51
|
Long intergenic noncoding RNA 00305 sponges miR-136 to regulate the hypoxia induced apoptosis of vascular endothelial cells. Biomed Pharmacother 2017; 94:238-243. [DOI: 10.1016/j.biopha.2017.07.099] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/11/2022] Open
|
52
|
Brady LK, Wang H, Radens CM, Bi Y, Radovich M, Maity A, Ivan C, Ivan M, Barash Y, Koumenis C. Transcriptome analysis of hypoxic cancer cells uncovers intron retention in EIF2B5 as a mechanism to inhibit translation. PLoS Biol 2017; 15:e2002623. [PMID: 28961236 PMCID: PMC5636171 DOI: 10.1371/journal.pbio.2002623] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 10/11/2017] [Accepted: 09/07/2017] [Indexed: 01/09/2023] Open
Abstract
Cells adjust to hypoxic stress within the tumor microenvironment by downregulating energy-consuming processes including translation. To delineate mechanisms of cellular adaptation to hypoxia, we performed RNA-Seq of normoxic and hypoxic head and neck cancer cells. These data revealed a significant down regulation of genes known to regulate RNA processing and splicing. Exon-level analyses classified > 1,000 mRNAs as alternatively spliced under hypoxia and uncovered a unique retained intron (RI) in the master regulator of translation initiation, EIF2B5. Notably, this intron was expressed in solid tumors in a stage-dependent manner. We investigated the biological consequence of this RI and demonstrate that its inclusion creates a premature termination codon (PTC), that leads to a 65kDa truncated protein isoform that opposes full-length eIF2Bε to inhibit global translation. Furthermore, expression of 65kDa eIF2Bε led to increased survival of head and neck cancer cells under hypoxia, providing evidence that this isoform enables cells to adapt to conditions of low oxygen. Additional work to uncover -cis and -trans regulators of EIF2B5 splicing identified several factors that influence intron retention in EIF2B5: a weak splicing potential at the RI, hypoxia-induced expression and binding of the splicing factor SRSF3, and increased binding of total and phospho-Ser2 RNA polymerase II specifically at the intron retained under hypoxia. Altogether, these data reveal differential splicing as a previously uncharacterized mode of translational control under hypoxia and are supported by a model in which hypoxia-induced changes to cotranscriptional processing lead to selective retention of a PTC-containing intron in EIF2B5.
Collapse
Affiliation(s)
- Lauren K. Brady
- Department of Radiation Oncology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Cellular and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Hejia Wang
- Department of Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Caleb M. Radens
- Cellular and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Yue Bi
- Department of Radiation Oncology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Oncology Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Milan Radovich
- Indiana University Health Precision Genomics Program, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Amit Maity
- Department of Radiation Oncology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Cristina Ivan
- Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Mircea Ivan
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Yoseph Barash
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
- Department of Computer and Information Science, University of Pennsylvania, Philadelphia, United States of America
| | - Constantinos Koumenis
- Department of Radiation Oncology Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
53
|
Boyle EC, Sedding DG, Haverich A. Targeting vasa vasorum dysfunction to prevent atherosclerosis. Vascul Pharmacol 2017; 96-98:5-10. [DOI: 10.1016/j.vph.2017.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/15/2017] [Indexed: 01/19/2023]
|
54
|
Circular RNA hsa_circ_0010729 regulates vascular endothelial cell proliferation and apoptosis by targeting the miR-186/HIF-1α axis. Biochem Biophys Res Commun 2017; 490:104-110. [PMID: 28571741 DOI: 10.1016/j.bbrc.2017.05.164] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 05/28/2017] [Indexed: 12/21/2022]
Abstract
Circular RNAs (circRNAs) are a group of non-protein-coding RNAs generated from back splicing. Emerging evidence has demonstrated its vital regulation on angiogenesis. However, the underlying mechanism responsible for circRNAs effects on vascular endothelial cells is still unclear. In the present study, we screened the expression profiles and investigated the physiological role of circRNAs in hypoxia-induced human umbilical vein endothelial cells (HUVECs). Using circRNA microarray analysis, we identified 36 circRNAs that were significantly dysregulated including 14 down-regulated circRNAs and 22 up-regulated with 2-fold change (P < 0.05). From the over-expressed circRNAs, hsa_circ_0010729 was selected as candidate circRNA and which was validated to be significantly up-regulated using RT-PCR. In loss-of-function experiments of HUVECs, hsa_circ_0010729 knockdown suppressed the proliferation and migration ability and enhanced apoptosis. Bioinformatic prediction and luciferase assay revealed that hsa_circ_0010729 and hypoxia inducible factor 1 alpha (HIF-1α) were targeted by miR-186. Validation experiments verified that hsa_circ_0010729 was co-expressed with HIF-1α, being negatively correlated with miR-186. Moreover, rescue experiments demonstrated that miR-186 inhibitor could reverse the role of hsa_circ_0010729 knockdown on HUVECs progression. Overall, the present study identifies the crucial regulation of hsa_circ_0010729 on vascular endothelial cell proliferation and apoptosis via targeting miR-186/HIF-1α axis.
Collapse
|
55
|
Eichhorn L, Dolscheid-Pommerich R, Erdfelder F, Ayub MA, Schmitz T, Werner N, Jansen F. Sustained apnea induces endothelial activation. Clin Cardiol 2017; 40:704-709. [PMID: 28464406 DOI: 10.1002/clc.22720] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Apnea diving has gained worldwide popularity, even though the pathophysiological consequences of this challenging sport on the human body are poorly investigated and understood. This study aims to assess the influence of sustained apnea in healthy volunteers on circulating microparticles (MPs) and microRNAs (miRs), which are established biomarkers reflecting vascular function. HYPOTHESIS Short intermittent hypoxia due to voluntary breath-holding affects circulating levels of endothelial cell-derived MPs (EMPs) and endothelial cell-derived miRs. METHODS Under dry laboratory conditions, 10 trained apneic divers performed maximal breath-hold. Venous blood samples were taken, once before and at 4 defined points in time after apnea. Samples were analyzed for circulating EMPs and endothelial miRs. RESULTS Average apnea time was 329 seconds (±103), and SpO2 at the end of apnea was 79% (±12). Apnea was associated with a time-dependent increase of circulating endothelial cell-derived EMPs and endothelial miRs. Levels of circulating EMPs in the bloodstream reached a peak 4 hours after the apnea period and returned to baseline levels after 24 hours. Circulating miR-126 levels were elevated at all time points after a single voluntary maximal apnea, whereas miR-26 levels were elevated significantly only after 30 minutes and 4 hours. Also miR-21 and miR-92 levels increased, but did not reach the level of significance. CONCLUSIONS Even a single maximal breath-hold induces acute endothelial activation and should be performed with great caution by subjects with preexisting vascular diseases. Voluntary apnea might be used as a model to simulate changes in endothelial function caused by hypoxia in humans.
Collapse
Affiliation(s)
- Lars Eichhorn
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Bonn, Bonn, Germany
| | | | - Felix Erdfelder
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Bonn, Bonn, Germany
| | | | - Theresa Schmitz
- Department of Medicine II, Heart Center Bonn, University Hospital of Bonn, Bonn, Germany
| | - Nikos Werner
- Department of Medicine II, Heart Center Bonn, University Hospital of Bonn, Bonn, Germany
| | - Felix Jansen
- Department of Medicine II, Heart Center Bonn, University Hospital of Bonn, Bonn, Germany
| |
Collapse
|
56
|
Bertero T, Rezzonico R, Pottier N, Mari B. Impact of MicroRNAs in the Cellular Response to Hypoxia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 333:91-158. [PMID: 28729029 DOI: 10.1016/bs.ircmb.2017.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In mammalian cells, hypoxia, or inadequate oxygen availability, regulates the expression of a specific set of MicroRNAs (MiRNAs), termed "hypoxamiRs." Over the past 10 years, the appreciation of the importance of hypoxamiRs in regulating the cellular adaptation to hypoxia has grown dramatically. At the cellular level, each hypoxamiR, including the master hypoxamiR MiR-210, can simultaneously regulate expression of multiple target genes in order to fine-tune the adaptive response of cells to hypoxia. This review addresses the complex molecular regulation of MiRNAs in both physiological and pathological conditions of low oxygen adaptation and the multiple functions of hypoxamiRs in various hypoxia-associated biological processes, including apoptosis, survival, proliferation, angiogenesis, inflammation, and metabolism. From a clinical perspective, we also discuss the potential use of hypoxamiRs as new biomarkers and/or therapeutic targets in cancer and aging-associated diseases including cardiovascular and fibroproliferative disorders.
Collapse
Affiliation(s)
- Thomas Bertero
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Nice, France
| | - Roger Rezzonico
- Université Côte d'Azur, CNRS, IPMC, FHU-OncoAge, Sophia-Antipolis, France
| | | | - Bernard Mari
- Université Côte d'Azur, CNRS, IPMC, FHU-OncoAge, Sophia-Antipolis, France.
| |
Collapse
|
57
|
Li P, Yin YL, Guo T, Sun XY, Ma H, Zhu ML, Zhao FR, Xu P, Chen Y, Wan GR, Jiang F, Peng QS, Liu C, Liu LY, Wang SX. Inhibition of Aberrant MicroRNA-133a Expression in Endothelial Cells by Statin Prevents Endothelial Dysfunction by Targeting GTP Cyclohydrolase 1 in Vivo. Circulation 2016; 134:1752-1765. [PMID: 27765794 PMCID: PMC5120771 DOI: 10.1161/circulationaha.116.017949] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 09/20/2016] [Indexed: 12/17/2022]
Abstract
Supplemental Digital Content is available in the text. Background: GTP cyclohydrolase 1 (GCH1) deficiency is critical for endothelial nitric oxide synthase uncoupling in endothelial dysfunction. MicroRNAs (miRs) are a class of regulatory RNAs that negatively regulate gene expression. We investigated whether statins prevent endothelial dysfunction via miR-dependent GCH1 upregulation. Methods: Endothelial function was assessed by measuring acetylcholine-induced vasorelaxation in the organ chamber. MiR-133a expression was assessed by quantitative reverse transcription polymerase chain reaction and fluorescence in situ hybridization. Results: We first demonstrated that GCH1 mRNA is a target of miR-133a. In endothelial cells, miR-133a was robustly induced by cytokines/oxidants and inhibited by lovastatin. Furthermore, lovastatin upregulated GCH1 and tetrahydrobiopterin, and recoupled endothelial nitric oxide synthase in stressed endothelial cells. These actions of lovastatin were abolished by enforced miR-133a expression and were mirrored by a miR-133a antagomir. In mice, hyperlipidemia- or hyperglycemia-induced ectopic miR-133a expression in the vascular endothelium, reduced GCH1 protein and tetrahydrobiopterin levels, and impaired endothelial function, which were reversed by lovastatin or miR-133a antagomir. These beneficial effects of lovastatin in mice were abrogated by in vivo miR-133a overexpression or GCH1 knockdown. In rats, multiple cardiovascular risk factors including hyperglycemia, dyslipidemia, and hyperhomocysteinemia resulted in increased miR-133a vascular expression, reduced GCH1 expression, uncoupled endothelial nitric oxide synthase function, and induced endothelial dysfunction, which were prevented by lovastatin. Conclusions: Statin inhibits aberrant miR-133a expression in the vascular endothelium to prevent endothelial dysfunction by targeting GCH1. Therefore, miR-133a represents an important therapeutic target for preventing cardiovascular diseases.
Collapse
Affiliation(s)
- Peng Li
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Ya-Ling Yin
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Tao Guo
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Xue-Ying Sun
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Hui Ma
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Mo-Li Zhu
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Fan-Rong Zhao
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Ping Xu
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Yuan Chen
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Guang-Rui Wan
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Fan Jiang
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Qi-Sheng Peng
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Chao Liu
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Li-Ying Liu
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.)
| | - Shuang-Xi Wang
- From School of Pharmacy and School of Basic Medical Sciences, Xinxiang Medical University, China (P.L., Y.-L.Y., M.-L.Z., F.-R.Z., P.X., G.-R.W., S.-X.W.); The Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China (T.G., H.M., Y.C., F.J., S.-X.W.); Department of Pharmacology, School of Pharmacy, Central South University, Changsha, China (X.-Y.S., L.-Y.L.); The Key Laboratory for Zoonosis Research, Institute of Zoonosis, Jilin University, Changchun, China (Q.-S.P.); and Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China (C.L.).
| |
Collapse
|
58
|
MicroRNA Stability in FFPE Tissue Samples: Dependence on GC Content. PLoS One 2016; 11:e0163125. [PMID: 27649415 PMCID: PMC5029930 DOI: 10.1371/journal.pone.0163125] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/03/2016] [Indexed: 11/30/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs responsible for fine-tuning of gene expression at post-transcriptional level. The alterations in miRNA expression levels profoundly affect human health and often lead to the development of severe diseases. Currently, high throughput analyses, such as microarray and deep sequencing, are performed in order to identify miRNA biomarkers, using archival patient tissue samples. MiRNAs are more robust than longer RNAs, and resistant to extreme temperatures, pH, and formalin-fixed paraffin-embedding (FFPE) process. Here, we have compared the stability of miRNAs in FFPE cardiac tissues using next-generation sequencing. The mode read length in FFPE samples was 11 nucleotides (nt), while that in the matched frozen samples was 22 nt. Although the read counts were increased 1.7-fold in FFPE samples, compared with those in the frozen samples, the average miRNA mapping rate decreased from 32.0% to 9.4%. These results indicate that, in addition to the fragmentation of longer RNAs, miRNAs are to some extent degraded in FFPE tissues as well. The expression profiles of total miRNAs in two groups were highly correlated (0.88 <r < 0.92). However, the relative read count of each miRNA was different depending on the GC content (p<0.0001). The unequal degradation of each miRNA affected the abundance ranking in the library, and miR-133a was shown to be the most abundant in FFPE cardiac tissues instead of miR-1, which was predominant before fixation. Subsequent quantitative PCR (qPCR) analyses revealed that miRNAs with GC content of less than 40% are more degraded than GC-rich miRNAs (p<0.0001). We showed that deep sequencing data obtained using FFPE samples cannot be directly compared with that of fresh frozen samples. The combination of miRNA deep sequencing and other quantitative analyses, such as qPCR, may improve the utility of archival FFPE tissue samples.
Collapse
|
59
|
Chistiakov DA, Orekhov AN, Bobryshev YV. The role of miR-126 in embryonic angiogenesis, adult vascular homeostasis, and vascular repair and its alterations in atherosclerotic disease. J Mol Cell Cardiol 2016; 97:47-55. [DOI: 10.1016/j.yjmcc.2016.05.007] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/19/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
|
60
|
Fritz JV, Heintz-Buschart A, Ghosal A, Wampach L, Etheridge A, Galas D, Wilmes P. Sources and Functions of Extracellular Small RNAs in Human Circulation. Annu Rev Nutr 2016; 36:301-36. [PMID: 27215587 PMCID: PMC5479634 DOI: 10.1146/annurev-nutr-071715-050711] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Various biotypes of endogenous small RNAs (sRNAs) have been detected in human circulation, including microRNAs, transfer RNAs, ribosomal RNA, and yRNA fragments. These extracellular sRNAs (ex-sRNAs) are packaged and secreted by many different cell types. Ex-sRNAs exhibit differences in abundance in several disease states and have, therefore, been proposed for use as effective biomarkers. Furthermore, exosome-borne ex-sRNAs have been reported to elicit physiological responses in acceptor cells. Exogenous ex-sRNAs derived from diet (most prominently from plants) and microorganisms have also been reported in human blood. Essential issues that remain to be conclusively addressed concern the (a) presence and sources of exogenous ex-sRNAs in human bodily fluids, (b) detection and measurement of ex-sRNAs in human circulation, (c) selectivity of ex-sRNA export and import, (d) sensitivity and specificity of ex-sRNA delivery to cellular targets, and (e) cell-, tissue-, organ-, and organism-wide impacts of ex-sRNA-mediated cell-to-cell communication. We survey the present state of knowledge of most of these issues in this review.
Collapse
MESH Headings
- Animals
- Biological Transport
- Biomarkers/blood
- Cell Communication
- Diet
- Gastrointestinal Microbiome/immunology
- Gene Expression Regulation
- Host-Parasite Interactions
- Host-Pathogen Interactions
- Humans
- Immunity, Innate
- MicroRNAs/blood
- MicroRNAs/metabolism
- Models, Biological
- RNA, Bacterial/blood
- RNA, Bacterial/metabolism
- RNA, Plant/blood
- RNA, Plant/metabolism
- RNA, Ribosomal/blood
- RNA, Ribosomal/metabolism
- RNA, Small Interfering/blood
- RNA, Small Interfering/metabolism
- RNA, Small Untranslated/blood
- RNA, Small Untranslated/metabolism
- RNA, Transfer/blood
- RNA, Transfer/metabolism
- RNA, Viral/blood
- RNA, Viral/metabolism
Collapse
Affiliation(s)
- Joëlle V Fritz
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
| | - Anna Heintz-Buschart
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
| | - Anubrata Ghosal
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Linda Wampach
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
| | - Alton Etheridge
- Pacific Northwest Diabetes Research Institute, Seattle, Washington 98122
| | - David Galas
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
- Pacific Northwest Diabetes Research Institute, Seattle, Washington 98122
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
| |
Collapse
|
61
|
Liu XS, Fan BY, Pan WL, Li C, Levin AM, Wang X, Zhang RL, Zervos TM, Hu J, Zhang XM, Chopp M, Zhang ZG. Identification of miRNomes associated with adult neurogenesis after stroke using Argonaute 2-based RNA sequencing. RNA Biol 2016; 14:488-499. [PMID: 27315491 DOI: 10.1080/15476286.2016.1196320] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurogenesis is associated with functional recovery after stroke. However, the underlying molecular mechanisms have not been fully investigated. Using an Ago2-based RNA immunoprecipitation to immunoprecipated Ago2-RNA complexes followed by RNA sequencing (Ago2 RIP-seq) approach, we profiled the miRNomes in neural progenitor cells (NPCs) harvested from the subventricular zone (SVZ) of the lateral ventricles of young adult rats. We identified more than 7 and 15 million reads in normal and ischemic NPC libraries, respectively. We found that stroke substantially changed Ago2-associated miRNA profiles in NPCs compared to those in non-ischemic NPCs. We also discovered a new complex repertoire of isomiRs and multiple miRNA-miRNA* pairs and numerous novel miRNAs in the non-ischemic and ischemic NPCs. Among them, pc-3p-17172 significantly regulated NPC proliferation and neuronal differentiation. Collectively, the present study reveals profiles of Ago2-associated miRNomes in non-ischemic and ischemic NPCs, which provide a molecular basis to further investigate the role of miRNAs in mediating adult neurogenesis under physiological and ischemic conditions.
Collapse
Affiliation(s)
- Xian Shuang Liu
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA
| | - Bao Yan Fan
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA
| | - Wan Long Pan
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA.,b Sichuan Key Laboratory of Medical Imaging and Department of Immunology , North Sichuan Medical University , Nanchong , Sichuan , China
| | - Chao Li
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA
| | - Albert M Levin
- c Department of Public Health Sciences , Henry Ford Health System , Detroit , MI , USA.,d Center for Bioinformatics , Henry Ford Health System , Detroit , MI , USA
| | - Xinli Wang
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA
| | - Rui Lan Zhang
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA
| | - Thomas M Zervos
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA
| | - Jiani Hu
- e Department of Radiology , Wayne State University , Detroit , MI , USA
| | - Xiao Ming Zhang
- f Sichuan Key Laboratory of Medical Imaging and Department of Radiology , Affiliated Hospital of North Sichuan Medical University , Nanchong , Sichuan , China
| | - Michael Chopp
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA.,g Department of Physics , Oakland University , Rochester , MI , USA
| | - Zheng Gang Zhang
- a Department of Neurology , Henry Ford Health System , Detroit , MI , USA
| |
Collapse
|
62
|
Schober A, Weber C. Mechanisms of MicroRNAs in Atherosclerosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:583-616. [DOI: 10.1146/annurev-pathol-012615-044135] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Andreas Schober
- Institute for Cardiovascular Prevention, Ludwig Maximilians University Munich, Munich 80336, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich 80336, Germany;
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig Maximilians University Munich, Munich 80336, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich 80336, Germany;
| |
Collapse
|
63
|
Poersch A, Grassi ML, Carvalho VPD, Lanfredi GP, Palma CDS, Greene LJ, de Sousa CB, Carrara HHA, Candido Dos Reis FJ, Faça VM. A proteomic signature of ovarian cancer tumor fluid identified by highthroughput and verified by targeted proteomics. J Proteomics 2016; 145:226-236. [PMID: 27222041 DOI: 10.1016/j.jprot.2016.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/27/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
Abstract
UNLABELLED Tumor fluid samples have emerged as a rich source for the identification of ovarian cancer in the context of proteomics studies. To uncover differences among benign and malignant ovarian samples, we performed a quantitative proteomic study consisting of albumin immunodepletion, isotope labeling with acrylamide and in-depth proteomic profiling by LC-MS/MS in a pool of 10 samples of each histological type. 1135 proteins were identified, corresponding to 505 gene products. 223 proteins presented associated quantification and the comparative analysis of histological types revealed 75 differentially abundant proteins. Based on this, we developed a panel for targeted proteomic analysis using the multiple reaction monitoring (MRM) method for validation of 51 proteins in individual samples of high-grade serous ovarian tumor fluids (malignant) and benign serous cystadenoma tumor fluids. This analysis showed concordant results in terms of average amounts of proteins, and APOE, SERPINF2, SERPING1, ADAM17, CD44 and OVGP1 were statistically significant between benign and malignant group. The results observed in the MRM for APOE were confirmed by western blotting, where APOE was more abundant in malignant samples. This molecular signature can contribute to improve tumor stratification and shall be investigated in combination with current biomarkers in larger cohorts to improve ovarian cancer diagnosis. BIOLOGICAL SIGNIFICANCE Despite advances in cancer research, ovarian cancer has a high mortality and remains a major challenge due to a number of particularities of the disease, especially late diagnosis caused by vague clinical symptoms, the cellular and molecular heterogeneity of tumors, and the lack of effective treatment. Thus, efforts are directed to better understand this neoplasia, its origin, development and, particularly the identification and validation of biomarkers for early detection of the disease in asymptomatic stage. In the present work, we confirmed by MRM method in individual ovarian tumor fluid samples the regulation of 27 proteins out of 33 identified in a highthroughput study. We speculate that the presence and/or differential abundance observed in tumor fluid is a cooperation primarily of high rates of secretion of such tumor proteins to extra tumor environment that will at the end accumulate in plasma, and also the accumulation of acute-phase proteins throughout the entire body. On top of that, consideration of physiological influences in the interpretation of expression observed, including age, menopause status, route-of-elimination kinetics and metabolism of the tumor marker, coexisting disease, hormonal imbalances, life-style influences (smoking, alcoholism, obesity), among others, are mandatory to enable the selection of good protein tumor marker candidates for extensive validation.
Collapse
Affiliation(s)
- Aline Poersch
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Department of Gynecology and Obstetrics, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil.
| | - Mariana Lopes Grassi
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil
| | - Vinícius Pereira de Carvalho
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Medical School of Barão de Mauá University, Rua Ramos de Azevedo, 423, 14090-180 Ribeirão Preto, SP, Brazil
| | - Guilherme Pauperio Lanfredi
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Camila de Souza Palma
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil
| | - Lewis Joel Greene
- Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil; Department of Cell and Molecular Biology and Pathogenic Bioagents, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Christiani Bisinoto de Sousa
- Department of Gynecology and Obstetrics, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Hélio Humberto Angotti Carrara
- Department of Gynecology and Obstetrics, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Francisco José Candido Dos Reis
- Department of Gynecology and Obstetrics, FMRP, University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, FMRP,University of São Paulo, Av. Bandeirantes, 3900, 14040-900 Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, Hemotherapy Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, 14051-140, Ribeirão Preto, SP, Brazil
| |
Collapse
|
64
|
Abstract
Activation of hypoxia pathways is both associated with and contributes to an aggressive phenotype across multiple types of solid cancers. The regulation of gene transcription by hypoxia-inducible factor (HIF) is a key element in this response. HIF directly upregulates the expression of many hundreds of protein-coding genes, which act to both improve oxygen delivery and to reduce oxygen demand. However, it is now becoming apparent that many classes of noncoding RNAs are also regulated by hypoxia, with several (e.g. micro RNAs, long noncoding RNAs and antisense RNAs) under direct transcriptional regulation by HIF. These hypoxia-regulated, noncoding RNAs may act as effectors of the indirect response to HIF by acting on specific coding transcripts or by affecting generic RNA-processing pathways. In addition, noncoding RNAs may also act as modulators of the HIF pathway, either by integrating other physiological responses or, in the case of HIF-regulated, noncoding RNAs, by providing negative or positive feedback and feedforward loops that affect upstream or downstream components of the HIF cascade. These hypoxia-regulated, noncoding transcripts play important roles in the aggressive hypoxic phenotype observed in cancer.
Collapse
|
65
|
Greco S, Zaccagnini G, Voellenkle C, Martelli F. microRNAs in ischaemic cardiovascular diseases. Eur Heart J Suppl 2016; 18:E31-E36. [PMID: 28533714 DOI: 10.1093/eurheartj/suw012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
microRNAs (miRNAs) are non-coding RNA molecules that modulate the stability and/or the translational efficiency of specific messenger RNAs. They have been shown to play a regulatory role in most biological processes and their expression is disrupted in many cardiovascular diseases. This review describes studies performed at Policlinico San Donato-IRCCS in cell cultures, animal models, and patients, showing a penetrant role of miRNAs in cell response to hypoxia and in ischaemic cardiovascular diseases. These experiments indicate miRNA as an emerging class of therapeutic targets.
Collapse
Affiliation(s)
- Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, via Morandi 30, 20097 San Donato Milanese, Milan, Italy
| | - Germana Zaccagnini
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, via Morandi 30, 20097 San Donato Milanese, Milan, Italy
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, via Morandi 30, 20097 San Donato Milanese, Milan, Italy
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, via Morandi 30, 20097 San Donato Milanese, Milan, Italy
| |
Collapse
|
66
|
Ouimet M, Koster S, Sakowski E, Ramkhelawon B, van Solingen C, Oldebeken S, Karunakaran D, Portal-Celhay C, Sheedy FJ, Ray TD, Cecchini K, Zamore PD, Rayner KJ, Marcel YL, Philips JA, Moore KJ. Mycobacterium tuberculosis induces the miR-33 locus to reprogram autophagy and host lipid metabolism. Nat Immunol 2016; 17:677-86. [PMID: 27089382 PMCID: PMC4873392 DOI: 10.1038/ni.3434] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 03/11/2016] [Indexed: 12/16/2022]
Abstract
Mycobacterium tuberculosis (Mtb) survives in macrophages by evading delivery to the lysosome and promoting the accumulation of lipid bodies, which serve as a bacterial source of nutrients. We found that by inducing the microRNA (miRNA) miR-33 and its passenger strand miR-33*, Mtb inhibited integrated pathways involved in autophagy, lysosomal function and fatty acid oxidation to support bacterial replication. Silencing of miR-33 and miR-33* by genetic or pharmacological means promoted autophagy flux through derepression of key autophagy effectors (such as ATG5, ATG12, LC3B and LAMP1) and AMPK-dependent activation of the transcription factors FOXO3 and TFEB, which enhanced lipid catabolism and Mtb xenophagy. These data define a mammalian miRNA circuit used by Mtb to coordinately inhibit autophagy and reprogram host lipid metabolism to enable intracellular survival and persistence in the host.
Collapse
Affiliation(s)
- Mireille Ouimet
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Stefan Koster
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Erik Sakowski
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Bhama Ramkhelawon
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Coen van Solingen
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Scott Oldebeken
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Denuja Karunakaran
- University of Ottawa Heart Institute and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Cynthia Portal-Celhay
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Frederick J Sheedy
- Department of Clinical Medicine, School of Medicine, Trinity College, Dublin, Ireland
| | - Tathagat Dutta Ray
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Katharine Cecchini
- RNA Therapeutics Institute, Howard Hughes Medical Institute, and Department of Biochemistry &Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Philip D Zamore
- RNA Therapeutics Institute, Howard Hughes Medical Institute, and Department of Biochemistry &Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Katey J Rayner
- University of Ottawa Heart Institute and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Yves L Marcel
- University of Ottawa Heart Institute and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Jennifer A Philips
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Medical Center, New York, New York, USA
| | - Kathryn J Moore
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York, USA
| |
Collapse
|
67
|
Implication of Long noncoding RNAs in the endothelial cell response to hypoxia revealed by RNA-sequencing. Sci Rep 2016; 6:24141. [PMID: 27063004 PMCID: PMC4827084 DOI: 10.1038/srep24141] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/21/2016] [Indexed: 01/01/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are non-protein coding RNAs regulating gene expression. Although for some lncRNAs a relevant role in hypoxic endothelium has been shown, the regulation and function of lncRNAs is still largely unknown in the vascular physio-pathology. Taking advantage of next-generation sequencing techniques, transcriptomic changes induced by endothelial cell exposure to hypoxia were investigated. Paired-end sequencing of polyadenylated RNA derived from human umbilical vein endothelial cells (HUVECs) exposed to 1% O2 or normoxia was performed. Bioinformatics analysis identified ≈2000 differentially expressed genes, including 122 lncRNAs. Extensive validation was performed by both microarray and qPCR. Among the validated lncRNAs, H19, MIR210HG, MEG9, MALAT1 and MIR22HG were also induced in a mouse model of hindlimb ischemia. To test the functional relevance of lncRNAs in endothelial cells, knockdown of H19 expression was performed. H19 inhibition decreased HUVEC growth, inducing their accumulation in G1 phase of the cell cycle; accordingly, p21 (CDKN1A) expression was increased. Additionally, H19 knockdown also diminished HUVEC ability to form capillary like structures when plated on matrigel. In conclusion, a high-confidence signature of lncRNAs modulated by hypoxia in HUVEC was identified and a significant impact of H19 lncRNA was shown.
Collapse
|
68
|
Hu J, Ni S, Cao Y, Zhang T, Wu T, Yin X, Lang Y, Lu H. The Angiogenic Effect of microRNA-21 Targeting TIMP3 through the Regulation of MMP2 and MMP9. PLoS One 2016; 11:e0149537. [PMID: 26872030 PMCID: PMC4752282 DOI: 10.1371/journal.pone.0149537] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/02/2016] [Indexed: 11/18/2022] Open
Abstract
microRNAs are a novel set of small, non-protein-coding nucleotide RNAs that negatively regulate the expression of target mRNAs. miRNA-21 is a microRNA that is highly enriched in endothelial cells. miRNA-21 has been shown to be a potential pro-angiogenic factor in some biological systems. Our previous study showed that the expression of miRNA-21 was up-regulated after spinal cord injury. However, the effect of miRNA-21 on angiogenesis in the spinal cord was unclear. In this study, to understand the role of miRNA-21 on injured endothelial cells exclusively, an oxygen and glucose deprivation model of endothelial cells was constructed, and the up-regulation of miRNA-21 was discovered in this model. An increased level of miRNA-21 by mimics promoted the survival, migration and tube formation of endothelial cells, which simultaneously inhibited tissue inhibitor of metalloproteinase-3 (TIMP3) expression and promoted matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9) expression and secretion. A decreased level of miRNA-21 by antagomir exerted an opposite effect. As is well known, survival, migration and tube formation of endothelial cells are necessary prerequisites for angiogenesis after injury. TIMP3 was validated as a direct target of miRNA-21 by dual-luciferase reporter assay. Silencing with small interfering RNA against TIMP3 promoted tube formation and increased MMP2 and MMP9 expression at the protein level. In vivo, we found that decreased levels of miRNA-21 inhibited angiogenesis after spinal cord injury in rats using synchrotron radiation micro-computed tomography. In summary, these findings suggest that miRNA-21 has a protective effect on angiogenesis by reducing cell death and promoting cell survival, migration and tube formation via partially targeting the TIMP3 by potentially regulating MMP2 and MMP9. TIMP3 is a functional target gene. Identifying the role of miRNA-21 in the protection of angiogenesis might offer a novel therapeutic target for secondary spinal cord injury, in which angiogenesis is indispensable.
Collapse
Affiliation(s)
- Jianzhong Hu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Shuangfei Ni
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Yong Cao
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Tao Zhang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Tianding Wu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Xianzhen Yin
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Ye Lang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Hongbin Lu
- Department of Sports Medicine, Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China
- * E-mail:
| |
Collapse
|
69
|
Abstract
Recent investigations have highlighted the importance of the non-coding genome in regions of hypoxia in tumours. Such regions are frequently found in solid tumours, and are associated with worse patient survival and therapy resistance. Hypoxia stabilises the transcription factors, hypoxia inducible factors (HIF1α and HIF2α) which coordinate transcriptomic changes that occur in hypoxia. The changes in gene expression induced by HIF1α and HIF2α contribute to many of the hallmarks of cancer phenotypes and enable tumour growth, survival and invasion in the hypoxic tumour microenvironment. Non-coding RNAs, in particular microRNAs (miRNAs), which regulate mRNA stability and translation, and long-non-coding RNAs (lncRNAs), which have diverse functions including chromatin modification and transcriptional regulation, are also important in enabling the key hypoxia regulated processes. They have roles in the regulation of metabolism, angiogenesis, autophagy, invasion and metastasis in the hypoxic microenvironment. Furthermore, HIF1α and HIF2α expression and stabilisation are also regulated by both miRNAs and lncRNAs. Here we review the recent developments in the expression, regulation and functions of miRNAs, lncRNAs and other non-coding RNA classes in tumour hypoxia.
Collapse
Affiliation(s)
- Hani Choudhry
- Department of Biochemistry, Faculty of Science, Center of Innovation in Personalized Medicine, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adrian L Harris
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Alan McIntyre
- Cancer Biology, Division of Cancer and Stem Cells, QMC, University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
70
|
Kuosmanen SM, Viitala S, Laitinen T, Peräkylä M, Pölönen P, Kansanen E, Leinonen H, Raju S, Wienecke-Baldacchino A, Närvänen A, Poso A, Heinäniemi M, Heikkinen S, Levonen AL. The Effects of Sequence Variation on Genome-wide NRF2 Binding--New Target Genes and Regulatory SNPs. Nucleic Acids Res 2016; 44:1760-75. [PMID: 26826707 PMCID: PMC4770247 DOI: 10.1093/nar/gkw052] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 01/16/2016] [Indexed: 12/11/2022] Open
Abstract
Transcription factor binding specificity is crucial for proper target gene regulation. Motif discovery algorithms identify the main features of the binding patterns, but the accuracy on the lower affinity sites is often poor. Nuclear factor E2-related factor 2 (NRF2) is a ubiquitous redox-activated transcription factor having a key protective role against endogenous and exogenous oxidant and electrophile stress. Herein, we decipher the effects of sequence variation on the DNA binding sequence of NRF2, in order to identify both genome-wide binding sites for NRF2 and disease-associated regulatory SNPs (rSNPs) with drastic effects on NRF2 binding. Interactions between NRF2 and DNA were studied using molecular modelling, and NRF2 chromatin immunoprecipitation-sequence datasets together with protein binding microarray measurements were utilized to study binding sequence variation in detail. The binding model thus generated was used to identify genome-wide binding sites for NRF2, and genomic binding sites with rSNPs that have strong effects on NRF2 binding and reside on active regulatory elements in human cells. As a proof of concept, miR-126–3p and -5p were identified as NRF2 target microRNAs, and a rSNP (rs113067944) residing on NRF2 target gene (Ferritin, light polypeptide, FTL) promoter was experimentally verified to decrease NRF2 binding and result in decreased transcriptional activity.
Collapse
Affiliation(s)
- Suvi M Kuosmanen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Sari Viitala
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Tuomo Laitinen
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Mikael Peräkylä
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Petri Pölönen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland Institute of Biomedicine, School of Medicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Emilia Kansanen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Hanna Leinonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Suresh Raju
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | | | - Ale Närvänen
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Antti Poso
- School of Pharmacy, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Anna-Liisa Levonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| |
Collapse
|
71
|
Richart A, Loyer X, Néri T, Howangyin K, Guérin CL, Ngkelo A, Bakker W, Zlatanova I, Rouanet M, Vilar J, Lévy B, Rothenberg M, Mallat Z, Pucéat M, Silvestre JS. MicroRNA-21 coordinates human multipotent cardiovascular progenitors therapeutic potential. Stem Cells 2015; 32:2908-22. [PMID: 25069679 DOI: 10.1002/stem.1789] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/06/2014] [Accepted: 06/23/2014] [Indexed: 12/11/2022]
Abstract
Published clinical trials in patients with ischemic diseases show limited benefit of adult stem cell-based therapy, likely due to their restricted plasticity and commitment toward vascular cell lineage. We aim to uncover the potent regenerative ability of MesP1/stage-specific embryonic antigen 1 (SSEA-1)-expressing cardiovascular progenitors enriched from human embryonic stem cells (hESCs). Injection of only 10(4) hESC-derived SSEA-1(+) /MesP1(+) cells, or their progeny obtained after treatment with VEGF-A or PDGF-BB, was effective enough to enhance postischemic revascularization in immunodeficient mice with critical limb ischemia (CLI). However, the rate of incorporation of hESC-derived SSEA-1(+) /MesP1(+) cells and their derivatives in ischemic tissues was modest. Alternatively, these cells possessed a unique miR-21 signature that inhibited phosphotase and tensin homolog (PTEN) thereby activating HIF-1α and the systemic release of VEGF-A. Targeting miR-21 limited cell survival and inhibited their proangiogenic capacities both in the Matrigel model and in mice with CLI. We next assessed the impact of mR-21 in adult angiogenesis-promoting cells. We observed an impaired postischemic angiogenesis in miR-21-deficient mice. Notably, miR-21 was highly expressed in circulating and infiltrated monocytes where it targeted PTEN/HIF-1α/VEGF-A signaling and cell survival. As a result, miR-21-deficient mice displayed an impaired number of infiltrated monocytes and a defective angiogenic phenotype that could be partially restored by retransplantation of bone marrow-derived cells from wild-type littermates. hESC-derived SSEA-1(+) /MesP1(+) cells progenitor cells are powerful key integrators of therapeutic angiogenesis in ischemic milieu and miR-21 is instrumental in this process as well as in the orchestration of the biological activity of adult angiogenesis-promoting cells.
Collapse
Affiliation(s)
- Adèle Richart
- INSERM UMRS 970, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Circulating miR-200-family micro-RNAs have altered plasma levels in patients with endometriosis and vary with blood collection time. Fertil Steril 2015. [PMID: 26206343 DOI: 10.1016/j.fertnstert.2015.06.029] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To determine whether circulating micro-RNA (miR) 200a, miR-200b, and miR-141 have altered levels in patients with endometriosis compared with control individuals. DESIGN Experimental laboratory study. SETTING University. PATIENT(S) Patients with endometriosis (n = 61), laparoscopically confirmed endometriosis-free women (n = 35), and self-reported healthy women (n = 30) were included in the study. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Plasma miRNA levels in endometriosis patients and control subjects. RESULT(S) We found that the levels of studied miRNAs varied with blood collection time, being lower in the morning than in the evening. When blood collection time was taken into account, the results revealed significantly lower levels of miR-200a and miR-141 in the evening plasma samples of women with endometriosis compared with surgically confirmed disease-free patients. However, the evening-sample levels of all three miRNAs were significantly lower in patients with stage I-II endometriosis than in endometriosis-free control subjects. In cases of stage III-IV endometriosis, only miR-200a levels were significantly lower compared with patients without endometriosis. Circulating miR-200a showed the best discriminative power to differentiate women with endometriosis from patients with similar complaints but without the disease. CONCLUSION(S) Our findings suggest that miR-200a and miR-141 have a potential as novel noninvasive biomarkers for endometriosis. In addition, we found that the plasma miR-200a, miR-200b and miR-141 levels vary with blood sampling time, so it is important to take the sample collection time into account when studying miRNAs as biomarkers.
Collapse
|
73
|
Ben-Dayan MM, MacCarthy T, Schlecht NF, Belbin TJ, Childs G, Smith RV, Prystowsky MB, Bergman A. Cancer as the Disintegration of Robustness: Population-Level Variance in Gene Expression Identifies Key Differences Between Tobacco- and HPV-Associated Oropharyngeal Carcinogenesis. Arch Pathol Lab Med 2015; 139:1362-72. [PMID: 26132601 DOI: 10.5858/arpa.2014-0624-oa] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CONTEXT Oropharyngeal squamous cell carcinoma is associated both with tobacco use and with human papillomavirus (HPV) infection. It is argued that carcinogen-driven tumorigenesis is a distinct disease from its virally driven counterpart. We hypothesized that tumorigenesis is the result of a loss of genotypic robustness resulting in an increase in phenotypic variation in tumors compared with adjacent histologically normal tissues, and that carcinogen-driven tumorigenesis results in greater variation than its virally driven counterpart. OBJECTIVES To examine the loss of robustness in carcinogen-driven and virally driven oropharyngeal squamous cell carcinoma samples, and to identify potential pathways involved. DESIGN We used coefficients of variation for messenger RNA and microRNA expression to measure the loss of robustness in oropharyngeal squamous cell carcinoma samples. Tumors were compared with matched normal tissues, and were further categorized by HPV and patient smoking status. Weighted gene coexpression networks were constructed for genes with highly variable expression among the HPV⁻ tumors from smokers. RESULTS We observed more genes with variable messenger RNA expression in tumors compared with normal tissues, regardless of HPV and smoking status, and more microRNAs with variable expression in HPV⁻ and HPV⁺ tumors from smoking patients than from nonsmokers. For both the messenger RNA and microRNA data, we observed more variance among HPV⁻ tumors from smokers compared with HPV⁺ tumors from nonsmokers. The gene coexpression network construction highlighted pathways that have lost robustness in carcinogen-induced tumors but appear stable in virally induced tumors. CONCLUSIONS Using coefficients of variation and coexpression networks, we identified multiple altered pathways that may play a role in carcinogen-driven tumorigenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Aviv Bergman
- From the Departments of Pathology (Ms Ben-Dayan and Drs Belbin, Childs, and Prystowsky), Epidemiology and Population Health (Dr Schlecht), and Computational and Systems Biology (Dr Bergman), Albert Einstein College of Medicine, Bronx, New York; the Department of Applied Mathematics and Statistics, SUNY Stony Brook, Stony Brook, New York (Dr MacCarthy); and the Department of Otorhinolaryngology, Montefiore Medical Center, Bronx, New York (Dr Smith)
| |
Collapse
|
74
|
Šerý O, Paclt I, Drtílková I, Theiner P, Kopečková M, Zvolský P, Balcar VJ. A 40-bp VNTR polymorphism in the 3'-untranslated region of DAT1/SLC6A3 is associated with ADHD but not with alcoholism. Behav Brain Funct 2015; 11:21. [PMID: 26058807 PMCID: PMC4472402 DOI: 10.1186/s12993-015-0066-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/27/2015] [Indexed: 11/18/2022] Open
Abstract
Background ADHD and alcoholism are psychiatric diseases with pathophysiology related to dopamine system. DAT1 belongs to the SLC6 family of transporters and is involved in the regulation of extracellular dopamine levels. A 40 bp variable number tandem repeat (VNTR) polymorphism in the 3′-untranslated region of DAT1/SLC6A3 gene was previously reported to be associated with various phenotypes involving disturbed regulation of dopaminergic neurotransmission. Methods A total of 1312 subjects were included and genotyped for 40 bp VNTR polymorphism of DAT1/SLC6A3 gene in this study (441 alcoholics, 400 non-alcoholic controls, 218 ADHD children and 253 non ADHD children). Using miRBase software, we have performed a computer analysis of VNTR part of DAT1 gene for presence of miRNA binding sites. Results We have found significant relationships between ADHD and the 40 bp VNTR polymorphisms of DAT1/SLC6A3 gene (P < 0.01). The 9/9 genotype appeared to reduce the risk of ADHD about 0.4-fold (p < 0.04). We also noted an occurrence of rare genotypes in ADHD (frequency different from controls at p < 0.01). No association between alcoholism and genotype frequencies of 40 bp VNTR polymorphism of DAT1/SLC6A3 gene has been detected. Conclusions We have found an association between 40 bp VNTR polymorphism of DAT1/SLC6A3 gene and ADHD in the Czech population; in a broad agreement with studies in other population samples. Furthermore, we detected rare genotypes 8/10, 7/10 and 10/11 present in ADHD boys only and identified miRNAs that should be looked at as potential novel targets in the research on ADHD.
Collapse
Affiliation(s)
- Omar Šerý
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic. .,Institute of Animal Physiology and Genetics, Academy of Science, Veveří 97, 602 00, Brno, Czech Republic.
| | - Ivo Paclt
- Department of Psychiatry, 1st Faculty of Medicine, Charles University, Ke Karlovu 11, 121 08, Prague, Czech Republic.
| | - Ivana Drtílková
- Department of Psychiatry, Masaryk University, Faculty of Medicine, Kamenice 5, 625 00, Brno, Czech Republic.
| | - Pavel Theiner
- Department of Psychiatry, Masaryk University, Faculty of Medicine, Kamenice 5, 625 00, Brno, Czech Republic.
| | - Marta Kopečková
- Department of Psychiatry, 1st Faculty of Medicine, Charles University, Ke Karlovu 11, 121 08, Prague, Czech Republic.
| | - Petr Zvolský
- Department of Psychiatry, 1st Faculty of Medicine, Charles University, Ke Karlovu 11, 121 08, Prague, Czech Republic.
| | - Vladimir J Balcar
- Laboratory of Neurochemistry, School of Medical Sciences (Discipline of Anatomy and Histology) and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
75
|
Mao G, Liu Y, Fang X, Liu Y, Fang L, Lin L, Liu X, Wang N. Tumor-derived microRNA-494 promotes angiogenesis in non-small cell lung cancer. Angiogenesis 2015; 18:373-82. [PMID: 26040900 DOI: 10.1007/s10456-015-9474-5] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/25/2015] [Indexed: 11/29/2022]
Abstract
Angiogenesis, a crucial step in tumor growth and metastasis, is regulated by various pro- or anti-angiogenic factors. Recently, microRNAs have been shown to modulate angiogenic processes by modulating the expression of critical angiogenic factors. However, roles of tumor-derived microRNAs in regulating tumor vascularization remain to be elucidated. In this study, we found that delivery of miR-494 into human vascular endothelial cells (ECs) enhanced the EC migration and promoted angiogenesis. The angiogenic effect of miR-494 was mediated by the targeting of PTEN and the subsequent activation of Akt/eNOS pathway. Importantly, co-culture experiments demonstrated that a lung cancer cell line, A549, secreted and delivered miR-494 into ECs via a microvesicle-mediated route. In addition, we found that the expression of miR-494 was induced in the tumor cells in response to hypoxia, likely via a HIF-1α-mediated mechanism. Furthermore, a specific miR-494 antagomiR effectively inhibited angiogenesis and attenuated the growth of tumor xenografts in nude mice. Taken together, these results demonstrated that miR-494 is a novel tumor-derived paracrine signal to promote angiogenesis and tumor growth under hypoxic condition.
Collapse
Affiliation(s)
- Guangmei Mao
- Institute of Cardiovascular Science, Peking University Health Science Center, 38 Xueyuan Rd, Beijing, 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Liu C, Lin Q, Yun Z. Cellular and molecular mechanisms underlying oxygen-dependent radiosensitivity. Radiat Res 2015; 183:487-96. [PMID: 25938770 DOI: 10.1667/rr13959.1] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Molecular oxygen has long been recognized as a powerful radiosensitizer that enhances the cell-killing efficiency of ionizing radiation. Radiosensitization by oxygen occurs at very low concentrations with the half-maximum radiosensitization at approximately 3 mmHg. However, robust hypoxia-induced signal transduction can be induced at <15 mmHg and can elicit a wide range of cellular responses that will affect therapy response as well as malignant progression. Great strides have been made, especially since the 1990s, toward identification and characterization of the oxygen-regulated molecular pathways that affect tumor response to ionizing radiation. In this review, we will discuss the current advances in our understanding of oxygen-dependent molecular modification and cellular signal transduction and their impact on tumor response to therapy. We will specifically address mechanistic distinctions between radiobiological hypoxia (0-3 mmHg) and pathological hypoxia (3-15 mmHg). We also propose a paradigm that hypoxia increases radioresistance by maintaining the cancer stem cell phenotype.
Collapse
Affiliation(s)
- Chao Liu
- a Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut 06520
| | | | | |
Collapse
|
77
|
Abstract
Atherosclerosis is characterised by the accumulation of lipid-laden macrophages in atherosclerotic lesions and occurs preferentially at arterial branching points, which are prone to inflammation during hyperlipidaemic stress. The increased susceptibility at branching sites of arteries is attributable to poor adaptation of arterial endothelial cells to disturbed blood flow. In the past 5 years, several studies have provided mechanistic insights into the regulatory roles of microRNAs (miRNAs) in inflammatory activation, proliferation, and regeneration of endothelial cells during this maladaptive process. The intercellular transfer of vesicle-bound miRNAs contributes to arterial homeostasis, and the combinatorial effect of multiple miRNAs controls the unresolved inflammation orchestrated by macrophages in atherosclerotic lesions. In this Review, we highlight the miRNA-dependent regulation of the endothelial phenotype and the proliferative reserve that occurs in response to altered haemodynamic conditions as a prerequisite for atherogenic inflammation. In particular, we discuss the regulation of transcriptional modules by miRNAs and the protective role of complementary strand pairs, which encompasses remote miRNA signalling. In addition, we review the roles of miRNA tandems and describe the relevance of RNA target selection and competition to the behaviour of lesional macrophages. Elucidating miRNA-mediated regulatory mechanisms can aid the development of novel diagnostic and therapeutic strategies for atherosclerosis.
Collapse
|
78
|
Zhuang X, Herbert JMJ, Lodhia P, Bradford J, Turner AM, Newby PM, Thickett D, Naidu U, Blakey D, Barry S, Cross DAE, Bicknell R. Identification of novel vascular targets in lung cancer. Br J Cancer 2015; 112:485-94. [PMID: 25535734 PMCID: PMC4453649 DOI: 10.1038/bjc.2014.626] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/13/2014] [Accepted: 11/26/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Lung cancer remains the leading cause of cancer-related death, largely owing to the lack of effective treatments. A tumour vascular targeting strategy presents an attractive alternative; however, the molecular signature of the vasculature in lung cancer is poorly explored. This work aimed to identify novel tumour vascular targets in lung cancer. METHODS Enzymatic digestion of fresh tissue followed by endothelial capture with Ulex lectin-coated magnetic beads was used to isolate the endothelium from fresh tumour specimens of lung cancer patients. Endothelial isolates from the healthy and tumour lung tissue were subjected to whole human genome expression profiling using microarray technology. RESULTS Bioinformatics analysis identified tumour endothelial expression of angiogenic factors, matrix metalloproteases and cell-surface transmembrane proteins. Predicted novel tumour vascular targets were verified by RNA-seq, quantitative real-time PCR analysis and immunohistochemistry. Further detailed expression profiling of STEAP1 on 82 lung cancer patients confirmed STEAP1 as a novel target in the tumour vasculature. Functional analysis of STEAP1 using siRNA silencing implicates a role in endothelial cell migration and tube formation. CONCLUSIONS The identification of cell-surface tumour endothelial markers in lung is of interest in therapeutic antibody and vaccine development.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Non-Small-Cell Lung/blood supply
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Gene Expression Profiling
- Genetic Association Studies/methods
- Humans
- Lung/blood supply
- Lung/metabolism
- Lung/pathology
- Lung Neoplasms/blood supply
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Male
- Microarray Analysis
- Middle Aged
- Molecular Targeted Therapy
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/genetics
- Real-Time Polymerase Chain Reaction
- Sequence Analysis, RNA
Collapse
Affiliation(s)
- X Zhuang
- School of Immunity and Infection,
Institute for Biomedical Research, College of Medical and Dental Sciences,
University of Birmingham, Edgbaston, Birmingham
B15 2TT, UK
- School of Cancer Sciences, College of
Medical and Dental Sciences, University of Birmingham,
Edgbaston, Birmingham
B15 2TT, UK
| | - J M J Herbert
- School of Immunity and Infection,
Institute for Biomedical Research, College of Medical and Dental Sciences,
University of Birmingham, Edgbaston, Birmingham
B15 2TT, UK
- Technology Hub Sequencing and
Bioinformatics, College of Medical and Dental Sciences,
Birmingham
B15, UK
| | - P Lodhia
- School of Immunity and Infection,
Institute for Biomedical Research, College of Medical and Dental Sciences,
University of Birmingham, Edgbaston, Birmingham
B15 2TT, UK
| | - J Bradford
- AstraZeneca, Mereside,
Alderley Park, Macclesfield, Cheshire
SK10 4TG, UK
| | - A M Turner
- School of Clinical and Experimental
Medicine, University of Birmingham, QEHB Research Laboratories,
Mindelsohn Way, Birmingham
B15 2WB, UK
- Birmingham Heartlands Hospital,
Bordesley Green, Birmingham
B9 5SS, UK
| | - P M Newby
- School of Immunity and Infection,
Institute for Biomedical Research, College of Medical and Dental Sciences,
University of Birmingham, Edgbaston, Birmingham
B15 2TT, UK
| | - D Thickett
- School of Clinical and Experimental
Medicine, University of Birmingham, QEHB Research Laboratories,
Mindelsohn Way, Birmingham
B15 2WB, UK
| | - U Naidu
- School of Clinical and Experimental
Medicine, University of Birmingham, QEHB Research Laboratories,
Mindelsohn Way, Birmingham
B15 2WB, UK
- Birmingham Heartlands Hospital,
Bordesley Green, Birmingham
B9 5SS, UK
| | - D Blakey
- AstraZeneca, Mereside,
Alderley Park, Macclesfield, Cheshire
SK10 4TG, UK
| | - S Barry
- AstraZeneca, Mereside,
Alderley Park, Macclesfield, Cheshire
SK10 4TG, UK
| | - D A E Cross
- AstraZeneca, Mereside,
Alderley Park, Macclesfield, Cheshire
SK10 4TG, UK
| | - R Bicknell
- School of Immunity and Infection,
Institute for Biomedical Research, College of Medical and Dental Sciences,
University of Birmingham, Edgbaston, Birmingham
B15 2TT, UK
- School of Cancer Sciences, College of
Medical and Dental Sciences, University of Birmingham,
Edgbaston, Birmingham
B15 2TT, UK
| |
Collapse
|
79
|
Rödel F, Frey B, Multhoff G, Gaipl U. Contribution of the immune system to bystander and non-targeted effects of ionizing radiation. Cancer Lett 2015; 356:105-13. [DOI: 10.1016/j.canlet.2013.09.015] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 08/13/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022]
|
80
|
Hamburg NM, Leeper NJ. Therapeutic Potential of Modulating MicroRNA in Peripheral Artery Disease. Curr Vasc Pharmacol 2015; 13:316-23. [PMID: 23713861 PMCID: PMC4886469 DOI: 10.2174/15701611113119990014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 02/12/2013] [Accepted: 02/13/2013] [Indexed: 12/16/2022]
Abstract
Peripheral artery disease (PAD) produces significant disability attributable to lower extremity ischemia. Limited treatment modalities exist to ameliorate clinical symptoms in patients with PAD. Growing evidence links microRNAs to key processes that govern disease expression in PAD including angiogenesis, endothelial function, inflammation, vascular regeneration, vascular smooth muscle cell function, restenosis, and mitochondrial function. MicroRNAs have been identified in circulation and may serve as novel biomarkers in PAD. This article reviews the potential contribution of microRNA to key pathways of disease development in PAD that may lead to microRNA-based diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Naomi M Hamburg
- Section of Cardiology, Boston Medical Center, 88 East Newton St., Boston, MA, 02118.
| | | |
Collapse
|
81
|
Noncoding RNAs in diabetes vascular complications. J Mol Cell Cardiol 2014; 89:42-50. [PMID: 25536178 DOI: 10.1016/j.yjmcc.2014.12.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 11/18/2014] [Accepted: 12/05/2014] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus is the most common metabolic disorder and is recognised as a dominant health threat of our time. Diabetes induces a widespread damage of the macro- and microvasculature in different organs and tissues and disrupts the endogenous vascular repair mechanisms, thus causing diffuse and severe complications. Moreover, diabetic patients respond poorly to surgical interventions aiming to "revascularise" (i.e., to restore blood flow supply) the ischemic myocardium or lower limbs. The molecular causes underpinning diabetes vascular complications are still underappreciated and druggable molecular targets for therapeutic interventions have not yet clearly emerged. Moreover, diabetes itself and diabetes complications are often silent killers, requiring new prognostic, diagnostic and predictive biomarkers for use in the clinical practice. Noncoding RNA (ncRNAs) are emerging as new fundamental regulators of gene expression. The small microRNAs (miRNAs, miRs) have opened the field capturing the attention of basic and clinical scientists for their potential to become new therapeutic targets and clinical biomarkers. More recently, long ncRNAs (lncRNAs) have started to be actively investigated, leading to first exciting reports, which further suggest their important and yet largely unexplored contribution to vascular physiology and disease. This review introduces the different ncRNA types and focuses at the ncRNA roles in diabetes vascular complications. Furthermore, we discuss the potential value of ncRNAs as clinical biomarkers, and we examine the possibilities for therapeutic intervention targeting ncRNs in diabetes. This article is part of a Special Issue titled: Non-coding RNAs.
Collapse
|
82
|
Kane NM, Thrasher AJ, Angelini GD, Emanueli C. Concise review: MicroRNAs as modulators of stem cells and angiogenesis. Stem Cells 2014; 32:1059-66. [PMID: 24449004 DOI: 10.1002/stem.1629] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 12/08/2013] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRs) are highly conserved, short noncoding RNA molecules that negatively regulate messenger RNA (mRNA) stability and/or translational efficiency. Since a given miR can control the expression of many mRNAs, their importance in governing gene expression in specific cell types including vascular cells and their progenitor cells has become increasingly clear. Understanding how the expression of miRs themselves is regulated and how miRs exert their influence on post-transcriptional gene control provides novel opportunities to dissect gene regulatory networks in clinically relevant cell types. A multitude of miRs have been identified with key roles in vascular development, homeostasis, function, disease, and regeneration. In this review, we will describe the impact of miRs on angiogenesis and their capacity to modulate the behavior of stem and progenitor cells which may be utilitarian for promoting vascular growth in ischemic tissue. Moreover, we summarize these strategies available for modulating miR expression and function and future therapeutic applications.
Collapse
Affiliation(s)
- Nicole M Kane
- Molecular Immunology Unit, Institute of Child Health, University College of London, London, United Kingdom
| | | | | | | |
Collapse
|
83
|
Abstract
miRNAs are highly conserved non-coding RNA molecules that negatively control gene expression by binding to target mRNAs promoting their degradation. A multitude of miRNAs have been reported to be involved in angiogenesis and vascular remodelling. In the present review, we aim to describe the effect of miRNAs in post-ischaemic repair. First, we describe the miRNAs reported in ischaemic diseases and in angiogenesis. Then we examine their capacity to modulate the behaviour of stem and progenitor cells which could be utilized for vascular repair. And finally we discuss the potential of miRNAs as new clinical biomarkers and therapeutic targets.
Collapse
|
84
|
Romay MC, Che N, Becker SN, Pouldar D, Hagopian R, Xiao X, Lusis AJ, Berliner JA, Civelek M. Regulation of NF-κB signaling by oxidized glycerophospholipid and IL-1β induced miRs-21-3p and -27a-5p in human aortic endothelial cells. J Lipid Res 2014; 56:38-50. [PMID: 25327529 DOI: 10.1194/jlr.m052670] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Exposure of endothelial cells (ECs) to agents such as oxidized glycerophospholipids (oxGPs) and cytokines, known to accumulate in atherosclerotic lesions, perturbs the expression of hundreds of genes in ECs involved in inflammatory and other biological processes. We hypothesized that microRNAs (miRNAs) are involved in regulating the inflammatory response in human aortic endothelial cells (HAECs) in response to oxGPs and interleukin 1β (IL-1β). Using next-generation sequencing and RT-quantitative PCR, we characterized the profile of expressed miRNAs in HAECs pre- and postexposure to oxGPs. Using this data, we identified miR-21-3p and miR-27a-5p to be induced 3- to 4-fold in response to oxGP and IL-1β treatment compared with control treatment. Transient overexpression of miR-21-3p and miR-27a-5p resulted in the downregulation of 1,253 genes with 922 genes overlapping between the two miRNAs. Gene Ontology functional enrichment analysis predicted that the two miRNAs were involved in the regulation of nuclear factor κB (NF-κB) signaling. Overexpression of these two miRNAs leads to changes in p65 nuclear translocation. Using 3' untranslated region luciferase assay, we identified 20 genes within the NF-κB signaling cascade as putative targets of miRs-21-3p and -27a-5p, implicating these two miRNAs as modulators of NF-κB signaling in ECs.
Collapse
Affiliation(s)
- Milagros C Romay
- Departments of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Nam Che
- Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Scott N Becker
- Departments of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Delila Pouldar
- Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Raffi Hagopian
- Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Xinshu Xiao
- Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095
| | - Aldons J Lusis
- Departments of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095 Medicine, University of California, Los Angeles, Los Angeles, CA 90095 Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Judith A Berliner
- Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Mete Civelek
- Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
85
|
Abstract
SIGNIFICANCE Hypoxia is a hallmark of the tumor microenvironment and represents a major source of failure in cancer therapy. RECENT ADVANCES Recent work has generated extensive evidence that microRNAs (miRNAs) are significant components of the adaptive response to low oxygen in tumors. Induction of specific miRNAs, collectively termed hypoxamiRs, has become an accepted feature of the hypoxic response in normal and transformed cells. CRITICAL ISSUES Overexpression of miR-210, the prototypical hypoxamiR, is detected in most solid tumors, and it has been linked to adverse prognosis in many tumor types. Several miR-210 target genes, including iron-sulfur (Fe-S) cluster scaffold protein (ISCU) and glycerol-3-phosphate dehydrogenase 1-like (GPD1L), have been correlated with prognosis in an inverse fashion to miR-210, suggesting that their down- regulation by miR-210 occurs in vivo and contributes to tumor growth. Additional miRNAs are modulated by decreased oxygen tension in a more tissue-specific fashion, adding another level of complexity over the classic hypoxia-regulated gene network. FUTURE DIRECTIONS From a biological standpoint, hypoxamiRs are emerging modifiers of cancer cell response to the adaptive challenges of the microenvironment. From a clinical perspective, assessing the status of these miRNAs may contribute to a detailed understanding of hypoxia-induced mechanisms of resistance and/or to the fine-tuning of future hypoxia-modifying therapies.
Collapse
Affiliation(s)
- Harriet E Gee
- 1 Department of Radiation Oncology, Sydney Cancer Centre, Royal Prince Alfred Hospital , Camperdown, Australia
| | | | | | | |
Collapse
|
86
|
Greco S, Gaetano C, Martelli F. HypoxamiR regulation and function in ischemic cardiovascular diseases. Antioxid Redox Signal 2014; 21:1202-19. [PMID: 24053126 PMCID: PMC4142792 DOI: 10.1089/ars.2013.5403] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE MicroRNAs (miRNAs) are deregulated and play a causal role in numerous cardiovascular diseases, including myocardial infarction, coronary artery disease, hypertension, heart failure, stroke, peripheral artery disease, kidney ischemia-reperfusion. RECENT ADVANCES One crucial component of ischemic cardiovascular diseases is represented by hypoxia. Indeed, hypoxia is a powerful stimulus regulating the expression of a specific subset of miRNAs, named hypoxia-induced miRNAs (hypoxamiR). These miRNAs are fundamental regulators of the cell responses to decreased oxygen tension. Certain hypoxamiRs seem to have a particularly pervasive role, such as miR-210 that is virtually induced in all ischemic diseases tested so far. However, its specific function may change according to the physiopathological context. CRITICAL ISSUES The discovery of HypoxamiR dates back 6 years. Thus, despite a rapid growth in knowledge and attention, a deeper insight of the molecular mechanisms underpinning hypoxamiR regulation and function is needed. FUTURE DIRECTIONS An extended understanding of the function of hypoxamiR in gene regulatory networks associated with cardiovascular diseases will allow the identification of novel molecular mechanisms of disease and indicate the development of innovative therapeutic approaches.
Collapse
Affiliation(s)
- Simona Greco
- 1 Molecular Cardiology Laboratory , IRCCS-Policlinico San Donato, Milan, Italy
| | | | | |
Collapse
|
87
|
Radovich M, Ragoussis J. Methods of quantifying microRNAs for hypoxia research: classic and next generation. Antioxid Redox Signal 2014; 21:1239-48. [PMID: 24328936 DOI: 10.1089/ars.2013.5716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE Recent evidence is uncovering the ever-increasing importance of microribonucleic acids (miRNAs) in the hypoxia response. In order to investigate the important roles that these small RNAs play, methods of quantification whether using classic single-gene methods or genome-wide technologies are necessary to obtain a global picture of the differential expression of miRNAs in hypoxia and their interplay with protein coding genes. RECENT ADVANCES Building on the groundwork of classic quantitative polymerase chain reaction (qPCR) and microarrays, the advent of next-generation sequencing technology has revolutionized how small RNAs can be detected and quantified on a genome-wide scale and without a priori knowledge of the small RNA sequence. This method delivers accurate and comprehensive data on the expression and sequence of all expressed small RNAs, and the data can be further combined with other sequencing modalities to better understand miRNAs via integrated genomic analyses. CRITICAL ISSUES Advancing technology has increased the need for better methods of sample and library preparation and for bioinformatics tools. Speed, cost, sample input, and analysis expertise remain the mainstay critical issues of small RNA sequencing. FUTURE DIRECTIONS Future hypoxia research will benefit from the application of genome-wide sequencing technologies. Analyses that combine genomic, transcriptomic, chromosome conformation, DNA/RNA-protein binding, and proteomics will help greatly advance hypoxia miRNA research.
Collapse
Affiliation(s)
- Milan Radovich
- 1 Department of Surgery, Indiana University School of Medicine , Indianapolis, Indiana
| | | |
Collapse
|
88
|
Bandara V, Michael MZ, Gleadle JM. Hypoxia represses microRNA biogenesis proteins in breast cancer cells. BMC Cancer 2014; 14:533. [PMID: 25052766 PMCID: PMC4223767 DOI: 10.1186/1471-2407-14-533] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 07/16/2014] [Indexed: 02/08/2023] Open
Abstract
Background Cancers are commonly characterised by hypoxia and also by global reductions in the levels of mature microRNAs. We have examined the hypothesis that hypoxia might mediate this reduction through repressive effects on microRNA biogenesis proteins. Methods Breast cancer cell lines were exposed to hypoxia and manipulations of hypoxia inducible factor (HIF) and HIF hydroxylase activity. The effects of hypoxia on the mRNA and protein levels of enzymes involved in microRNA biogenesis (Dicer, Drosha, TARPB2, DCGR8, XPO5) was determined by RT PCR and immunoblotting. The effect of hypoxia on microRNAs was determined with microarray studies, RT PCR and reporter assays. Results In breast cancer lines there was significant reduction of Dicer mRNA and protein levels in cells exposed to hypoxia. This effect was independent of HIF but dependent on the HIF hydroxylase PHD2 and was partly mediated by feedback effects via microRNAs. Furthermore, several other proteins with critical roles in microRNA biogenesis (Drosha, TARBP2 and DCGR8) also showed significant and co-ordinated repression under hypoxic conditions. Despite these substantial alterations no, or modest, changes were observed in mature microRNA production. Conclusion These observations provide further and important interfaces between oxygen availability and gene expression and a potential mechanistic explanation for the reduced levels of microRNAs observed in some cancers. They provide further support for the existence of feedback mechanisms in the regulation of the microRNA biogenesis pathway and the relative stability of microRNAs.
Collapse
Affiliation(s)
| | | | - Jonathan M Gleadle
- Renal Department, Flinders Medical Centre, Flinders University School of Medicine, Bedford Park, Adelaide, South Australia 5042, Australia.
| |
Collapse
|
89
|
Fu X, Huang X, Li P, Chen W, Xia M. 7-Ketocholesterol inhibits isocitrate dehydrogenase 2 expression and impairs endothelial function via microRNA-144. Free Radic Biol Med 2014; 71:1-15. [PMID: 24642088 DOI: 10.1016/j.freeradbiomed.2014.03.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/15/2014] [Accepted: 03/10/2014] [Indexed: 11/30/2022]
Abstract
Oxysterol is associated with the induction of endothelial oxidative stress and impaired endothelial function. Mitochondria play a central role in oxidative energy metabolism and the maintenance of proper redox status. The purpose of this study was to determine the effects and mechanisms of 7-ketocholesterol (7-KC) on isocitrate dehydrogenase 2 (IDH2) and its impact on endothelial function in both human aortic endothelial cells (HAECs) and C57BL/6J mice. HAECs treated with 7-KC showed significant reductions of IDH2 mRNA and protein levels and enzyme activity, leading to decreased NADPH concentration and an increased ratio of reduced-to-oxidized glutathione in the mitochondria. 7-KC induced the expression of a specific microRNA, miR-144, which in turn targets and downregulates IDH2. In silico analysis predicted that miR-144 could bind to the 3'-untranslated region of IDH2 mRNA. Overexpression of miR-144 decreased the expression of IDH2 and the levels of NADPH. A complementary finding is that a miR-144 inhibitor increased the mRNA and protein expression levels of IDH2. Furthermore, miR-144 level was elevated in HAECs in response to 7-KC. Anti-Ago1/2 immunoprecipitation coupled with a real-time polymerase chain reaction assay revealed that 7-KC increased the functional targeting of miR-144/IDH2 mRNA in HAECs. Infusion of 7-KC in vivo decreased vascular IDH2 expression and impaired vascular reactivity via miR-144. 7-KC controls miR-144 expression, which in turn decreases IDH2 expression and attenuates NO bioavailability to impair endothelial homeostasis. The newly identified 7-KC-miR-144-IDH2 pathway may contribute to atherosclerosis progression and provides new insight into 7-KC function and microRNA biology in cardiovascular disease.
Collapse
Affiliation(s)
- Xiaodong Fu
- Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xiuwei Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China; Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China
| | - Ping Li
- Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Weiyu Chen
- Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China; Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China.
| |
Collapse
|
90
|
Kent OA, McCall MN, Cornish TC, Halushka MK. Lessons from miR-143/145: the importance of cell-type localization of miRNAs. Nucleic Acids Res 2014; 42:7528-38. [PMID: 24875473 PMCID: PMC4081080 DOI: 10.1093/nar/gku461] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
miR-143 and miR-145 are co-expressed microRNAs (miRNAs) that have been extensively studied as potential tumor suppressors. These miRNAs are highly expressed in the colon and are consistently reported as being downregulated in colorectal and other cancers. Through regulation of multiple targets, they elicit potent effects on cancer cell growth and tumorigenesis. Importantly, a recent discovery demonstrates that miR-143 and miR-145 are not expressed in colonic epithelial cells; rather, these two miRNAs are highly expressed in mesenchymal cells such as fibroblasts and smooth muscle cells. The expression patterns of miR-143 and miR-145 and other miRNAs were initially determined from tissue level data without consideration that multiple different cell types, each with their own unique miRNA expression patterns, make up each tissue. Herein, we discuss the early reports on the identification of dysregulated miR-143 and miR-145 expression in colorectal cancer and how lack of consideration of cellular composition of normal tissue led to the misconception that these miRNAs are downregulated in cancer. We evaluate mechanistic data from miR-143/145 studies in context of their cell type-restricted expression pattern and the potential of these miRNAs to be considered tumor suppressors. Further, we examine other examples of miRNAs being investigated in inappropriate cell types modulating pathways in a non-biological fashion. Our review highlights the importance of determining the cellular expression pattern of each miRNA, so that downstream studies are conducted in the appropriate cell type.
Collapse
Affiliation(s)
- Oliver A Kent
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Room 8-703, Toronto Medical Discovery Tower, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Matthew N McCall
- University of Rochester, Department of Biostatistics, Rochester, NY 14642, USA
| | - Toby C Cornish
- Johns Hopkins University, Department of Pathology, Baltimore, MD 21205, USA
| | - Marc K Halushka
- Johns Hopkins University, Department of Pathology, Baltimore, MD 21205, USA
| |
Collapse
|
91
|
Chevillet JR, Lee I, Briggs HA, He Y, Wang K. Issues and prospects of microRNA-based biomarkers in blood and other body fluids. Molecules 2014; 19:6080-105. [PMID: 24830712 PMCID: PMC6271291 DOI: 10.3390/molecules19056080] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/07/2014] [Accepted: 05/12/2014] [Indexed: 12/21/2022] Open
Abstract
Cell-free circulating microRNAs (miRNAs) in the blood are good diagnostic biomarker candidates for various physiopathological conditions, including cancer, neurodegeneration, diabetes and other diseases. Since their discovery in 2008 as blood biomarkers, the field has expanded rapidly with a number of important findings. Despite the initial optimistic views of their potential for clinical application, there are currently no circulating miRNA-based diagnostics in use. In this article, we review the status of circulating miRNAs, examine different analytical approaches, and address some of the challenges and opportunities.
Collapse
Affiliation(s)
| | - Inyoul Lee
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Hilary A Briggs
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, NY 10065, USA
| | - Yuqing He
- Institute of Medical Systems Biology, Guangdong Medical College, Dongguan, Guangdong 523808, China.
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA 98109, USA.
| |
Collapse
|
92
|
Poissonnier L, Villain G, Soncin F, Mattot V. miR126-5p repression of ALCAM and SetD5 in endothelial cells regulates leucocyte adhesion and transmigration. Cardiovasc Res 2014; 102:436-47. [PMID: 24562769 DOI: 10.1093/cvr/cvu040] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AIMS miR126-5p is processed from the miR126-3p/-5p duplex, which is expressed in endothelial cells and gives rise to the guide strand miR126-3p and the passenger strand miR126-5p. miR126-3p has prominent roles in vascular development and diseases, whereas the expression and physiological functions of miR126-5p are unknown. The purpose of this study was to evaluate the expression and role of miR126-5p in blood vessel endothelial cells. METHODS AND RESULTS miR126-5p is mostly expressed in blood vessel endothelial cells in vivo and in vitro. Gain- and loss-of-function approaches revealed that miR126-5p promotes leucocyte adhesion and represses leucocyte transendothelial migration. Two distinct target genes of miR126-5p in endothelial cells were identified: the activated leucocyte cell adhesion molecule (ALCAM) gene which codes for an adhesion molecule involved in leucocyte transendothelial migration and SetD5, a gene with previously unknown functions. Using either a blocking antibody or target protectors which specifically disrupt the miRNA/mRNA target pairing, we showed that miR126-5p promotes leucocyte adhesion by controlling the expression of SetD5 and represses transendothelial migration via the regulation of ALCAM. miR126-5p controls ALCAM and SetD5 expression in vivo in separate tissues and regulates leucocyte infiltration into inflamed lungs by repressing ALCAM expression. CONCLUSION miR126-5p is a functional, endothelial-enriched microRNA that participates in the control of leucocyte trafficking by regulating the expression of ALCAM and SetD5.
Collapse
Affiliation(s)
- Loïc Poissonnier
- CNRS, UMR8161, Institut de Biologie de Lille, Institut Pasteur de Lille, Université Lille-Nord de France, 1 rue du Pr Calmette, Lille Cedex 59021, France
| | - Gaëlle Villain
- CNRS, UMR8161, Institut de Biologie de Lille, Institut Pasteur de Lille, Université Lille-Nord de France, 1 rue du Pr Calmette, Lille Cedex 59021, France
| | - Fabrice Soncin
- CNRS, UMR8161, Institut de Biologie de Lille, Institut Pasteur de Lille, Université Lille-Nord de France, 1 rue du Pr Calmette, Lille Cedex 59021, France
| | - Virginie Mattot
- CNRS, UMR8161, Institut de Biologie de Lille, Institut Pasteur de Lille, Université Lille-Nord de France, 1 rue du Pr Calmette, Lille Cedex 59021, France
| |
Collapse
|
93
|
Camps C, Saini HK, Mole DR, Choudhry H, Reczko M, Guerra-Assunção JA, Tian YM, Buffa FM, Harris AL, Hatzigeorgiou AG, Enright AJ, Ragoussis J. Integrated analysis of microRNA and mRNA expression and association with HIF binding reveals the complexity of microRNA expression regulation under hypoxia. Mol Cancer 2014; 13:28. [PMID: 24517586 PMCID: PMC3928101 DOI: 10.1186/1476-4598-13-28] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 02/05/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND In mammalians, HIF is a master regulator of hypoxia gene expression through direct binding to DNA, while its role in microRNA expression regulation, critical in the hypoxia response, is not elucidated genome wide. Our aim is to investigate in depth the regulation of microRNA expression by hypoxia in the breast cancer cell line MCF-7, establish the relationship between microRNA expression and HIF binding sites, pri-miRNA transcription and microRNA processing gene expression. METHODS MCF-7 cells were incubated at 1% Oxygen for 16, 32 and 48 h. SiRNA against HIF-1α and HIF-2α were performed as previously published. MicroRNA and mRNA expression were assessed using microRNA microarrays, small RNA sequencing, gene expression microarrays and Real time PCR. The Kraken pipeline was applied for microRNA-seq analysis along with Bioconductor packages. Microarray data was analysed using Limma (Bioconductor), ChIP-seq data were analysed using Gene Set Enrichment Analysis and multiple testing correction applied in all analyses. RESULTS Hypoxia time course microRNA sequencing data analysis identified 41 microRNAs significantly up- and 28 down-regulated, including hsa-miR-4521, hsa-miR-145-3p and hsa-miR-222-5p reported in conjunction with hypoxia for the first time. Integration of HIF-1α and HIF-2α ChIP-seq data with expression data showed overall association between binding sites and microRNA up-regulation, with hsa-miR-210-3p and microRNAs of miR-27a/23a/24-2 and miR-30b/30d clusters as predominant examples. Moreover the expression of hsa-miR-27a-3p and hsa-miR-24-3p was found positively associated to a hypoxia gene signature in breast cancer. Gene expression analysis showed no full coordination between pri-miRNA and microRNA expression, pointing towards additional levels of regulation. Several transcripts involved in microRNA processing were found regulated by hypoxia, of which DICER (down-regulated) and AGO4 (up-regulated) were HIF dependent. DICER expression was found inversely correlated to hypoxia in breast cancer. CONCLUSIONS Integrated analysis of microRNA, mRNA and ChIP-seq data in a model cell line supports the hypothesis that microRNA expression under hypoxia is regulated at transcriptional and post-transcriptional level, with the presence of HIF binding sites at microRNA genomic loci associated with up-regulation. The identification of hypoxia and HIF regulated microRNAs relevant for breast cancer is important for our understanding of disease development and design of therapeutic interventions.
Collapse
Affiliation(s)
- Carme Camps
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Harpreet K Saini
- EMBL—European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - David R Mole
- Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Hani Choudhry
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Martin Reczko
- Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, 34 Fleming Street, Vari 16672, Greece
| | | | - Ya-Min Tian
- Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Francesca M Buffa
- Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Adrian L Harris
- Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Artemis G Hatzigeorgiou
- Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, 34 Fleming Street, Vari 16672, Greece
| | - Anton J Enright
- EMBL—European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Jiannis Ragoussis
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
- Institute of Molecular Oncology, Biomedical Sciences Research Center “Alexander Fleming”, 34 Fleming Street, Vari 16672, Greece
- Present Address: McGill University and Genome Quebec Innovation Centre, 740 DR Penfield Ave, Montreal H3A 0G1, Canada
| |
Collapse
|
94
|
Abstract
Recent discoveries of microRNAs (miRNAs) that control high-density lipoprotein abundance and function have expanded our knowledge of the mechanisms regulating this important lipoprotein subclass. miRNAs have been shown to regulate gene networks that control high-density lipoprotein biogenesis and uptake, as well as discrete steps in the reverse cholesterol transport pathway. Furthermore, high-density lipoprotein itself has been shown to transport miRNAs selectively in health and disease, offering new possibilities of how this lipoprotein may alter gene expression in distal target cells and tissues. Collectively, these discoveries offer new insights into the mechanisms governing high-density lipoprotein metabolism and function and open new avenues for the development of therapeutics for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Katey J Rayner
- From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada (K.J.R.); and Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine (K.J.M.)
| | | |
Collapse
|
95
|
Nie Z, Zhou F, Li D, Lv Z, Chen J, Liu Y, Shu J, Sheng Q, Yu W, Zhang W, Jiang C, Yao Y, Yao J, Jin Y, Zhang Y. RIP-seq of BmAgo2-associated small RNAs reveal various types of small non-coding RNAs in the silkworm, Bombyx mori. BMC Genomics 2013; 14:661. [PMID: 24074203 PMCID: PMC3849828 DOI: 10.1186/1471-2164-14-661] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 09/26/2013] [Indexed: 12/21/2022] Open
Abstract
Background Small non-coding RNAs (ncRNAs) are important regulators of gene expression in eukaryotes. Previously, only microRNAs (miRNAs) and piRNAs have been identified in the silkworm, Bombyx mori. Furthermore, only ncRNAs (50-500nt) of intermediate size have been systematically identified in the silkworm. Results Here, we performed a systematic identification and analysis of small RNAs (18-50nt) associated with the Bombyx mori argonaute2 (BmAgo2) protein. Using RIP-seq, we identified various types of small ncRNAs associated with BmAGO2. These ncRNAs showed a multimodal length distribution, with three peaks at ~20nt, ~27nt and ~33nt, which included tRNA-, transposable element (TE)-, rRNA-, snoRNA- and snRNA-derived small RNAs as well as miRNAs and piRNAs. The tRNA-derived fragments (tRFs) were found at an extremely high abundance and accounted for 69.90% of the BmAgo2-associated small RNAs. Northern blotting confirmed that many tRFs were expressed or up-regulated only in the BmNPV-infected cells, implying that the tRFs play a prominent role by binding to BmAgo2 during BmNPV infection. Additional evidence suggested that there are potential cleavage sites on the D, anti-codon and TψC loops of the tRNAs. TE-derived small RNAs and piRNAs also accounted for a significant proportion of the BmAgo2-associated small RNAs, suggesting that BmAgo2 could be involved in the maintenance of genome stability by suppressing the activities of transposons guided by these small RNAs. Finally, Northern blotting was also used to confirm the Bombyx 5.8 s rRNA-derived small RNAs, demonstrating that various novel small RNAs exist in the silkworm. Conclusions Using an RIP-seq method in combination with Northern blotting, we identified various types of small RNAs associated with the BmAgo2 protein, including tRNA-, TE-, rRNA-, snoRNA- and snRNA-derived small RNAs as well as miRNAs and piRNAs. Our findings provide new clues for future functional studies of the role of small RNAs in insect development and evolution.
Collapse
Affiliation(s)
- Zuoming Nie
- College of Life Sciences, Zhejiang Sci-Tech University, Hanghzou 310018, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Baran-Gale J, Fannin EE, Kurtz CL, Sethupathy P. Beta cell 5'-shifted isomiRs are candidate regulatory hubs in type 2 diabetes. PLoS One 2013; 8:e73240. [PMID: 24039891 PMCID: PMC3767796 DOI: 10.1371/journal.pone.0073240] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 07/18/2013] [Indexed: 01/02/2023] Open
Abstract
Next-generation deep sequencing of small RNAs has unveiled the complexity of the microRNA (miRNA) transcriptome, which is in large part due to the diversity of miRNA sequence variants (“isomiRs”). Changes to a miRNA’s seed sequence (nucleotides 2–8), including shifted start positions, can redirect targeting to a dramatically different set of RNAs and alter biological function. We performed deep sequencing of small RNA from mouse insulinoma (MIN6) cells (widely used as a surrogate for the study of pancreatic beta cells) and developed a bioinformatic analysis pipeline to profile isomiR diversity. Additionally, we applied the pipeline to recently published small RNA-seq data from primary human beta cells and whole islets and compared the miRNA profiles with that of MIN6. We found that: (1) the miRNA expression profile in MIN6 cells is highly correlated with those of primary human beta cells and whole islets; (2) miRNA loci can generate multiple highly expressed isomiRs with different 5′-start positions (5′-isomiRs); (3) isomiRs with shifted start positions (5′-shifted isomiRs) are highly expressed, and can be as abundant as their unshifted counterparts (5′-reference miRNAs). Finally, we identified 10 beta cell miRNA families as candidate regulatory hubs in a type 2 diabetes (T2D) gene network. The most significant candidate hub was miR-29, which we demonstrated regulates the mRNA levels of several genes critical to beta cell function and implicated in T2D. Three of the candidate miRNA hubs were novel 5′-shifted isomiRs: miR-375+1, miR-375-1 and miR-183-5p+1. We showed by in silico target prediction and in vitro transfection studies that both miR-375+1 and miR-375-1 are likely to target an overlapping, but distinct suite of beta cell genes compared to canonical miR-375. In summary, this study characterizes the isomiR profile in beta cells for the first time, and also highlights the potential functional relevance of 5′-shifted isomiRs to T2D.
Collapse
Affiliation(s)
- Jeanette Baran-Gale
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America ; Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | | | | | | |
Collapse
|
97
|
Nallamshetty S, Chan SY, Loscalzo J. Hypoxia: a master regulator of microRNA biogenesis and activity. Free Radic Biol Med 2013; 64:20-30. [PMID: 23712003 PMCID: PMC3762925 DOI: 10.1016/j.freeradbiomed.2013.05.022] [Citation(s) in RCA: 223] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 05/01/2013] [Accepted: 05/16/2013] [Indexed: 01/08/2023]
Abstract
Hypoxia, or low oxygen tension, is a unique environmental stress that induces global changes in a complex regulatory network of transcription factors and signaling proteins to coordinate cellular adaptations in metabolism, proliferation, DNA repair, and apoptosis. Several lines of evidence now establish microRNAs (miRNAs), which are short noncoding RNAs that regulate gene expression through posttranscriptional mechanisms, as key elements in this response to hypoxia. Oxygen deprivation induces a distinct shift in the expression of a specific group of miRNAs, termed hypoxamirs, and emerging evidence indicates that hypoxia regulates several facets of hypoxamir transcription, maturation, and function. Transcription factors such as hypoxia-inducible factor are upregulated under conditions of low oxygen availability and directly activate the transcription of a subset of hypoxamirs. Conversely, hypoxia selectively represses other hypoxamirs through less well characterized mechanisms. In addition, oxygen deprivation has been directly implicated in epigenetic modifications such as DNA demethylation that control specific miRNA transcription. Finally, hypoxia also modulates the activity of key proteins that control posttranscriptional events in the maturation and activity of miRNAs. Collectively, these findings establish hypoxia as an important proximal regulator of miRNA biogenesis and function. It will be important for future studies to address the relative contributions of transcriptional and posttranscriptional events in the regulation of specific hypoxamirs and how such miRNAs are coordinated in order to integrate into the complex hierarchical regulatory network induced by hypoxia.
Collapse
Affiliation(s)
- Shriram Nallamshetty
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Stephen Y. Chan
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
98
|
Derecka K, Blythe MJ, Malla S, Genereux DP, Guffanti A, Pavan P, Moles A, Snart C, Ryder T, Ortori CA, Barrett DA, Schuster E, Stöger R. Transient exposure to low levels of insecticide affects metabolic networks of honeybee larvae. PLoS One 2013; 8:e68191. [PMID: 23844170 PMCID: PMC3699529 DOI: 10.1371/journal.pone.0068191] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/27/2013] [Indexed: 01/21/2023] Open
Abstract
The survival of a species depends on its capacity to adjust to changing environmental conditions, and new stressors. Such new, anthropogenic stressors include the neonicotinoid class of crop-protecting agents, which have been implicated in the population declines of pollinating insects, including honeybees (Apis mellifera). The low-dose effects of these compounds on larval development and physiological responses have remained largely unknown. Over a period of 15 days, we provided syrup tainted with low levels (2 µg/L(-1)) of the neonicotinoid insecticide imidacloprid to beehives located in the field. We measured transcript levels by RNA sequencing and established lipid profiles using liquid chromatography coupled with mass spectrometry from worker-bee larvae of imidacloprid-exposed (IE) and unexposed, control (C) hives. Within a catalogue of 300 differentially expressed transcripts in larvae from IE hives, we detect significant enrichment of genes functioning in lipid-carbohydrate-mitochondrial metabolic networks. Myc-involved transcriptional response to exposure of this neonicotinoid is indicated by overrepresentation of E-box elements in the promoter regions of genes with altered expression. RNA levels for a cluster of genes encoding detoxifying P450 enzymes are elevated, with coordinated downregulation of genes in glycolytic and sugar-metabolising pathways. Expression of the environmentally responsive Hsp90 gene is also reduced, suggesting diminished buffering and stability of the developmental program. The multifaceted, physiological response described here may be of importance to our general understanding of pollinator health. Muscles, for instance, work at high glycolytic rates and flight performance could be impacted should low levels of this evolutionarily novel stressor likewise induce downregulation of energy metabolising genes in adult pollinators.
Collapse
Affiliation(s)
- Kamila Derecka
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire, United Kingdom
| | - Martin J. Blythe
- Deep Seq, Centre for Genetics and Genomics, University of Nottingham, Nottingham, United Kingdom
| | - Sunir Malla
- Deep Seq, Centre for Genetics and Genomics, University of Nottingham, Nottingham, United Kingdom
| | - Diane P. Genereux
- Biology Department, Westfield State University, Westfield, Massachusetts, United States of America
| | | | | | | | - Charles Snart
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire, United Kingdom
| | | | - Catharine A. Ortori
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - David A. Barrett
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Eugene Schuster
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Reinhard Stöger
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire, United Kingdom
| |
Collapse
|
99
|
Martin FA, Murphy RP, Cummins PM. Thrombomodulin and the vascular endothelium: insights into functional, regulatory, and therapeutic aspects. Am J Physiol Heart Circ Physiol 2013; 304:H1585-97. [PMID: 23604713 PMCID: PMC7212260 DOI: 10.1152/ajpheart.00096.2013] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Thrombomodulin (TM) is a 557-amino acid protein with a broad cell and tissue distribution consistent with its wide-ranging physiological roles. When expressed on the lumenal surface of vascular endothelial cells in both large vessels and capillaries, its primary function is to mediate endothelial thromboresistance. The complete integral membrane-bound protein form displays five distinct functional domains, although shorter soluble (functional) variants comprising the extracellular domains have also been reported in fluids such as serum and urine. TM-mediated binding of thrombin is known to enhance the specificity of the latter serine protease toward both protein C and thrombin activatable fibrinolysis inhibitor (TAFI), increasing their proteolytic activation rate by almost three orders of magnitude with concomitant anticoagulant, antifibrinolytic, and anti-inflammatory benefits to the vascular wall. Recent years have seen an abundance of research into the cellular mechanisms governing endothelial TM production, processing, and regulation (including flow-mediated mechanoregulation)--from transcriptional and posttranscriptional (miRNA) regulation of TM gene expression, to posttranslational processing and release of the expressed protein--facilitating greater exploitation of its therapeutic potential. The goal of the present paper is to comprehensively review the endothelial/TM system from these regulatory perspectives and draw some fresh conclusions. This paper will conclude with a timely examination of the current status of TM's growing therapeutic appeal, from novel strategies to improve the clinical efficacy of recombinant TM analogs for resolution of vascular disorders such as disseminated intravascular coagulation (DIC), to an examination of the complex pleiotropic relationship between statin treatment and TM expression.
Collapse
Affiliation(s)
- Fiona A Martin
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | | |
Collapse
|
100
|
McGahon MK, Yarham JM, Daly A, Guduric-Fuchs J, Ferguson LJ, Simpson DA, Collins A. Distinctive profile of IsomiR expression and novel microRNAs in rat heart left ventricle. PLoS One 2013; 8:e65809. [PMID: 23799049 PMCID: PMC3683050 DOI: 10.1371/journal.pone.0065809] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/03/2013] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) are single-stranded non-coding RNAs that negatively regulate target gene expression through mRNA cleavage or translational repression. There is mounting evidence that they play critical roles in heart disease. The expression of known miRNAs in the heart has been studied at length by microarray and quantitative PCR but it is becoming evident that microRNA isoforms (isomiRs) are potentially physiologically important. It is well known that left ventricular (patho)physiology is influenced by transmural heterogeneity of cardiomyocyte phenotype, and this likely reflects underlying heterogeneity of gene expression. Given the significant role of miRNAs in regulating gene expression, knowledge of how the miRNA profile varies across the ventricular wall will be crucial to better understand the mechanisms governing transmural physiological heterogeneity. To determinine miRNA/isomiR expression profiles in the rat heart we investigated tissue from different locations across the left ventricular wall using deep sequencing. We detected significant quantities of 145 known rat miRNAs and 68 potential novel orthologs of known miRNAs, in mature, mature* and isomiR formation. Many isomiRs were detected at a higher frequency than their canonical sequence in miRBase and have different predicted targets. The most common miR-133a isomiR was more effective at targeting a construct containing a sequence from the gelsolin gene than was canonical miR-133a, as determined by dual-fluorescence assay. We identified a novel rat miR-1 homolog from a second miR-1 gene; and a novel rat miRNA similar to miR-676. We also cloned and sequenced the rat miR-486 gene which is not in miRBase (v18). Signalling pathways predicted to be targeted by the most highly detected miRNAs include Ubiquitin-mediated Proteolysis, Mitogen-Activated Protein Kinase, Regulation of Actin Cytoskeleton, Wnt signalling, Calcium Signalling, Gap junctions and Arrhythmogenic Right Ventricular Cardiomyopathy. Most miRNAs are not expressed in a gradient across the ventricular wall, with exceptions including miR-10b, miR-21, miR-99b and miR-486.
Collapse
Affiliation(s)
- Mary K. McGahon
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Janet M. Yarham
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Aideen Daly
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Jasenka Guduric-Fuchs
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Lyndsey J. Ferguson
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - David A. Simpson
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Anthony Collins
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| |
Collapse
|