting. Here we showed that the protein expression of CLDN-1 was increased exposed to TGF-β1 in a dose- and time-dependent manner. Knockdown of CLDN-1 using small interfering CLDN-1 RNA (siCLDN-1) prevented the migration and invasion in NHBE and BEAS-2B cells. Moreover, depletion of CLDN-1 promoted the E-cadherin expression and decreased the mRNA and protein levels of N-cadherin, α-SMA, and Vimentin induced by TGF-β1. Furthermore, CLDN-1 silencing resulted in the reduction of the Notch intracellular domain (NICD) and hairy enhancer of split-1 (Hes-1) in mRNA and protein level. Jagged-1, an activator of Notch signaling pathway, abrogated the protective function of siCLDN-1 in migration and EMT. In conclusion, CLDN-1 promoted the migration and EMT through the Notch signaling pathway.
Collapse
Affiliation(s)
- Jing Lv
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China
| | - Baohua Sun
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China.
| | - Zhitao Mai
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China
| | - Mingming Jiang
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China
| | - Junfeng Du
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, People's Republic of China
| |
Collapse
|
55
|
Computational Analysis of Specific MicroRNA Biomarkers for Noninvasive Early Cancer Detection. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4680650. [PMID: 28357401 PMCID: PMC5357545 DOI: 10.1155/2017/4680650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/13/2017] [Indexed: 01/18/2023]
Abstract
Cancer is a complex disease residing in various tissues of human body, accompanied with many abnormalities and mutations in genomes, transcriptome, and epigenome. Early detection plays a crucial role in extending survival time of all major cancer types. Recent advances in microarray and sequencing techniques have given more support to identifying effective biomarkers for early detection of cancer. MicroRNAs (miRNAs) are more and more frequently used as candidates for biomarkers in cancer related studies due to their regulation of target gene expression. In this paper, the comparative analysis is used to discover miRNA expression patterns in cancer versus normal samples on early stage of eight prevalent cancer types. Our work focuses on the specific miRNAs biomarkers identification and function analysis. Several identified miRNA biomarkers in this paper are matched well with those reported in existing researches, and most of them could serve as potential candidate indicators for clinical early diagnosis applications.
Collapse
|
56
|
Zhang Y, Huang B, Wang HY, Chang A, Zheng XFS. Emerging Role of MicroRNAs in mTOR Signaling. Cell Mol Life Sci 2017; 74:2613-2625. [PMID: 28238105 DOI: 10.1007/s00018-017-2485-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 02/01/2017] [Accepted: 02/06/2017] [Indexed: 12/15/2022]
Abstract
Mechanistic target of rapamycin (mTOR) is a conserved serine/threonine kinase that plays a critical role in the control of cellular growth and metabolism. Hyperactivation of mTOR pathway is common in human cancers, driving uncontrolled proliferation. MicroRNA (miRNA) is a class of short noncoding RNAs that regulate the expression of a wide variety of genes. Deregulation of miRNAs is a hallmark of cancer. Recent studies have revealed interplays between miRNAs and the mTOR pathway during cancer development. Such interactions appear to provide a fine-tuning of various cellular functions and contribute qualitatively to the behavior of cancer. Here we provide an overview of current knowledge regarding the reciprocal relationship between miRNAs and mTOR pathway: regulation of mTOR signaling by miRNAs and control of miRNA biogenesis by mTOR. Further research in this area may prove important for the diagnosis and therapy of human cancer.
Collapse
Affiliation(s)
- Yanjie Zhang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201999, China.
| | - Bo Huang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201999, China
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Rutgers Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA.,Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ, 08903, USA
| | - Augustus Chang
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ, 08903, USA
| | - X F Steven Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China. .,Rutgers Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA. .,Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
57
|
Li X, You M, Liu YJ, Ma L, Jin PP, Zhou R, Zhang ZX, Hua B, Ji XJ, Cheng XY, Yin F, Chen Y, Yin W. Reversal of the Apoptotic Resistance of Non-Small-Cell Lung Carcinoma towards TRAIL by Natural Product Toosendanin. Sci Rep 2017; 7:42748. [PMID: 28209994 PMCID: PMC5314365 DOI: 10.1038/srep42748] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/13/2017] [Indexed: 11/18/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively triggers cancer cell death via its association with death receptors on the cell membrane, but exerts negligible side effects on normal cells. However, some non-small-cell lung carcinoma (NSCLC) patients exhibited resistance to TRAIL treatment in clinical trials, and the mechanism varies. In this study, we described for the first time that toosendanin (TSN), a triterpenoid derivative used in Chinese medicine for pain management, could significantly sensitize human primary NSCLC cells or NSCLC cell lines to TRAIL-mediated apoptosis both in vitro and in vivo, while showing low toxicity against human primary cells or tissues. The underlying apoptotic mechanisms involved upregulation of death receptor 5 (DR5) and CCAAT/enhancer binding protein homologous protein, which is related to the endoplasmic reticulum stress response, and is further associated with reactive oxygen species generation and Ca2+ accumulation. Surprisingly, TSN also induced autophagy in NSCLC cells, which recruited membrane DR5, and subsequently antagonized the apoptosis-sensitizing effect of TSN. Taken together, TSN can be used to sensitize tumors and the combination of TRAIL and TSN may represent a useful strategy for NSCLC therapy; moreover, autophagy serves as an important drug resistance mechanism for TSN.
Collapse
Affiliation(s)
- Xin Li
- The State Key Lab of Pharmaceutical Biotechnology, College of life Sciences, Nanjing University, Nanjing, 210093, China.,Jiangsu Key Lab of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming You
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Yong-Jian Liu
- The State Key Lab of Pharmaceutical Biotechnology, College of life Sciences, Nanjing University, Nanjing, 210093, China
| | - Lin Ma
- The State Key Lab of Pharmaceutical Biotechnology, College of life Sciences, Nanjing University, Nanjing, 210093, China
| | - Pei-Pei Jin
- Department of Anesthesiology and Intensive Care Unit, Changhai Hospital, Affiliated Hospital of the Second Military Medical University, Shanghai, China
| | - Ri Zhou
- The State Key Lab of Pharmaceutical Biotechnology, College of life Sciences, Nanjing University, Nanjing, 210093, China
| | - Zhao-Xin Zhang
- The State Key Lab of Pharmaceutical Biotechnology, College of life Sciences, Nanjing University, Nanjing, 210093, China
| | - Baojin Hua
- Guang'anmen hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao-Jun Ji
- The State Key Lab of Pharmaceutical Biotechnology, College of life Sciences, Nanjing University, Nanjing, 210093, China
| | - Xiao-Ying Cheng
- The State Key Lab of Pharmaceutical Biotechnology, College of life Sciences, Nanjing University, Nanjing, 210093, China
| | - Fangzhou Yin
- College of Pharmacy, Nanjing University of Chinese medicine, China
| | - Yan Chen
- Guang'anmen hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Jiangsu Cancer Hospital &Institute Affiliated to Nanjing Medical University, China
| | - Wu Yin
- The State Key Lab of Pharmaceutical Biotechnology, College of life Sciences, Nanjing University, Nanjing, 210093, China.,Jiangsu Key Lab of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
58
|
Li H, Batth IS, Qu X, Xu L, Song N, Wang R, Liu Y. IGF-IR signaling in epithelial to mesenchymal transition and targeting IGF-IR therapy: overview and new insights. Mol Cancer 2017; 16:6. [PMID: 28137302 PMCID: PMC5282886 DOI: 10.1186/s12943-016-0576-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/19/2016] [Indexed: 01/06/2023] Open
Abstract
The insulin-like growth factor-I (IGF-I) signaling induces epithelial to mesenchymal transition (EMT) program and contributes to metastasis and drug resistance in several subtypes of tumors. In preclinical studies, targeting of the insulin-like growth factor-I receptor (IGF-IR) showed promising anti-tumor effects. Unfortunately, high expectations for anti-IGF-IR therapy encountered challenge and disappointment in numerous clinical trials. This review summarizes the regulation of EMT by IGF-I/IGF-IR signaling pathway and drug resistance mechanisms of targeting IGF-IR therapy. Most importantly, we address several factors in the regulation of IGF-I/IGF-IR-associated EMT progression that may be potential predictive biomarkers in targeted therapy.
Collapse
Affiliation(s)
- Heming Li
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China.,Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Izhar Singh Batth
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiujuan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Ling Xu
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Na Song
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Ruoyu Wang
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China.
| | - Yunpeng Liu
- Department of Medical Oncology, the First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China.
| |
Collapse
|
59
|
Mansoori B, Mohammadi A, Shirjang S, Baghbani E, Baradaran B. Micro RNA 34a and Let-7a Expression in Human Breast Cancers is Associated with Apoptotic Expression Genes. Asian Pac J Cancer Prev 2017; 17:1887-90. [PMID: 27221871 DOI: 10.7314/apjcp.2016.17.4.1887] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Breast cancer is the most common cause of cancer-related death among women in the whole world. MiR- 34a and let-7a are well known tumor suppressors that participate in the regulation of apoptosis, invasion and other cellular functions. In this study, expression of miR-34a, let-7a and apoptosis pathway genes such as Bcl-2, Caspase-3 and P53 were evaluated using quantitative real-time PCR in 45 paired samples of normal margin and tumor tissue collected from breast cancer patient at advanced stage (3-4). MiR-34a, let-7a, caspase-3 and P53 expression are reduced and Bcl-2 expression is increased within tumoral tissues in comparison with normal margin tissues. P53 expression directly or indirectly was correlated with miR-34a, let-7a, Bcl-2 and caspase-3 expression. In This study we found that MiR-34a and let-7a expression are reduced in the tumoral tissues. Down- regulation of these two molecules correlated with expression of genes associated with apoptosis. These results suggest that due to the correlation of miR-34a and let-7a with apoptotic and anti-apoptotic pathways these molecules could participate as regulators in advanced clinical stages of breast cancer and should be considered as markers for diagnosis, prognostic assessment and targeted therapy.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran E-mail :
| | | | | | | | | |
Collapse
|
60
|
Matrine induces RIP3-dependent necroptosis in cholangiocarcinoma cells. Cell Death Discov 2017; 3:16096. [PMID: 28179994 PMCID: PMC5253620 DOI: 10.1038/cddiscovery.2016.96] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/04/2016] [Accepted: 11/08/2016] [Indexed: 12/22/2022] Open
Abstract
The development of acquired resistance to pro-apoptotic antitumor agents is a major impediment to the cure of cholangiocarcinoma (CCA). Antitumor drugs inducing non-apoptotic cell death are considered as a new approach to overcome such drug resistance. Here, we reported for the first time that matrine-induced necroptosis in CCA cell lines, differing from its classical role to induce apoptosis in many other kinds of cancer cells. CCA cells under matrine treatment exhibited typical necrosis-like but not apoptotic morphologic change. These matrine-induced morphologic change and cell death in CCA cells were greatly attenuated by necroptosis inhibitor necrostatin-1, but not apoptosis inhibitor z-VAD-fmk. Unlike many cancer cells with negative receptor-interacting protein 3 (RIP3) expression, moderate expression of RIP3 in CCA cells was observed and was required for matrine to induce necroptosis, which was switched to apoptosis after knocking down endogenous RIP3. Moreover, matrine could increase RIP3 expression level, which may facilitate the necroptosis process. Translocation of mixed lineage kinase-domain like (MLKL) from cytoplasm to plasma membrane as a downstream event of RIP3, as well as the increased production of reactive oxygen species (ROS) by RIP3/MLKL, was critical for matrine to induce necroptosis. In clinical study, we found RIP3 was lower but still moderately expressed in most CCA tissue samples compared with adjacent normal tissues. Taken together, we identified matrine as a necroptosis inducer in CCA by enhancing RIP3 expression and the following RIP3/MLKL/ROS signaling pathway, which provided new individualized strategies based on RIP3 expression to overcome chemoresistance in CCA therapy.
Collapse
|
61
|
Abstract
The pseudokinase complement of the human kinase superfamily consists of approximately 60 signaling proteins, which lacks one or more of the amino acids typically required to correctly align ATP and metal ions, and phosphorylate protein substrates. Recent studies in the pseudokinase field have begun to expose the biological relevance of pseudokinases, which are now thought to perform a diverse range of physiological roles and are connected to a multitude of human diseases, including cancer. In this review, we discuss how and why members of the 'pseudokinome' represent important new targets for drug discovery, and describe how knowledge of protein structure and function provides informative clues to help guide the rational chemical design or repurposing of inhibitors to target pseudokinases.
Collapse
|
62
|
Li J, Chen Y, Jin M, Wang J, Li S, Chen Z, Yu W. MicroRNA-134 reverses multidrug resistance in human lung adenocarcinoma cells by targeting FOXM1. Oncol Lett 2017; 13:1451-1455. [PMID: 28454276 DOI: 10.3892/ol.2017.5574] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/03/2016] [Indexed: 01/01/2023] Open
Abstract
Multidrug resistance (MDR) is the primary barrier to the success of chemotherapy for lung adenocarcinoma. MicroRNA (miR)-134, which is downregulated in lung adenocarcinoma, influences cell proliferation, apoptosis and invasion of lung adenocarcinoma. However, the function of miR-134 in the MDR of lung adenocarcinoma remains unclear. In the present study, it was identified that miR-134 expression is significantly downregulated in A549/cisplatin MDR lung adenocarcinoma cells, as compared with A549 parental cells. miR-134 regulates the sensitivity of lung adenocarcinoma cells to certain anticancer drugs. Furthermore, it was demonstrated that forkhead box M1 and multidrug resistance-associated protein 1 are functional targets of miR-134. These data revealed an important role for miR-134 in the regulation of MDR in lung adenocarcinoma.
Collapse
Affiliation(s)
- Jipeng Li
- Department of Central Laboratory, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Ying Chen
- Department of Central Laboratory, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Mingwei Jin
- Department of Central Laboratory, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Jianhua Wang
- Department of Central Laboratory, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Shanfeng Li
- Department of Central Laboratory, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Zhe Chen
- Department of Central Laboratory, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Wanjun Yu
- Department of Respiratory and Critical Care Medicine, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
63
|
Filella X, Foj L. miRNAs as novel biomarkers in the management of prostate cancer. ACTA ACUST UNITED AC 2017; 55:715-736. [DOI: 10.1515/cclm-2015-1073] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/29/2015] [Indexed: 12/15/2022]
Abstract
AbstractmicroRNAs (miRNAs) are small non-coding RNAs that control gene expression posttranscriptionally and are part of the giant non codifying genoma. Cumulating data suggest that miRNAs are promising potential biomarkers for many diseases, including cancer. Prostate cancer (PCa) detection is currently based in the serum prostate-specific antigen biomarker and digital rectal examination. However, these methods are limited by a low predictive value and the adverse consequences associated with overdiagnosis and overtreatment. New biomarkers that could be used for PCa detection and prognosis are still needed. Recent studies have demonstrated that aberrant expressions of microRNAs are associated with the underlying mechanisms of PCa. This review attempts to extensively summarize the current knowledge of miRNA expression patterns, as well as their targets and involvement in PCa pathogenesis. We focused our review in the value of circulating and urine miRNAs as biomarkers in PCa patients, highlighting the existing discrepancies between different studies, probably associated with the important methodological issues related to their quantitation and normalization. The majority of studies have been performed in serum or plasma, but urine obtained after prostate massage appears as a new way to explore the usefulness of miRNAs. Large screening studies to select a miRNA profile have been completed, but bioinformatics tools appear as a new approach to select miRNAs that are relevant in PCa development. Promising preliminary results were published concerning miR-141, miR-375 and miR-21, but larger and prospective studies using standardized methodology are necessary to define the value of miRNAs in the detection and prognosis of PCa.
Collapse
|
64
|
Proteomic profile of the Bradysia odoriphaga in response to the microbial secondary metabolite benzothiazole. Sci Rep 2016; 6:37730. [PMID: 27883048 PMCID: PMC5121901 DOI: 10.1038/srep37730] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/31/2016] [Indexed: 01/15/2023] Open
Abstract
Benzothiazole, a microbial secondary metabolite, has been demonstrated to possess fumigant activity against Sclerotinia sclerotiorum, Ditylenchus destructor and Bradysia odoriphaga. However, to facilitate the development of novel microbial pesticides, the mode of action of benzothiazole needs to be elucidated. Here, we employed iTRAQ-based quantitative proteomics analysis to investigate the effects of benzothiazole on the proteomic expression of B. odoriphaga. In response to benzothiazole, 92 of 863 identified proteins in B. odoriphaga exhibited altered levels of expression, among which 14 proteins were related to the action mechanism of benzothiazole, 11 proteins were involved in stress responses, and 67 proteins were associated with the adaptation of B. odoriphaga to benzothiazole. Further bioinformatics analysis indicated that the reduction in energy metabolism, inhibition of the detoxification process and interference with DNA and RNA synthesis were potentially associated with the mode of action of benzothiazole. The myosin heavy chain, succinyl-CoA synthetase and Ca+-transporting ATPase proteins may be related to the stress response. Increased expression of proteins involved in carbohydrate metabolism, energy production and conversion pathways was responsible for the adaptive response of B. odoriphaga. The results of this study provide novel insight into the molecular mechanisms of benzothiazole at a large-scale translation level and will facilitate the elucidation of the mechanism of action of benzothiazole.
Collapse
|
65
|
Durand N, Storz P. Targeting reactive oxygen species in development and progression of pancreatic cancer. Expert Rev Anticancer Ther 2016; 17:19-31. [PMID: 27841037 DOI: 10.1080/14737140.2017.1261017] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDA) is characterized by expression of oncogenic KRas which drives all aspects of tumorigenesis. Oncogenic KRas induces the formation of reactive oxygen species (ROS) which have been implicated in initiation and progression of PDA. To facilitate tumor promoting levels and to avoid oncogene-induced senescence or cytotoxicity, ROS homeostasis in PDA cells is balanced by additional up-regulation of antioxidant systems. Areas covered: We examine the sources of ROS in PDA, the mechanisms by which ROS homeostasis is maintained, and the biological consequences of ROS in PDA. Additionally, we discuss the potential mechanisms for targeting ROS homoeostasis as a point of therapeutic intervention. An extensive review of the relevant literature as it relates to the topic was conducted using PubMed. Expert commentary: Even though oncogenic mutations in the KRAS gene have been detected in over 95% of human pancreatic adenocarcinoma, targeting its gene product, KRas, has been difficult. The dependency of PDA cells on balancing ROS homeostasis could be an angle for new prevention or treatment strategies. These include use of antioxidants to prevent formation or progression of precancerous lesions, or methods to increase ROS in tumor cells to toxic levels.
Collapse
Affiliation(s)
- Nisha Durand
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| | - Peter Storz
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| |
Collapse
|
66
|
Maina PK, Shao P, Liu Q, Fazli L, Tyler S, Nasir M, Dong X, Qi HH. c-MYC drives histone demethylase PHF8 during neuroendocrine differentiation and in castration-resistant prostate cancer. Oncotarget 2016; 7:75585-75602. [PMID: 27689328 PMCID: PMC5342763 DOI: 10.18632/oncotarget.12310] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 09/20/2016] [Indexed: 02/06/2023] Open
Abstract
Epigenetic factors play critical roles in prostate cancer (PCa) development. However, how they contribute to neuroendocrine differentiation (NED) and castration-resistant PCa (CRPC) is not fully understood. Using bioinformatics and biochemical approaches to analyze cell-based models of NED and CRPC, we found a cluster of epigenetic factors whose expression is downregulated during NED and upregulated in CRPC (i.e. follow a Down-Up pattern). Two histone demethylases within this cluster, PHF8 and KDM3A, are post-transcriptionally regulated by c-MYC through miR-22, which targets both PHF8 and KDM3A. We also found that the c-MYC/miR-22/PHF8 axis is downstream of androgen receptor (AR) signaling in CRPC cells. The co-expression of PHF8 with AR in clinical CRPC samples, normal mouse prostate, and adenocarcinomas of the prostate during PCa progression in a transgenic (TRAMP) mouse model supports the connection between PHF8 and AR. Knockdown of PHF8 impedes cell cycle progression in CRPC cells and has more profound effects on their growth than on the parental LNCaP cell line. Furthermore, PHF8 knockdown sensitizes LNCaP-Abl cells to the AR antagonist enzalutamide. Our data reveal novel mechanisms that underlie the regulation of PHF8 and KDM3A during NED and in CRPC, and support the candidacy of PHF8 as a therapeutic target in CRPC.
Collapse
MESH Headings
- 3' Untranslated Regions
- Animals
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Cycle Checkpoints/genetics
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cell Survival/genetics
- Epigenesis, Genetic
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, myc
- Histone Demethylases/genetics
- Humans
- Interleukin-6/metabolism
- Interleukin-6/pharmacology
- Jumonji Domain-Containing Histone Demethylases/genetics
- Jumonji Domain-Containing Histone Demethylases/metabolism
- Male
- Mice
- MicroRNAs/genetics
- Models, Biological
- Neoplasm Grading
- Neuroendocrine Tumors/genetics
- Neuroendocrine Tumors/metabolism
- Neuroendocrine Tumors/pathology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- RNA Interference
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Transcription Factors/genetics
- Transcriptome
Collapse
Affiliation(s)
- Peterson Kariuki Maina
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Peng Shao
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Qi Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Ladan Fazli
- Vancouver Prostate Center, Department of Urology Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Scott Tyler
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Moman Nasir
- Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Xuesen Dong
- Vancouver Prostate Center, Department of Urology Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Hank Heng Qi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| |
Collapse
|
67
|
Sun XF, Sun JP, Hou HT, Li K, Liu X, Ge QX. MicroRNA-27b exerts an oncogenic function by targeting Fbxw7 in human hepatocellular carcinoma. Tumour Biol 2016; 37:15325-15332. [PMID: 27704356 DOI: 10.1007/s13277-016-5444-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/23/2016] [Indexed: 10/20/2022] Open
Abstract
Aberrant expression of microRNAs (miRNAs) plays fundamental effect on the pathogenesis of hepatocellular carcinoma (HCC). MiR-27b was previously found to play important roles in human cancers. However, its expression status, clinical significance, and biological functions in HCC remain largely unclear. The expression status of miR-27b in HCC specimens and cells were determined with qRT-PCR. MTT, 5-bromodeoxyuridine (BrdU) proliferation assays, and flow cytometry analysis were carried out to assay proliferation, cell cycle, and apoptosis. A subcutaneous model was used to evaluated the HCC tumor growth in vivo. The putative target gene of miR-27b was disclosed by TargetScan and a luciferase reporter assay. The levels of miR-27b were overexpressed in HCC. Overexpression of miR-27b was correlated with adverse prognostic features and reduced survival rate. Inhibition of miR-27b in SMMC-7721 cells remarkably suppressed proliferative ability and cell-cycle progression while enhanced apoptosis. In contrast, miR-27b overexpression resulted in prominent increased proliferation and process of cell cycle and reduced apoptosis of Hep3B cells. In vivo studies showed that knockdown of miR-27b inhibited the in vivo growth of SMMC-7721 cells in mouse xenograft model. Furthermore, we confirmed that Fbxw7 was directly regulated by miR-27b and mediated the roles of miR-27b in HCC. We suggest that miR-27b serves as an oncogenic miRNA in HCC by modulating proliferation, cell-cycle progression, and apoptosis, and its oncogenic effect is mediated by its downstream target gene, Fbxw7.
Collapse
Affiliation(s)
- Xin-Fang Sun
- Department of Gastroenterology, Huaihe Hospital, Henan University, No. 8 Baobei Road, Kaifeng, Henan Province, 475000, China
| | - Jin-Ping Sun
- Department of Gastroenterology, Huaihe Hospital, Henan University, No. 8 Baobei Road, Kaifeng, Henan Province, 475000, China
| | - Hong-Tao Hou
- Department of Gastroenterology, Huaihe Hospital, Henan University, No. 8 Baobei Road, Kaifeng, Henan Province, 475000, China
| | - Ke Li
- Department of Gastroenterology, Huaihe Hospital, Henan University, No. 8 Baobei Road, Kaifeng, Henan Province, 475000, China
| | - Xin Liu
- Department of Gastroenterology, Huaihe Hospital, Henan University, No. 8 Baobei Road, Kaifeng, Henan Province, 475000, China
| | - Quan-Xing Ge
- Department of Gastroenterology, Huaihe Hospital, Henan University, No. 8 Baobei Road, Kaifeng, Henan Province, 475000, China.
| |
Collapse
|
68
|
Ranji P, Salmani Kesejini T, Saeedikhoo S, Alizadeh AM. Targeting cancer stem cell-specific markers and/or associated signaling pathways for overcoming cancer drug resistance. Tumour Biol 2016; 37:13059-13075. [DOI: 10.1007/s13277-016-5294-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
|
69
|
Nigita G, Acunzo M, Romano G, Veneziano D, Laganà A, Vitiello M, Wernicke D, Ferro A, Croce CM. microRNA editing in seed region aligns with cellular changes in hypoxic conditions. Nucleic Acids Res 2016; 44:6298-308. [PMID: 27298257 PMCID: PMC4994866 DOI: 10.1093/nar/gkw532] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/22/2022] Open
Abstract
RNA editing is a finely tuned, dynamic mechanism for post-transcriptional gene regulation that has been thoroughly investigated in the last decade. Nevertheless, RNA editing in non-coding RNA, such as microRNA (miRNA), have caused great debate and have called for deeper investigation. Until recently, in fact, inadequate methodologies and experimental contexts have been unable to provide detailed insights for further elucidation of RNA editing affecting miRNAs, especially in cancer.In this work, we leverage on recent innovative bioinformatics approaches applied to a more informative experimental context in order to analyze the variations in miRNA seed region editing activity during a time course of a hypoxia-exposed breast cancer cell line. By investigating its behavior in a dynamic context, we found that miRNA editing events in the seed region are not depended on miRNA expression, unprecedentedly providing insights on the targetome shifts derived from these modifications. This reveals that miRNA editing acts under the influence of environmentally induced stimuli.Our results show a miRNA editing activity trend aligning with cellular pathways closely associated to hypoxia, such as the VEGF and PI3K/Akt pathways, providing important novel insights on this poorly elucidated phenomenon.
Collapse
Affiliation(s)
- Giovanni Nigita
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Mario Acunzo
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Giulia Romano
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Dario Veneziano
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Alessandro Laganà
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Marika Vitiello
- Department of Clinicaland Molecular Biomedicine, University of Catania, Catania 95125, Italy
| | - Dorothee Wernicke
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Alfredo Ferro
- Department of Structural & Functional Biology, University 'Federico II' of Naples, Naples 80126, Italy
| | - Carlo M Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
70
|
Kojima S, Goto Y, Naya Y. The roles of microRNAs in the progression of castration-resistant prostate cancer. J Hum Genet 2016; 62:25-31. [PMID: 27278789 DOI: 10.1038/jhg.2016.69] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 04/28/2016] [Accepted: 05/10/2016] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) is one of the leading causes of cancer-related death in men. PCa is androgen-dependent, and androgen-deprivation therapy is effective for first-line hormonal treatment, but the androgen-independent phenotype of PCa eventually develops, which is difficult to treat and has no effective cure. Recently, microRNAs have been discovered to have important roles in the initiation and progression of PCa, suggesting their use in diagnosis, predicting prognosis and development of treatment for castration-resistant PCa (CRPC). Understanding the networks of microRNAs and their target genes is necessary to ascertain their roles and importance in the development and progression of PCa. This review summarizes the current knowledge about microRNAs regulating PCa progression and elucidates the mechanism of progression to CRPC.
Collapse
Affiliation(s)
- Satoko Kojima
- Department of Urology, Teikyo University Chiba Medical Center, Ichihara, Japan
| | - Yusuke Goto
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yukio Naya
- Department of Urology, Teikyo University Chiba Medical Center, Ichihara, Japan
| |
Collapse
|
71
|
Keller SM, Roth TL. Environmental influences on the female epigenome and behavior. ENVIRONMENTAL EPIGENETICS 2016; 2:dvw007. [PMID: 27746953 PMCID: PMC5065103 DOI: 10.1093/eep/dvw007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 06/06/2023]
Abstract
Environmental factors have long-lasting effects on brain development and behavior. One way experiences are propagated is via epigenetic modifications to the genome. Environmentally-driven epigenetic modifications show incredible brain region- and sex-specificity, and many brain regions affected are ones involved in maternal behavior. In rodent models, females are typically the primary caregiver and thus, any environmental factors that modulate the epigenotype of the mother could have consequences for her current and future offspring. Here we review evidence of the susceptibility of the female epigenome to environmental factors, with a focus on brain regions involved in maternal behavior. Accordingly, implications for interventions that target the mother's epigenome and parenting behavior are discussed.
Collapse
Affiliation(s)
- Samantha M. Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Tania L. Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
72
|
Yang P, Wei J, Li W, He F, Zeng S, Zhang T, Sun Z, Cao J. High expression of growth factor receptor-bound protein 14 predicts poor prognosis for colorectal cancer patients. Biotechnol Lett 2016; 38:1043-7. [PMID: 26965150 DOI: 10.1007/s10529-016-2077-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/01/2016] [Indexed: 12/18/2022]
Abstract
OBJECTS To explore the roles of growth factor receptor-bound protein 14 (GRB14) in colorectal cancer (CRC) and its correlation with clinicopathological characteristics and prognosis of CRC patients. RESULTS GRB14 was localized in the cytoplasm of CRC and benign glandular epithelium cells, showing higher levels in CRC tissues compared with normal colon samples (P < 0.001). High GRB14 was associated with a high pathological grade (P = 0.045), advanced clinical stage (P = 0.018), enhanced tumor invasion (P < 0.001) and lymph node metastasis (P = 0.028). The cancer genome atlas (TCGA) mRNA sequence data showed that GRB14 was upregulated in CRC at an advanced clinical stage (P = 0.011) with enhanced tumor invasion (P < 0.001) and lymph node metastasis (P = 0.014). Kaplan-Meier survival curves revealed that CRC patients with high GRB14 levels had a shorter survival compared with those showing low GRB14 expression (P = 0.007). High GRB14 expression was an independent prognostic factor for CRC patients (HR 2.847, 95 %CI 1.058-7.659; P = 0.038). CONCLUSIONS GRB14 may be an important cancer promoter that enhances CRC progression. Upregulated GRB14 levels may predict a poor clinical outcome in CRC patients.
Collapse
Affiliation(s)
- Ping Yang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Jianchang Wei
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Wanglin Li
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Feng He
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Shanqi Zeng
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Tong Zhang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Zheng Sun
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou Medical University, Panfu Road No. 1, Guangzhou, 510180, China.
| |
Collapse
|
73
|
Mikhaylenko DS, Efremov GD, Sivkov AV, Zaletaev DV. Hormone resistance and neuroendocrine differentiation due to accumulation of genetic lesions during clonal evolution of prostate cancer. Mol Biol 2016. [DOI: 10.1134/s0026893315060187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
74
|
Aakula A, Kohonen P, Leivonen SK, Mäkelä R, Hintsanen P, Mpindi JP, Martens-Uzunova E, Aittokallio T, Jenster G, Perälä M, Kallioniemi O, Östling P. Systematic Identification of MicroRNAs That Impact on Proliferation of Prostate Cancer Cells and Display Changed Expression in Tumor Tissue. Eur Urol 2015; 69:1120-8. [PMID: 26489476 DOI: 10.1016/j.eururo.2015.09.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/14/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Systematic approaches to functionally identify key players in microRNA (miRNA)-target networks regulating prostate cancer (PCa) proliferation are still missing. OBJECTIVE To comprehensively map miRNA regulation of genes relevant for PCa proliferation through phenotypic screening and tumor expression data. DESIGN, SETTING, AND PARTICIPANTS Gain-of-function screening with 1129 miRNA molecules was performed in five PCa cell lines, measuring proliferation, viability, and apoptosis. These results were integrated with changes in miRNA expression from two cohorts of human PCa (188 tumors in total). For resulting miRNAs, the predicted targets were collected and analyzed for patterns with gene set enrichment analysis, and for their association with biochemical recurrence free survival. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Rank product statistical analysis was used to evaluate miRNA effects in phenotypic screening and for expression differences in the prostate tumor cohorts. Expression data were analyzed using the significance analysis of microarrays (SAM) method and the patient material was subjected to Kaplan-Meier statistics. RESULTS AND LIMITATIONS Functional screening identified 25 miRNAs increasing and 48 miRNAs decreasing cell viability. Data integration resulted in 14 miRNAs, with aberrant expression and effect on proliferation. These miRNAs are predicted to regulate >3700 genes, of which 28 were found up-regulated and 127 down-regulated in PCa compared with benign tissue. Seven genes, FLNC, MSRB3, PARVA, PCDH7, PRNP, RAB34, and SORBS1, showed an inverse association to their predicted miRNA, and were identified to significantly correlate with biochemical recurrence free survival in PCa patients. CONCLUSIONS A systematic in vitro screening approach combined with in vivo expression and gene set enrichment analysis provide unbiased means for revealing novel miRNA-target links, possibly driving the oncogenic processes in PCa. PATIENT SUMMARY This study identified novel regulatory molecules, which impact on PCa proliferation and are aberrantly expressed in clinical tumors. Thus, our study reveals regulatory nodes with potential for therapy.
Collapse
Affiliation(s)
- Anna Aakula
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland; VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland(1); Turku Centre for Biotechnology, University of Turku, Turku, Finland(1).
| | - Pekka Kohonen
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland(1); Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Suvi-Katri Leivonen
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland(1); Genome-Scale Biology Research Program, University of Helsinki, Helsinki, Finland
| | - Rami Mäkelä
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland(1); Misvik Biology Corporation, Turku, Finland
| | - Petteri Hintsanen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - John Patrick Mpindi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland; VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland(1)
| | | | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Guido Jenster
- Department of Urology, Erasmus MC, Rotterdam, The Netherlands
| | - Merja Perälä
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland(1); Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland; VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland(1)
| | - Päivi Östling
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland; VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland(1)
| |
Collapse
|
75
|
Pasqualini L, Bu H, Puhr M, Narisu N, Rainer J, Schlick B, Schäfer G, Angelova M, Trajanoski Z, Börno ST, Schweiger MR, Fuchsberger C, Klocker H. miR-22 and miR-29a Are Members of the Androgen Receptor Cistrome Modulating LAMC1 and Mcl-1 in Prostate Cancer. Mol Endocrinol 2015; 29:1037-54. [PMID: 26052614 DOI: 10.1210/me.2014-1358] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The normal prostate as well as early stages and advanced prostate cancer (PCa) require a functional androgen receptor (AR) for growth and survival. The recent discovery of microRNAs (miRNAs) as novel effector molecules of AR disclosed the existence of an intricate network between AR, miRNAs and downstream target genes. In this study DUCaP cells, characterized by high content of wild-type AR and robust AR transcriptional activity, were chosen as the main experimental model. By integrative analysis of chromatin immunoprecipitation-sequencing (ChIP-seq) and microarray expression profiling data, miRNAs putatively bound and significantly regulated by AR were identified. A direct AR regulation of miR-22, miR-29a, and miR-17-92 cluster along with their host genes was confirmed. Interestingly, endogenous levels of miR-22 and miR-29a were found to be reduced in PCa cells expressing AR. In primary tumor samples, miR-22 and miR-29a were less abundant in the cancerous tissue compared with the benign counterpart. This specific expression pattern was associated with a differential DNA methylation of the genomic AR binding sites. The identification of laminin gamma 1 (LAMC1) and myeloid cell leukemia 1 (MCL1) as direct targets of miR-22 and miR-29a, respectively, suggested a tumor-suppressive role of these miRNAs. Indeed, transfection of miRNA mimics in PCa cells induced apoptosis and diminished cell migration and viability. Collectively, these data provide additional information regarding the complex regulatory machinery that guides miRNAs activity in PCa, highlighting an important contribution of miRNAs in the AR signaling.
Collapse
Affiliation(s)
- Lorenza Pasqualini
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Huajie Bu
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Martin Puhr
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Narisu Narisu
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Johannes Rainer
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Bettina Schlick
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Georg Schäfer
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Mihaela Angelova
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Zlatko Trajanoski
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Stefan T Börno
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Michal R Schweiger
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Christian Fuchsberger
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| | - Helmut Klocker
- Department of Urology (L.P., H.B., M.P., B.S., G.S., H.K.), Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Research Institute for Biomedical Aging Research (H.B.), University of Innsbruck, 6020 Innsbruck, Austria; Medical Genomics and Metabolic Genetics Branch (N.N.), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892; Biocenter Innsbruck (J.R.), Section for Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; Center for Biomedicine (J.R., C.F.), EURAC Bolzano, 39100 Bolzano, Italy; Oncotyrol (B.S.), Center for Personalized Cancer Medicine, 6020 Innsbruck, Austria; Department of Pathology (G.S.), Medical University of Innsbruck, 6020 Innsbruck, Austria; Biocenter Innsbruck (M.A., Z.T.), Division of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Max Planck Institute for Molecular Genetics (S.T.B., M.R.S.), 14195 Berlin, Germany; Cologne Center for Genomics (M.R.S.), University of Cologne, 50931 Cologne, Germany; and Department of Biostatistic (C.F.), University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
76
|
Wang L, Wang X, Jin X, Jia R, Huang Q, Tan Y, Guo A. Comparative proteomics of Bt-transgenic and non-transgenic cotton leaves. Proteome Sci 2015; 13:15. [PMID: 25949214 PMCID: PMC4422549 DOI: 10.1186/s12953-015-0071-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/03/2015] [Indexed: 01/05/2023] Open
Abstract
Background As the rapid growth of the commercialized acreage in genetically modified (GM) crops, the unintended effects of GM crops’ biosafety assessment have been given much attention. To investigate whether transgenic events cause unintended effects, comparative proteomics of cotton leaves between the commercial transgenic Bt + CpTI cotton SGK321 (BT) clone and its non-transgenic parental counterpart SY321 wild type (WT) was performed. Results Using enzyme linked immunosorbent assay (ELISA), Cry1Ac toxin protein was detected in the BT leaves, while its content was only 0.31 pg/g. By 2-DE, 58 differentially expressed proteins (DEPs) were detected. Among them 35 were identified by MS. These identified DEPs were mainly involved in carbohydrate transport and metabolism, chaperones related to post-translational modification and energy production. Pathway analysis revealed that most of the DEPs were implicated in carbon fixation and photosynthesis, glyoxylate and dicarboxylate metabolism, and oxidative pentose phosphate pathway. Thirteen identified proteins were involved in protein-protein interaction. The protein interactions were mainly involved in photosynthesis and energy metabolite pathway. Conclusions Our study demonstrated that exogenous DNA in a host cotton genome can affect the plant growth and photosynthesis. Although some unintended variations of proteins were found between BT and WT cotton, no toxic proteins or allergens were detected. This study verified genetically modified operation did not sharply alter cotton leaf proteome, and the target proteins were hardly checked by traditional proteomic analysis. Electronic supplementary material The online version of this article (doi:10.1186/s12953-015-0071-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Limin Wang
- Chinese Academy of Tropical Agricultural Sciences, The Institute of Tropical Biosciences and Biotechnology, Haikou, Hainan 571101 China ; Chinese Academy of Agricultural Sciences, The Oilcrops Research Institute, Wuhan, 430062 China
| | - Xuchu Wang
- Chinese Academy of Tropical Agricultural Sciences, The Institute of Tropical Biosciences and Biotechnology, Haikou, Hainan 571101 China
| | - Xiang Jin
- Chinese Academy of Tropical Agricultural Sciences, The Institute of Tropical Biosciences and Biotechnology, Haikou, Hainan 571101 China
| | - Ruizong Jia
- Chinese Academy of Tropical Agricultural Sciences, The Institute of Tropical Biosciences and Biotechnology, Haikou, Hainan 571101 China
| | - Qixing Huang
- Chinese Academy of Tropical Agricultural Sciences, The Institute of Tropical Biosciences and Biotechnology, Haikou, Hainan 571101 China
| | - Yanhua Tan
- Chinese Academy of Tropical Agricultural Sciences, The Institute of Tropical Biosciences and Biotechnology, Haikou, Hainan 571101 China
| | - Anping Guo
- Chinese Academy of Tropical Agricultural Sciences, The Institute of Tropical Biosciences and Biotechnology, Haikou, Hainan 571101 China
| |
Collapse
|
77
|
Tang RJ, Shen SN, Zhao XY, Nie YZ, Xu YJ, Ren J, Lv MM, Hou YY, Wang TT. Mesenchymal stem cells-regulated Treg cells suppress colitis-associated colorectal cancer. Stem Cell Res Ther 2015; 6:71. [PMID: 25889203 PMCID: PMC4414289 DOI: 10.1186/s13287-015-0055-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 08/30/2014] [Accepted: 03/18/2015] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Previous studies have produced controversial results regarding whether mesenchymal stem cells (MSCs) promote or inhibit tumor development. Given the dual role of MSCs in inflammation and cancer, in this study the colitis-associated colorectal cancer (CAC) model was used to examine whether umbilical cord tissue-derived MSCs could prevent neoplasm by inhibiting chronic inflammation. METHODS MSCs were obtained and identified using flow cytometry. Colitis-associated colorectal cancer model was induced using azoxymethane (AOM) and dextran sulfate sodium (DSS) and MSCs were injected intravenously twice. Levels of immune cells in mesenteric lymph node including regulatory T (Treg) cells were detected using flow cytometry. Naïve T cells and Jurkat cells were co-cultured with MSCs and the effect of MSCs on Treg cells differentiation was evaluated. RESULTS After injection through tail vein, MSCs could migrate to colon and suppress colitis-related neoplasm. This tumor suppressive effect was characterized by longer colon length, decreased tumor numbers and decreased expression of Ki-67. Moreover, MSCs alleviated the pathology of inflammation in the colitis stage of CAC model and inhibited inflammation cytokines both in colon and serum. Furthermore, Treg cells were accumulated in mesenteric lymph node of MSCs-treated mice while the percentage of T helper cells 2 (Th2) and Th17 were not changed. Of note, MSCs secreted transforming growth factor-β (TGF-β) enhanced the induction of Treg cells from naïve T cells. The conditioned medium of MSCs also activated Smad2 signaling, which has been reported to regulate Treg cells. CONCLUSIONS These results proved that MSCs could migrate to colon tissues and induce the differentiation of Treg cells via Smad2 as so to inhibit the colitis and suppress the development of CAC.
Collapse
Affiliation(s)
- Rui-jing Tang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Su-nan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| | - Xiao-yin Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yun-zhong Nie
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yu-jun Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Jing Ren
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Ming-ming Lv
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Ya-yi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| | - Ting-ting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| |
Collapse
|
78
|
Segal CV, Koufaris C, Powell C, Gooderham NJ. Effects of treatment with androgen receptor ligands on microRNA expression of prostate cancer cells. Toxicology 2015; 333:45-52. [PMID: 25846647 DOI: 10.1016/j.tox.2015.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/01/2015] [Accepted: 04/02/2015] [Indexed: 11/26/2022]
Abstract
Post-transcriptional regulation by microRNA (miRNA) is an important aspect of androgen receptor (AR) signalling in prostate cancer cells. However, the global profiling of miRNA expression in prostate cancer cells following treatment with AR ligands has not been reported so far. In this study we examined the effect of treatment with two AR agonists (mibolerone (MIB) and dihydrotestosterone (DHT)) and an AR antagonist (bicalutamide (BIC)) on miRNA expression in the human androgen-dependent LNCaP prostate cancer cell line using microarray technology and verification of selected miRNA using quantitative real-time PCR (qRT-PCR). No miRNA was identified as differentially expressed following treatment with the AR antagonist BIC. In contrast, a number of common and compound-specific alterations in miRNA expression were observed following treatment with AR agonists. Unexpectedly it was found that treatment with the AR agonists resulted in the repression of miR-221, a miRNA previously established to be involved with prostate cancer development. This observation indicates that this miRNA may have a more complex role in prostate cancer development than considered previously. Treatment with MIB led to an induction of miR-210 expression, a hypoxia-related miRNA. This miRNA is reported to be involved in cell adaptation to hypoxia and thus induction in conditions of normoxia may be important in driving metabolic changes observed in prostate cancer. Thus examining the effect of AR agonists and antagonists on miRNA expression can provide novel insights into the response of cells to AR ligands and subsequent downstream events.
Collapse
Affiliation(s)
- Corrinne V Segal
- Department of Surgery & Cancer, Biomolecular Medicine, Imperial College London, London SW72AZ, UK
| | - Costas Koufaris
- Department of Cytogenetics and Genomics, Cyprus Institute of Neurology and Genetics, Cyprus
| | | | - Nigel J Gooderham
- Department of Surgery & Cancer, Biomolecular Medicine, Imperial College London, London SW72AZ, UK.
| |
Collapse
|
79
|
Aakula A, Leivonen SK, Hintsanen P, Aittokallio T, Ceder Y, Børresen-Dale AL, Perälä M, Östling P, Kallioniemi O. MicroRNA-135b regulates ERα, AR and HIF1AN and affects breast and prostate cancer cell growth. Mol Oncol 2015; 9:1287-300. [PMID: 25907805 DOI: 10.1016/j.molonc.2015.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/05/2015] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) regulate a wide range of cellular signaling pathways and biological processes in both physiological and pathological states such as cancer. We have previously identified miR-135b as a direct regulator of androgen receptor (AR) protein level in prostate cancer (PCa). We wanted to further explore the relationship of miR-135b to hormonal receptors, particularly estrogen receptor α (ERα). Here we show that miR-135b expression is lower in ERα-positive breast tumors as compared to ERα-negative samples in two independent breast cancer (BCa) patient cohorts (101 and 1302 samples). Additionally, the miR-135b expression is higher in AR-low PCa patient samples (47 samples). We identify ERα as a novel miR-135b target by demonstrating miR-135b binding to the 3'UTR of the ERα and decreased ERα protein and mRNA level upon miR-135b overexpression in BCa cells. MiR-135b reduces proliferation of ERα-positive BCa cells MCF-7 and BT-474 as well as AR-positive PCa cells LNCaP and 22Rv1 when grown in 2D. To identify other genes regulated by miR-135b we performed gene expression studies and found a link to the hypoxia inducible factor 1α (HIF1α) pathway. We show that miR-135b influences the protein level of the inhibitor for hypoxia inducible factor 1α (HIF1AN) and is able to bind to HIF1AN 3'UTR. Our study demonstrates that miR-135b regulates ERα, AR and HIF1AN protein levels through interaction with their 3'UTR regions, and proliferation in ERα-positive BCa and AR-positive PCa cells.
Collapse
Affiliation(s)
- Anna Aakula
- Institute for Molecular Medicine Finland, FIMM, Helsinki, Finland; VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland; Turku Centre for Biotechnology, University of Turku, Turku, Finland.
| | - Suvi-Katri Leivonen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway; The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Tero Aittokallio
- Institute for Molecular Medicine Finland, FIMM, Helsinki, Finland
| | - Yvonne Ceder
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway; The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Merja Perälä
- VTT Technical Research Centre of Finland, Medical Biotechnology, Turku, Finland
| | - Päivi Östling
- Institute for Molecular Medicine Finland, FIMM, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland, FIMM, Helsinki, Finland
| |
Collapse
|
80
|
Xuan H, Xue W, Pan J, Sha J, Dong B, Huang Y. Downregulation of miR-221, -30d, and -15a contributes to pathogenesis of prostate cancer by targeting Bmi-1. BIOCHEMISTRY (MOSCOW) 2015; 80:276-83. [DOI: 10.1134/s0006297915030037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
81
|
Torres IO, Kuchenbecker KM, Nnadi CI, Fletterick RJ, Kelly MJS, Fujimori DG. Histone demethylase KDM5A is regulated by its reader domain through a positive-feedback mechanism. Nat Commun 2015; 6:6204. [PMID: 25686748 DOI: 10.1038/ncomms7204] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 01/05/2015] [Indexed: 12/13/2022] Open
Abstract
The retinoblastoma binding protein KDM5A removes methyl marks from lysine 4 of histone H3 (H3K4). Misregulation of KDM5A contributes to the pathogenesis of lung and gastric cancers. In addition to its catalytic jumonji C domain, KDM5A contains three PHD reader domains, commonly recognized as chromatin recruitment modules. It is unknown whether any of these domains in KDM5A have functions beyond recruitment and whether they regulate the catalytic activity of the demethylase. Here using biochemical and nuclear magnetic resonance (NMR)-based structural studies, we show that the PHD1 preferentially recognizes unmethylated H3K4 histone tail, product of KDM5A-mediated demethylation of tri-methylated H3K4 (H3K4me3). Binding of unmodified H3 peptide to the PHD1 stimulates catalytic domain-mediated removal of methyl marks from H3K4me3 peptide and nucleosome substrates. This positive-feedback mechanism--enabled by the functional coupling between a reader and a catalytic domain in KDM5A--suggests a model for the spread of demethylation on chromatin.
Collapse
Affiliation(s)
- Idelisse Ortiz Torres
- 1] Department of Cellular and Molecular Pharmacology, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA [2] Chemistry and Chemical Biology Graduate Program, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA
| | - Kristopher M Kuchenbecker
- 1] Department of Biochemistry and Biophysics, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA [2] Biophysics Graduate Program, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA
| | - Chimno I Nnadi
- 1] Department of Cellular and Molecular Pharmacology, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA [2] Chemistry and Chemical Biology Graduate Program, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA [3] UCSF Medical Scientist Training Program, University of California, 513 Parnassus Avenue, San Francisco, California 94143, USA
| | - Robert J Fletterick
- Department of Biochemistry and Biophysics, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA
| | - Mark J S Kelly
- Department of Pharmaceutical Chemistry, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA
| | - Danica Galonić Fujimori
- 1] Department of Cellular and Molecular Pharmacology, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA [2] Department of Pharmaceutical Chemistry, University of California, 600 16th Street, Genentech Hall, San Francisco, California 94158, USA
| |
Collapse
|
82
|
Translationally controlled tumor protein in prostatic adenocarcinoma: correlation with tumor grading and treatment-related changes. BIOMED RESEARCH INTERNATIONAL 2015; 2015:985950. [PMID: 25667934 PMCID: PMC4312572 DOI: 10.1155/2015/985950] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/21/2014] [Accepted: 09/30/2014] [Indexed: 12/02/2022]
Abstract
Prostate cancer is the second leading cause of cancer-related death. The androgen deprivation therapy is the standard treatment for advanced stages. Unfortunately, virtually all tumors become resistant to androgen withdrawal. The progression to castration-resistance is not fully understood, although a recent paper has suggested translationally controlled tumor protein to be implicated in the process. The present study was designed to investigate the role of this protein in prostate cancer, focusing on the correlation between its expression level with tumor differentiation and response to treatment. We retrieved 292 prostatic cancer specimens; of these 153 had been treated only by radical prostatectomy and 139 had undergone radical prostatectomy after neoadjuvant treatment with combined androgen blockade therapy. Non-neoplastic controls were represented by 102 prostatic peripheral zone specimens. In untreated patients, the expression of the protein, evaluated by RT-qPCR and immunohistochemistry, was significantly higher in tumor specimens than in non-neoplastic control, increasing as Gleason pattern and score progressed. In treated prostates, the staining was correlated with the response to treatment. An association between protein expression and the main clinicopathological factors involved in prostate cancer aggressiveness was identified. These findings suggest that the protein may be a promising prognostic factor and a target for therapy.
Collapse
|
83
|
Abstract
The introduction of novel technologies that can be applied to the investigation of the molecular underpinnings of human cancer has allowed for new insights into the mechanisms associated with tumor development and progression. They have also advanced the diagnosis, prognosis and treatment of cancer. These technologies include microarray and other analysis methods for the generation of large-scale gene expression data on both mRNA and miRNA, next-generation DNA sequencing technologies utilizing a number of platforms to perform whole genome, whole exome, or targeted DNA sequencing to determine somatic mutational differences and gene rearrangements, and a variety of proteomic analysis platforms including liquid chromatography/mass spectrometry (LC/MS) analysis to survey alterations in protein profiles in tumors. One other important advancement has been our current ability to survey the methylome of human tumors in a comprehensive fashion through the use of sequence-based and array-based methylation analysis (Bock et al., Nat Biotechnol 28:1106-1114, 2010; Harris et al., Nat Biotechnol 28:1097-1105, 2010). The focus of this chapter is to present and discuss the evidence for key genes involved in prostate tumor development, progression, or resistance to therapy that are regulated by methylation-induced silencing.
Collapse
Affiliation(s)
- Tawnya C McKee
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, Diagnostic Biomarkers and Technology Branch, National Cancer Institute, Bethesda, MD, 20892-7430, USA
| | | |
Collapse
|
84
|
Rajendiran S, Parwani AV, Hare RJ, Dasgupta S, Roby RK, Vishwanatha JK. MicroRNA-940 suppresses prostate cancer migration and invasion by regulating MIEN1. Mol Cancer 2014; 13:250. [PMID: 25406943 PMCID: PMC4246551 DOI: 10.1186/1476-4598-13-250] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/23/2014] [Indexed: 12/21/2022] Open
Abstract
Background MicroRNAs (miRNAs) are crucial molecules that regulate gene expression and hence pathways that are key to prostate cancer progression. These non-coding RNAs are highly deregulated in prostate cancer thus facilitating progression of the disease. Among the many genes that have gained importance in this disease, Migration and invasion enhancer 1 (MIEN1), a novel gene located next to HER2/neu in the 17q12 amplicon of the human chromosome, has been shown to enhance prostate cancer cell migration and invasion, two key processes in cancer progression. MIEN1 is differentially expressed between normal and cancer cells and tissues. Understanding the regulation of MIEN1 by microRNA may enable development of better targeting strategies. Methods The miRNAs that could target MIEN1 were predicted by in silico algorithms and microarray analysis. The validation for miRNA expression was performed by qPCR and northern blotting in cells and by in situ hybridization in tissues. MIEN1 and levels of other molecules upon miRNA regulation was determined by Western blotting, qPCR, and immunofluorescence. The functional effects of miRNA on cells were determined by wound healing cell migration, Boyden chamber cell invasion, clonal and colony formation assays. For knockdown or overexpression of the miRNA or overexpression of MIEN1 3′UTR, cells were transfected with the oligomiRs and plasmids, respectively. Results A novel miRNA, hsa-miR-940 (miR-940), identified and validated to be highly expressed in immortalized normal cells compared to cancer cells, is a regulator of MIEN1. Analysis of human prostate tumors and their matched normal tissues confirmed that miR-940 is highly expressed in the normal tissues compared to its low to negligible expression in the tumors. While MIEN1 is a direct target of miR-940, miR-940 alters MIEN1 RNA, in a quantity as well as cell dependent context, along with altering its downstream effectors. The miR-940 inhibited migratory and invasive potential of cells, attenuated their anchorage-independent growth ability, and increased E-cadherin expression, implicating its role in mesenchymal-to-epithelial transition (MET). Conclusions These results, for the first time, implicate miR-940, a regulator of MIEN1, as a promising novel diagnostic and prognostic tool for prostate cancer. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-250) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Jamboor K Vishwanatha
- From the Department of Molecular and Medical Genetics and Institute for Cancer Research, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX 76107, USA.
| |
Collapse
|
85
|
Cannistraci A, Di Pace AL, De Maria R, Bonci D. MicroRNA as new tools for prostate cancer risk assessment and therapeutic intervention: results from clinical data set and patients' samples. BIOMED RESEARCH INTERNATIONAL 2014; 2014:146170. [PMID: 25309903 PMCID: PMC4182080 DOI: 10.1155/2014/146170] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/18/2014] [Accepted: 08/21/2014] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) is one of the leading causes of cancer-related death in men. Despite considerable advances in prostate cancer early detection and clinical management, validation of new biomarkers able to predict the natural history of tumor progression is still necessary in order to reduce overtreatment and to guide therapeutic decisions. MicroRNAs are endogenous noncoding RNAs which offer a fast fine-tuning and energy-saving mechanism for posttranscriptional control of protein expression. Growing evidence indicate that these RNAs are able to regulate basic cell functions and their aberrant expression has been significantly correlated with cancer development. Therefore, detection of microRNAs in tumor tissues and body fluids represents a new tool for early diagnosis and patient prognosis prediction. In this review, we summarize current knowledge about microRNA deregulation in prostate cancer mainly focusing on the different clinical aspects of the disease. We also highlight the potential roles of microRNAs in PCa management, while also discussing several current challenges and needed future research.
Collapse
Affiliation(s)
- Alessio Cannistraci
- Departement of Hematology, Oncology and Molecular Medicine, Istituto Superiore Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Anna Laura Di Pace
- Departement of Hematology, Oncology and Molecular Medicine, Istituto Superiore Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Ruggero De Maria
- Regina Elena National Cancer Institute, Via Fermo Ognibene, 00144 Rome, Italy
| | - Désirée Bonci
- Departement of Hematology, Oncology and Molecular Medicine, Istituto Superiore Sanità, Viale Regina Elena 299, 00161 Rome, Italy
- Regina Elena National Cancer Institute, Via Fermo Ognibene, 00144 Rome, Italy
| |
Collapse
|
86
|
Yan B, Wang ZH, Liu JY, Tao ZF, Li XM, Qin J. Long noncoding RNAs: versatile players in biologcial processes and human disorders. Epigenomics 2014; 6:375-9. [PMID: 25333847 DOI: 10.2217/epi.14.29] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Biao Yan
- Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Zhen-Hua Wang
- The Institute of Digital Ocean, College of InformationTechnology, Shanghai Ocean University, Shanghai, China
| | - Jing-Yu Liu
- Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Zhi-Fu Tao
- Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xiu-Miao Li
- Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Jiang Qin
- Eye Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|