51
|
Hou F, Li D, Yu H, Kong Q. The mechanism and potential targets of class II HDACs in angiogenesis. J Cell Biochem 2017; 119:2999-3006. [PMID: 29091298 DOI: 10.1002/jcb.26476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/31/2017] [Indexed: 12/21/2022]
Abstract
Angiogenesis refers to the new blood vessels deriving from the existing blood vessels, and it is a complex regulatory process. Angiogenesis is associated with the normal development of the body and tumor growth and migration. The imbalance of histone deacetylase, as an epigenetic modification, could induce the production of diseases, such as cancer, metabolic diseases, etc., and it also plays an important role in angiogenesis. Many researches indicate that class II HDACs nuclear shuttle and its phosphorylation are necessary for the diseases and the protection of the collective itself. This paper will make a review for the relationship between II HDACs and angiogenesis under physiological and pathologic categories, looking forward to the disease treatment in the future.
Collapse
Affiliation(s)
- Fei Hou
- Department of Basic Medical College, Jining Medical University, Jining, Shandong, China.,College of Science, Qufu Normal University, Qufu, Shandong, China
| | - Dandan Li
- Department of Basic Medical College, Jining Medical University, Jining, Shandong, China.,College of Science, Qufu Normal University, Qufu, Shandong, China
| | - Honglian Yu
- Department of Basic Medical College, Jining Medical University, Jining, Shandong, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining, Shandong, China
| | - Qingsheng Kong
- Department of Basic Medical College, Jining Medical University, Jining, Shandong, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining, Shandong, China
| |
Collapse
|
52
|
Lv M, Chen H, Shao Y, Li C, Zhang W, Zhao X, Jin C, Xiong J. miR-92a regulates coelomocytes apoptosis in sea cucumber Apostichopus japonicus via targeting Aj14-3-3ζ in vivo. FISH & SHELLFISH IMMUNOLOGY 2017; 69:211-217. [PMID: 28860073 DOI: 10.1016/j.fsi.2017.08.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/21/2017] [Accepted: 08/27/2017] [Indexed: 06/07/2023]
Abstract
miR-92a, a well-documented oncogene, was previously found to be differentially expressed in diseased sea cucumber Apostichopus japonicus by high-throughput sequencing. In this study, we identified Aj14-3-3ζ as a novel target of miR-92a in this species and investigated their regulatory roles in vivo. The negative expression profiles between miR-92a and Aj14-3-3ζ protein were detected in both LPS-exposed primary coelomocytes and Vibrio splendidus-challenged sea cucumbers. Over-expression of miR-92a by injection of miR-92a agomir significantly depressed the mRNA and protein expression of Aj14-3-3ζ and promoted coelomocytes apoptosis with 5.04-fold increase in vivo, which was consistent with those from siRNA-mediated Aj14-3-3ζ knockdown assay. In contrast, miR-92a antagomir significantly elevated the mRNA and protein expression of Aj14-3-3ζ and decreased coelomocytes apoptosis. Taken together, our result confirmed that miR-92a is involved in apoptotic signaling pathway regulation perhaps via targeting Aj14-3-3ζ in sea cucumbers, which will enhance our understanding of miR-92a regulatory roles in sea cucumber pathogenesis.
Collapse
Affiliation(s)
- Miao Lv
- School of Marine Sciences, Ningbo University, PR China
| | - Huahui Chen
- School of Marine Sciences, Ningbo University, PR China
| | - Yina Shao
- School of Marine Sciences, Ningbo University, PR China
| | - Chenghua Li
- School of Marine Sciences, Ningbo University, PR China.
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, PR China
| | - Xuelin Zhao
- School of Marine Sciences, Ningbo University, PR China
| | - Chunhua Jin
- School of Marine Sciences, Ningbo University, PR China
| | - Jinbo Xiong
- School of Marine Sciences, Ningbo University, PR China
| |
Collapse
|
53
|
Abstract
Endothelial cells (ECs) are confirmed as important regulators of vascular integrity, particularly in relation to angiogenesis, wound repair post-injury, and during embryogenesis. Futher, miRNAs have been implicated in EC function and proliferation. Moreover, knockdown of these miRNAs resulted in altered expressions of several important regulators of endothelial biology and angiogenesis including vascular endothelial growth factor receptor 2, endothelial nitric oxide synthase and tubule formation capacity. Several miRNAs have been identified to play a role in the regulation of function, proliferation and growth of vascular ECs. These miRNAs may be important therapeutic targets in the treatment of a range of ischemic diseases, as well as in the regulation of angiogenesis during cancer and tumour progression. The present review discuss some of the important miRNAs having confirmed regulatory role in EC in connection espically with cardiovascular disease.
Collapse
Affiliation(s)
- Yong Cao
- Department of Cardiology, Xuzhou Hospital of Traditional Chinese Medicine, Xuzhou, Jiangsu 221009, P.R. China
| | - Pei-Ying Zhang
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
54
|
Ferronato S, Mombello A, Posenato I, Candiani P, Scuro A, Setacci C, Gomez-Lira M. Expression of Circulating miR-17-92 Cluster and HDAC9 Gene in Atherosclerotic Patients with Unstable and Stable Carotid Plaques. Genet Test Mol Biomarkers 2017; 21:402-405. [PMID: 28436693 DOI: 10.1089/gtmb.2016.0384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIMS The miR-17-92 cluster and the HDAC9 gene are involved in inflammatory, apoptotic, and angiogenic processes that are activated in the vulnerable carotid plaque. The aim of this research was to determine whether expression of one or more of the miRs of the miR-17-92 cluster and/or HDAC9 expression could represent biomarkers for patients with unstable atherosclerotic carotid plaques. MATERIALS AND METHODS Plasma levels of miRs and HDAC9 expression in peripheral blood were analyzed by real-time PCR in patients with histologically classified stable or unstable plaques. RESULTS No differences were observed between the two groups. DISCUSSION AND CONCLUSIONS Levels of the miR-17-92 cluster in plasma and HDAC9 gene expression in peripheral blood cannot be considered appropriate biomarkers to identify patients with unstable plaques at risk of rupture.
Collapse
Affiliation(s)
- Silvia Ferronato
- 1 Department of Neurological, Biomedical and Movement Sciences, University of Verona , Verona, Italy
| | - Aldo Mombello
- 2 Department of Diagnostics and Public Health, University of Verona , Verona, Italy
| | - Ilaria Posenato
- 2 Department of Diagnostics and Public Health, University of Verona , Verona, Italy
| | - Paola Candiani
- 3 Department of Surgery, University of Verona , Verona, Italy
| | - Alberto Scuro
- 3 Department of Surgery, University of Verona , Verona, Italy
| | - Carlo Setacci
- 4 Division of Vascular Surgery, University of Siena , Siena, Italy
| | - Macarena Gomez-Lira
- 1 Department of Neurological, Biomedical and Movement Sciences, University of Verona , Verona, Italy
| |
Collapse
|
55
|
Berndsen RH, Abdul UK, Weiss A, Zoetemelk M, te Winkel MT, Dyson PJ, Griffioen AW, Nowak-Sliwinska P. Epigenetic approach for angiostatic therapy: promising combinations for cancer treatment. Angiogenesis 2017; 20:245-267. [DOI: 10.1007/s10456-017-9551-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 03/10/2017] [Indexed: 12/15/2022]
|
56
|
Dimopoulos A, Sicko RJ, Kay DM, Rigler SL, Fan R, Romitti PA, Browne ML, Druschel CM, Caggana M, Brody LC, Mills JL. Copy number variants in a population-based investigation of Klippel-Trenaunay syndrome. Am J Med Genet A 2017; 173:352-359. [PMID: 27901321 PMCID: PMC6205266 DOI: 10.1002/ajmg.a.37868] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 06/16/2016] [Indexed: 01/19/2023]
Abstract
Klippel-Trenaunay syndrome (KTS) is a rare congenital vascular disorder that is thought to occur sporadically; however, reports of familial occurrence suggest a genetic component. We examined KTS cases to identify novel, potentially causal copy number variants (CNVs). We identified 17 KTS cases from all live-births occurring in New York (1998-2010). Extracted DNA was genotyped using Illumina microarrays and CNVs were called using PennCNV software. CNVs selected for follow-up had ≥10 single nucleotide polymorphisms (SNPs) and minimal overlap with in-house controls or controls from the Database of Genomic Variants. We identified 15 candidate CNVs in seven cases; among them a deletion in two cases within transcripts of HDAC9, a histone deacetylase essential for angiogenic sprouting of endothelial cells. One of them also had a duplication upstream of SALL3, a transcription factor essential for embryonic development that inhibits DNMT3A, a DNA methyltransferase responsible for embryonic de novo DNA methylation. Another case had a duplication spanning ING5, a histone acetylation regulator active during embryogenesis. We identified rare genetic variants related to chromatin modification which may have a key role in regulating vascular development during embryogenesis. Further investigation of their implications in the pathogenesis of KTS is warranted. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aggeliki Dimopoulos
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Robert J. Sicko
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York
| | - Denise M. Kay
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York
| | - Shannon L. Rigler
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Ruzong Fan
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Paul A. Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, Iowa
| | - Marilyn L. Browne
- Congenital Malformations Registry, New York State Department of Health, Albany, New York
- Department of Epidemiology and Biostatistics, University at Albany School of Public Health, Rensselaer, New York
| | - Charlotte M. Druschel
- Congenital Malformations Registry, New York State Department of Health, Albany, New York
- Department of Epidemiology and Biostatistics, University at Albany School of Public Health, Rensselaer, New York
| | - Michele Caggana
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York
| | - Lawrence C. Brody
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - James L. Mills
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
57
|
Hrgovic I, Doll M, Pinter A, Kaufmann R, Kippenberger S, Meissner M. Histone deacetylase inhibitors interfere with angiogenesis by decreasing endothelial VEGFR-2 protein half-life in part via a VE-cadherin-dependent mechanism. Exp Dermatol 2017; 26:194-201. [DOI: 10.1111/exd.13159] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Igor Hrgovic
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Monika Doll
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Andreas Pinter
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Roland Kaufmann
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Stefan Kippenberger
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Markus Meissner
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| |
Collapse
|
58
|
Tanimoto A, Takeuchi S, Arai S, Fukuda K, Yamada T, Roca X, Ong ST, Yano S. Histone Deacetylase 3 Inhibition Overcomes BIM Deletion Polymorphism-Mediated Osimertinib Resistance in EGFR-Mutant Lung Cancer. Clin Cancer Res 2016; 23:3139-3149. [PMID: 27986747 DOI: 10.1158/1078-0432.ccr-16-2271] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 12/02/2016] [Accepted: 12/09/2016] [Indexed: 11/16/2022]
Abstract
Purpose: The BIM deletion polymorphism is associated with apoptosis resistance to EGFR tyrosine kinase inhibitors (EGFR-TKI), such as gefitinib and erlotinib, in non-small cell lung cancer (NSCLC) harboring EGFR mutations. Here, we investigated whether the BIM deletion polymorphism contributes to resistance against osimertinib, a third-generation EGFR-TKI. In addition, we determined the efficacy of a histone deacetylase (HDAC) inhibitor, vorinostat, against this form of resistance and elucidated the underlying mechanism.Experimental Design: We used EGFR-mutated NSCLC cell lines, which were either heterozygous or homozygous for the BIM deletion polymorphism, to evaluate the effect of osimertinib in vitro and in vivo Protein expression was examined by Western blotting. Alternative splicing of BIM mRNA was analyzed by RT-PCR.Results:EGFR-mutated NSCLC cell lines with the BIM deletion polymorphism exhibited apoptosis resistance to osimertinib in a polymorphism dosage-dependent manner, and this resistance was overcome by combined use with vorinostat. Experiments with homozygous BIM deletion-positive cells revealed that vorinostat affected the alternative splicing of BIM mRNA in the deletion allele, increased the expression of active BIM protein, and thereby induced apoptosis in osimertinib-treated cells. These effects were mediated predominantly by HDAC3 inhibition. In xenograft models, combined use of vorinostat with osimertinib could regress tumors in EGFR-mutated NSCLC cells homozygous for the BIM deletion polymorphism. Moreover, this combination could induce apoptosis even when tumor cells acquired EGFR-T790M mutations.Conclusions: These findings indicate the importance of developing HDAC3-selective inhibitors, and their combined use with osimertinib, for treating EGFR-mutated lung cancers carrying the BIM deletion polymorphism. Clin Cancer Res; 23(12); 3139-49. ©2016 AACR.
Collapse
Affiliation(s)
- Azusa Tanimoto
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Shinji Takeuchi
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Sachiko Arai
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Koji Fukuda
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Tadaaki Yamada
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - S Tiong Ong
- Cancer & Stem Cell Biology Signature Research Programme, Duke-NUS Medical School, Singapore.,Department of Medical Oncology, National Cancer Centre Singapore, Singapore.,Department of Haematology, Singapore General Hospital, Singapore.,Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Seiji Yano
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
59
|
Craven KE, Gore J, Wilson JL, Korc M. Angiogenic gene signature in human pancreatic cancer correlates with TGF-beta and inflammatory transcriptomes. Oncotarget 2016; 7:323-41. [PMID: 26586478 PMCID: PMC4808001 DOI: 10.18632/oncotarget.6345] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/08/2015] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinomas (PDACs) are hypovascular, but overexpress pro-angiogenic factors and exhibit regions of microvasculature. Using RNA-seq data from The Cancer Genome Atlas (TCGA), we previously reported that ∼12% of PDACs have an angiogenesis gene signature with increased expression of multiple pro-angiogenic genes. By analyzing the recently expanded TCGA dataset, we now report that this signature is present in ∼35% of PDACs but that it is mostly distinct from an angiogenesis signature present in pancreatic neuroendocrine tumors (PNETs). These PDACs exhibit a transcriptome that reflects active TGF-β signaling, and up-regulation of several pro-inflammatory genes, and many members of JAK signaling pathways. Moreover, expression of SMAD4 and HDAC9 correlates with endothelial cell abundance in PDAC tissues. Concomitantly targeting the TGF-β type I receptor (TβRI) kinase with SB505124 and JAK1-2 with ruxolitinib suppresses JAK1 phosphorylation and blocks proliferative cross-talk between human pancreatic cancer cells (PCCs) and human endothelial cells (ECs), and these anti-proliferative effects were mimicked by JAK1 silencing in ECs. By contrast, either inhibitor alone does not suppress their enhanced proliferation in 3D co-cultures. These findings suggest that targeting both TGF-β and JAK1 signaling could be explored therapeutically in the 35% of PDAC patients whose cancers exhibit an angiogenesis gene signature.
Collapse
Affiliation(s)
- Kelly E Craven
- Departments of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jesse Gore
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,The Pancreatic Cancer Signature Center at Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Julie L Wilson
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Murray Korc
- Departments of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,The Pancreatic Cancer Signature Center at Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
60
|
Li Y, Seto E. HDACs and HDAC Inhibitors in Cancer Development and Therapy. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a026831. [PMID: 27599530 DOI: 10.1101/cshperspect.a026831] [Citation(s) in RCA: 862] [Impact Index Per Article: 95.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the last several decades, it has become clear that epigenetic abnormalities may be one of the hallmarks of cancer. Posttranslational modifications of histones, for example, may play a crucial role in cancer development and progression by modulating gene transcription, chromatin remodeling, and nuclear architecture. Histone acetylation, a well-studied posttranslational histone modification, is controlled by the opposing activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs). By removing acetyl groups, HDACs reverse chromatin acetylation and alter transcription of oncogenes and tumor suppressor genes. In addition, HDACs deacetylate numerous nonhistone cellular substrates that govern a wide array of biological processes including cancer initiation and progression. This review will discuss the role of HDACs in cancer and the therapeutic potential of HDAC inhibitors (HDACi) as emerging drugs in cancer treatment.
Collapse
Affiliation(s)
- Yixuan Li
- George Washington University Cancer Center, Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC 20037
| | - Edward Seto
- George Washington University Cancer Center, Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC 20037
| |
Collapse
|
61
|
Recchioni R, Marcheselli F, Antonicelli R, Lazzarini R, Mensà E, Testa R, Procopio AD, Olivieri F. Physical activity and progenitor cell-mediated endothelial repair in chronic heart failure: Is there a role for epigenetics? Mech Ageing Dev 2016; 159:71-80. [DOI: 10.1016/j.mad.2016.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/17/2016] [Accepted: 03/21/2016] [Indexed: 02/09/2023]
|
62
|
Kemmerer M, Wittig I, Richter F, Brüne B, Namgaladze D. AMPK activates LXRα and ABCA1 expression in human macrophages. Int J Biochem Cell Biol 2016; 78:1-9. [DOI: 10.1016/j.biocel.2016.06.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/10/2016] [Accepted: 06/21/2016] [Indexed: 11/16/2022]
|
63
|
Man HSJ, Yan MS, Lee JJ, Marsden PA. Epigenetic determinants of cardiovascular gene expression: vascular endothelium. Epigenomics 2016; 8:959-79. [PMID: 27381277 DOI: 10.2217/epi-2016-0012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The modern landscape of gene regulation involves interacting factors that ultimately lead to gene activation or repression. Epigenetic mechanisms provide a perspective of cellular phenotype as dynamically regulated and responsive to input. This perspective is supported by the generation of induced pluripotent stem cells from fully differentiated cell types. In vascular endothelial cells, evidence suggests that epigenetic mechanisms play a major role in the expression of endothelial cell-specific genes such as the endothelial nitric oxide synthase (NOS3/eNOS). These mechanisms are also important for eNOS expression in response to environmental stimuli such as hypoxia and shear stress. A newer paradigm in epigenetics, long noncoding RNAs offer a link between genetic variation, epigenetic regulation and disease. While the understanding of epigenetic mechanisms is early in its course, it is becoming clear that approaches to understanding the interaction of these factors and their inputs will be necessary to improve outcomes in cardiovascular disease.
Collapse
Affiliation(s)
- Hon-Sum Jeffrey Man
- Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Departments of Respirology & Critical Care, University Health Network & Mt Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Matthew S Yan
- Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - John Jy Lee
- Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Philip A Marsden
- Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Nephrology, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
64
|
Affiliation(s)
- Jan Fiedler
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integrated Research and Treatment Center Transplantation (J.F., T.T.), and Excellence Cluster REBIRTH (J.F., T.T.), Hannover Medical School, Hannover, Germany; and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Thomas Thum
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integrated Research and Treatment Center Transplantation (J.F., T.T.), and Excellence Cluster REBIRTH (J.F., T.T.), Hannover Medical School, Hannover, Germany; and National Heart and Lung Institute, Imperial College London, London, UK (T.T.).
| |
Collapse
|
65
|
Magnusson M, Larsson P, Lu EX, Bergh N, Carén H, Jern S. Rapid and specific hypomethylation of enhancers in endothelial cells during adaptation to cell culturing. Epigenetics 2016; 11:614-24. [PMID: 27302749 DOI: 10.1080/15592294.2016.1192734] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Epigenetics, including DNA methylation, is one way for a cell to respond to the surrounding environment. Traditionally, DNA methylation has been perceived as a quite stable modification; however, lately, there have been reports of a more dynamic CpG methylation that can be affected by, for example, long-term culturing. We recently reported that methylation in the enhancer of the gene encoding the key fibrinolytic enzyme tissue-type plasminogen activator (t-PA) was rapidly erased during cell culturing. In the present study we used sub-culturing of human umbilical vein endothelial cells (HUVECs) as a model of environmental challenge to examine how fast genome-wide methylation changes can arise. To assess genome-wide DNA methylation, the Infinium HumanMethylation450 BeadChip was used on primary, passage 0, and passage 4 HUVECs. Almost 2% of the analyzed sites changed methylation status to passage 4, predominantly displaying hypomethylation. Sites annotated as enhancers were overrepresented among the differentially methylated sites (DMSs). We further showed that half of the corresponding genes concomitantly altered their expression, most of them increasing in expression. Interestingly, the stroke-related gene HDAC9 increased its expression several hundredfold. This study reveals a rapid hypomethylation of CpG sites in enhancer elements during the early stages of cell culturing. As many methods for methylation analysis are biased toward CpG rich promoter regions, we suggest that such methods may not always be appropriate for the study of methylation dynamics. In addition, we found that significant changes in expression arose in genes with enhancer DMSs. HDAC9 displayed the most prominent increase in expression, indicating, for the first time, that dynamic enhancer methylation may be central in regulating this important stroke-associated gene.
Collapse
Affiliation(s)
- Mia Magnusson
- a Wallenberg Laboratory, Department of Molecular and Clinical Medicine , Institute of Medicine, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Pia Larsson
- a Wallenberg Laboratory, Department of Molecular and Clinical Medicine , Institute of Medicine, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Emma Xuchun Lu
- a Wallenberg Laboratory, Department of Molecular and Clinical Medicine , Institute of Medicine, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Niklas Bergh
- a Wallenberg Laboratory, Department of Molecular and Clinical Medicine , Institute of Medicine, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Helena Carén
- b Sahlgrenska Cancer Center, Department of Pathology , Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Sverker Jern
- a Wallenberg Laboratory, Department of Molecular and Clinical Medicine , Institute of Medicine, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
66
|
Welten S, Goossens E, Quax P, Nossent A. The multifactorial nature of microRNAs in vascular remodelling. Cardiovasc Res 2016; 110:6-22. [DOI: 10.1093/cvr/cvw039] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/07/2016] [Indexed: 12/22/2022] Open
|
67
|
Li Q, Hu B, Hu GW, Chen CY, Niu X, Liu J, Zhou SM, Zhang CQ, Wang Y, Deng ZF. tRNA-Derived Small Non-Coding RNAs in Response to Ischemia Inhibit Angiogenesis. Sci Rep 2016; 6:20850. [PMID: 26865164 PMCID: PMC4749989 DOI: 10.1038/srep20850] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 01/12/2016] [Indexed: 12/31/2022] Open
Abstract
Ischemic injuries will lead to necrotic tissue damage, and post-ischemia angiogenesis plays critical roles in blood flow restoration and tissue recovery. Recently, several types of small RNAs have been reported to be involved in this process. In this study, we first generated a rat brain ischemic model to investigate the involvement of new types of small RNAs in ischemia. We utilized deep sequencing and bioinformatics analyses to demonstrate that the level of small RNA fragments derived from tRNAs strikingly increased in the ischemic rat brain. Among these sequences, tRNAVal- and tRNAGly-derived small RNAs account for the most abundant segments. The up-regulation of tRNAVal- and tRNAGly-derived fragments was verified through northern blot and quantitative PCR analyses. The levels of these two fragments also increased in a mouse hindlimb ischemia model and cellular hypoxia model. Importantly, up-regulation of the tRNAVal- and tRNAGly-derived fragments in endothelial cells inhibited cell proliferation, migration and tube formation. Furthermore, we showed that these small RNAs are generated by angiogenin cleavage. Our results indicate that tRNA-derived fragments are involved in tissue ischemia, and we demonstrate for the first time that tRNAVal- and tRNAGly-derived fragments inhibit angiogenesis by modulating the function of endothelial cells.
Collapse
Affiliation(s)
- Qing Li
- Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Bin Hu
- Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Guo-Wen Hu
- Department of Neurosurgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Graduate School of Nanchang University, Nanchang 330006, China
| | - Chun-Yuan Chen
- Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Graduate School of Nanchang University, Nanchang 330006, China
| | - Xin Niu
- Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Juan Liu
- Department of Neurosurgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Shu-Min Zhou
- Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chang-Qing Zhang
- Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Department of Orthopedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yang Wang
- Institute of Microsurgery on Extremities, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zhi-Feng Deng
- Department of Neurosurgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
68
|
Sambri I, Crespo J, Aguiló S, Ingrosso D, Rodríguez C, Martínez González J. miR-17 and -20a Target the Neuron-Derived Orphan Receptor-1 (NOR-1) in Vascular Endothelial Cells. PLoS One 2015; 10:e0141932. [PMID: 26600038 PMCID: PMC4658114 DOI: 10.1371/journal.pone.0141932] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/14/2015] [Indexed: 11/19/2022] Open
Abstract
Neuron-derived orphan receptor-1 (NOR-1) plays a major role in vascular biology by controlling fibroproliferative and inflammatory responses. Because microRNAs (miRNAs) have recently emerged as key players in the regulation of gene expression in the vasculature, here we have investigated the regulation of NOR-1 by miRNAs in endothelial cells. Computational algorithms suggest that NOR-1 could be targeted by members of the miR-17 family. Accordingly, ectopic over-expression of miR-17 or miR-20a in endothelial cells using synthetic premiRNAs attenuated the up-regulation of NOR-1 expression induced by VEGF (as evidenced by real time PCR, Western blot and immunocitochemistry). Conversely, the antagonism of these miRNAs by specific antagomirs prevented the down-regulation of NOR-1 promoted by miR-17 or miR-20a in VEGF-stimulated cells. Disruption of the miRNA-NOR-1 mRNA interaction using a custom designed target protector evidenced the selectivity of these responses. Further, luciferase reporter assays and seed-sequence mutagenesis confirmed that miR-17 and -20a bind to NOR-1 3’-UTR. Finally, miR-17 and -20a ameliorated the up-regulation of VCAM-1 mediated by NOR-1 in VEGF-stimulated cells. Therefore, miR-17 and -20a target NOR-1 thereby regulating NOR-1-dependent gene expression.
Collapse
Affiliation(s)
- Irene Sambri
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
- Department of Biochemistry, Biophysics & General Pathology, School of Medicine & Surgery, Second University of Naples, Naples, Italy
| | - Javier Crespo
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Silvia Aguiló
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Diego Ingrosso
- Department of Biochemistry, Biophysics & General Pathology, School of Medicine & Surgery, Second University of Naples, Naples, Italy
| | - Cristina Rodríguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - José Martínez González
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
- * E-mail:
| |
Collapse
|
69
|
Crochiere M, Kashyap T, Kalid O, Shechter S, Klebanov B, Senapedis W, Saint-Martin JR, Landesman Y. Deciphering mechanisms of drug sensitivity and resistance to Selective Inhibitor of Nuclear Export (SINE) compounds. BMC Cancer 2015; 15:910. [PMID: 26573568 PMCID: PMC4647283 DOI: 10.1186/s12885-015-1790-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Exportin 1 (XPO1) is a well-characterized nuclear export protein whose expression is up-regulated in many types of cancers and functions to transport key tumor suppressor proteins (TSPs) from the nucleus. Karyopharm Therapeutics has developed a series of small-molecule Selective Inhibitor of Nuclear Export (SINE) compounds, which have been shown to block XPO1 function both in vitro and in vivo. The drug candidate, selinexor (KPT-330), is currently in Phase-II/IIb clinical trials for treatment of both hematologic and solid tumors. The present study sought to decipher the mechanisms that render cells either sensitive or resistant to treatment with SINE compounds, represented by KPT-185, an early analogue of KPT-330. METHODS Using the human fibrosarcoma HT1080 cell line, resistance to SINE was acquired over a period of 10 months of constant incubation with increasing concentration of KPT-185. Cell viability was assayed by MTT. Immunofluorescence was used to compare nuclear export of TSPs. Fluorescence activated cell sorting (FACS), quantitative polymerase chain reaction (qPCR), and immunoblots were used to measure effects on cell cycle, gene expression, and cell death. RNA from naïve and drug treated parental and resistant cells was analyzed by Affymetrix microarrays. RESULTS Treatment of HT1080 cells with gradually increasing concentrations of SINE resulted in >100 fold decrease in sensitivity to SINE cytotoxicity. Resistant cells displayed prolonged cell cycle, reduced nuclear accumulation of TSPs, and similar changes in protein expression compared to parental cells, however the magnitude of the protein expression changes were more significant in parental cells. Microarray analyses comparing parental to resistant cells indicate that a number of key signaling pathways were altered in resistant cells including expression changes in genes involved in adhesion, apoptosis, and inflammation. While the patterns of changes in transcription following drug treatment are similar in parental and resistant cells, the extent of response was more robust in the parental cells. CONCLUSIONS These results suggest that SINE resistance is conferred by alterations in signaling pathways downstream of XPO1 inhibition. Modulation of these pathways could potentially overcome the resistance to nuclear export inhibitors.
Collapse
Affiliation(s)
- Marsha Crochiere
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Trinayan Kashyap
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Ori Kalid
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Sharon Shechter
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - Boris Klebanov
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | - William Senapedis
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| | | | - Yosef Landesman
- Karyopharm Therapeutics Inc., 85 Wells Avenue, Newton, MA 02459, USA.
| |
Collapse
|
70
|
Genome-Wide Association Study of Peripheral Arterial Disease in a Japanese Population. PLoS One 2015; 10:e0139262. [PMID: 26488411 PMCID: PMC4619060 DOI: 10.1371/journal.pone.0139262] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/10/2015] [Indexed: 02/07/2023] Open
Abstract
Characteristics of peripheral arterial disease (PAD) are the occlusion or stenosis of multiple vessel sites caused mainly by atherosclerosis and chronic lower limb ischemia. To identify PAD susceptible loci, we conducted a genome-wide association study (GWAS) with 785 cases and 3,383 controls in a Japanese population using 431,666 single nucleotide polymorphisms (SNP). After staged analyses including a total of 3,164 cases and 20,134 controls, we identified 3 novel PAD susceptibility loci at IPO5/RAP2A, EDNRA and HDAC9 with genome wide significance (combined P = 6.8 x 10−14, 5.3 x 10−9 and 8.8 x 10−8, respectively). Fine-mapping at the IPO5/RAP2A locus revealed that rs9584669 conferred risk of PAD. Luciferase assay showed that the risk allele at this locus reduced expression levels of IPO5. To our knowledge, these are the first genetic risk factors for PAD.
Collapse
|
71
|
Chamorro-Jorganes A, Lee MY, Araldi E, Landskroner-Eiger S, Fernández-Fuertes M, Sahraei M, Quiles Del Rey M, van Solingen C, Yu J, Fernández-Hernando C, Sessa WC, Suárez Y. VEGF-Induced Expression of miR-17-92 Cluster in Endothelial Cells Is Mediated by ERK/ELK1 Activation and Regulates Angiogenesis. Circ Res 2015; 118:38-47. [PMID: 26472816 PMCID: PMC4703066 DOI: 10.1161/circresaha.115.307408] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/15/2015] [Indexed: 01/19/2023]
Abstract
Supplemental Digital Content is available in the text. Several lines of evidence indicate that the regulation of microRNA (miRNA) levels by different stimuli may contribute to the modulation of stimulus-induced responses. The miR-17–92 cluster has been linked to tumor development and angiogenesis, but its role in vascular endothelial growth factor–induced endothelial cell (EC) functions is unclear and its regulation is unknown.
Collapse
Affiliation(s)
- Aránzazu Chamorro-Jorganes
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Monica Y Lee
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Elisa Araldi
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Shira Landskroner-Eiger
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Marta Fernández-Fuertes
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Mahnaz Sahraei
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Maria Quiles Del Rey
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Coen van Solingen
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Jun Yu
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Carlos Fernández-Hernando
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - William C Sessa
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.)
| | - Yajaira Suárez
- From the Vascular Biology and Therapeutics Program (A.C.-J., M.Y.L., E.A., M.F.-F., M.S., J.Y., C.F.-H., W.C.S., Y.S.), Section of Comparative Medicine (A.C.-J., E.A., M.F.-F., M.S., M.Q.R., C.F.-H., Y.S.), Departments of Pathology (A.C.-J., E.A., M.F.-F., M.S., C.F.-H., Y.S.); Pharmacology (M.Y.L., S.L.-E., W.C.S.), and Internal Medicine, Section of Cardiovascular Medicine (J.Y.), Yale University School of Medicine, New Haven, CT; and Department of Medicine, New York University School of Medicine (C.S.).
| |
Collapse
|
72
|
Association of GWAS-supported loci rs2107595 in HDAC9 gene with ischemic stroke in southern Han Chinese. Gene 2015; 570:282-7. [DOI: 10.1016/j.gene.2015.06.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 06/06/2015] [Accepted: 06/15/2015] [Indexed: 11/22/2022]
|
73
|
Yan MS, Marsden PA. Epigenetics in the Vascular Endothelium: Looking From a Different Perspective in the Epigenomics Era. Arterioscler Thromb Vasc Biol 2015; 35:2297-306. [PMID: 26404488 DOI: 10.1161/atvbaha.115.305043] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 09/14/2015] [Indexed: 01/11/2023]
Abstract
Cardiovascular diseases are commonly thought to be complex, non-Mendelian diseases that are influenced by genetic and environmental factors. A growing body of evidence suggests that epigenetic pathways play a key role in vascular biology and might be involved in defining and transducing cardiovascular disease inheritability. In this review, we argue the importance of epigenetics in vascular biology, especially from the perspective of endothelial cell phenotype. We highlight and discuss the role of epigenetic modifications across the transcriptional unit of protein-coding genes, especially the role of intragenic chromatin modifications, which are underappreciated and not well characterized in the current era of genome-wide studies. Importantly, we describe the practical application of epigenetics in cardiovascular disease therapeutics.
Collapse
Affiliation(s)
- Matthew S Yan
- From the Department of Medical Biophysics (M.S.Y., P.A.M.) and Department of Medicine, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.S.Y., P.A.M.), University of Toronto, Toronto, Ontario, Canada
| | - Philip A Marsden
- From the Department of Medical Biophysics (M.S.Y., P.A.M.) and Department of Medicine, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.S.Y., P.A.M.), University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
74
|
Renaud L, Harris LG, Mani SK, Kasiganesan H, Chou JC, Baicu CF, Van Laer A, Akerman AW, Stroud RE, Jones JA, Zile MR, Menick DR. HDACs Regulate miR-133a Expression in Pressure Overload-Induced Cardiac Fibrosis. Circ Heart Fail 2015; 8:1094-104. [PMID: 26371176 DOI: 10.1161/circheartfailure.114.001781] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 09/02/2015] [Indexed: 01/04/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) and histone deacetylases (HDACs) serve a significant role in the pathogenesis of a variety of cardiovascular diseases. The transcriptional regulation of miRNAs is poorly understood in cardiac hypertrophy. We investigated whether the expression of miR-133a is epigenetically regulated by class I and IIb HDACs during hypertrophic remodeling. METHODS AND RESULTS Transverse aortic constriction (TAC) was performed in CD1 mice to induce pressure overload hypertrophy. Mice were treated with class I and IIb HDAC inhibitor (HDACi) via drinking water for 2 and 4 weeks post TAC. miRNA expression was determined by real-time polymerase chain reaction. Echocardiography was performed at baseline and post TAC end points for structural and functional assessment. Chromatin immunoprecipitation was used to identify HDACs and transcription factors associated with miR-133a promoter. miR-133a expression was downregulated by 0.7- and 0.5-fold at 2 and 4 weeks post TAC, respectively, when compared with vehicle control (P<0.05). HDAC inhibition prevented this significant decrease 2 weeks post TAC and maintained miR-133a expression near vehicle control levels, which coincided with (1) a decrease in connective tissue growth factor expression, (2) a reduction in cardiac fibrosis and left atrium diameter (marker of end-diastolic pressure), suggesting an improvement in diastolic function. Chromatin immunoprecipitation analysis revealed that HDAC1 and HDAC2 are present on the miR-133a enhancer regions. CONCLUSIONS The results reveal that HDACs play a role in the regulation of pressure overload-induced miR-133a downregulation. This work is the first to provide insight into an epigenetic-miRNA regulatory pathway in pressure overload-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Ludivine Renaud
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Lillianne G Harris
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Santhosh K Mani
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Harinath Kasiganesan
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - James C Chou
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Catalin F Baicu
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - An Van Laer
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Adam W Akerman
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Robert E Stroud
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Jeffrey A Jones
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Michael R Zile
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Donald R Menick
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.).
| |
Collapse
|
75
|
Rader DJ. Human genetics of atherothrombotic disease and its risk factors. Arterioscler Thromb Vasc Biol 2015; 35:741-7. [PMID: 25810293 DOI: 10.1161/atvbaha.115.305492] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Daniel J Rader
- From the Departments of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| |
Collapse
|
76
|
Kim Y, Eom S, Park D, Kim H, Jeoung D. The Hyaluronic Acid-HDAC3-miRNA Network in Allergic Inflammation. Front Immunol 2015; 6:210. [PMID: 25983734 PMCID: PMC4415435 DOI: 10.3389/fimmu.2015.00210] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/17/2015] [Indexed: 12/19/2022] Open
Abstract
We previously reported the anti-allergic effect of high molecular weight form of hyaluronic acid (HMW-HA). In doing so, HA targets CD44 and inhibits FcεRI signaling and cross-talk between epidermal growth factor receptor (EGFR) and FcεRI. We previously reported the role of histone deacetylases (HDACs) in allergic inflammation and allergic inflammation-promoted enhanced tumorigenic potential. We reported regulatory role of HA in the expression of HDAC3. In this review, we will discuss molecular mechanisms associated with anti-allergic effect of HA in relation with HDACs. The role of microRNAs (miRNAs) in allergic inflammation has been reported. We will also discuss the role of miRNAs in allergic inflammation in relation with HA-mediated anti-allergic effects.
Collapse
Affiliation(s)
- Youngmi Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University , Chuncheon , South Korea
| | - Sangkyung Eom
- Department of Biochemistry, College of Natural Sciences, Kangwon National University , Chuncheon , South Korea
| | - Deokbum Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University , Chuncheon , South Korea
| | - Hyuna Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University , Chuncheon , South Korea
| | - Dooil Jeoung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University , Chuncheon , South Korea
| |
Collapse
|
77
|
Gore J, Craven KE, Wilson JL, Cote GA, Cheng M, Nguyen HV, Cramer HM, Sherman S, Korc M. TCGA data and patient-derived orthotopic xenografts highlight pancreatic cancer-associated angiogenesis. Oncotarget 2015; 6:7504-21. [PMID: 25762644 PMCID: PMC4480696 DOI: 10.18632/oncotarget.3233] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 01/28/2015] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinomas (PDACs) overexpress pro-angiogenic factors but are not viewed as vascular. Using data from The Cancer Genome Atlas we demonstrate that a subset of PDACs exhibits a strong pro-angiogenic signature that includes 37 genes, such as HDAC9, that are overexpressed in PDAC arising in KRC mice, which express mutated Kras and lack RB. Moreover, patient-derived orthotopic xenografts can exhibit tumor angiogenesis, whereas conditioned media (CM) from KRC-derived pancreatic cancer cells (PCCs) enhance endothelial cell (EC) growth and migration, and activate canonical TGF-β signaling and STAT3. Inhibition of the type I TGF-β receptor with SB505124 does not alter endothelial activation in vitro, but decreases pro-angiogenic gene expression and suppresses angiogenesis in vivo. Conversely, STAT3 silencing or JAK1-2 inhibition with ruxolitinib blocks CM-enhanced EC proliferation. STAT3 disruption also suppresses endothelial HDAC9 and blocks CM-induced HDAC9 expression, whereas HDAC9 re-expression restores CM-enhanced endothelial proliferation. Moreover, ruxolitinib blocks mitogenic EC/PCC cross-talk, and suppresses endothelial p-STAT3 and HDAC9, and PDAC progression and angiogenesis in vivo, while markedly prolonging survival of KRC mice. Thus, targeting JAK1-2 with ruxolitinib blocks a final pathway that is common to multiple pro-angiogenic factors, suppresses EC-mediated PCC proliferation, and may be useful in PDACs with a strong pro-angiogenic signature.
Collapse
Affiliation(s)
- Jesse Gore
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- The Melvin and Bren Simon Cancer Center, and the Center for Pancreatic Cancer Research, Indianapolis, IN 46202, USA
| | - Kelly E. Craven
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Julie L. Wilson
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gregory A. Cote
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- The Melvin and Bren Simon Cancer Center, and the Center for Pancreatic Cancer Research, Indianapolis, IN 46202, USA
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Monica Cheng
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hai V. Nguyen
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Harvey M. Cramer
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stuart Sherman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- The Melvin and Bren Simon Cancer Center, and the Center for Pancreatic Cancer Research, Indianapolis, IN 46202, USA
| | - Murray Korc
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- The Melvin and Bren Simon Cancer Center, and the Center for Pancreatic Cancer Research, Indianapolis, IN 46202, USA
| |
Collapse
|
78
|
Swierczynski S, Klieser E, Illig R, Alinger-Scharinger B, Kiesslich T, Neureiter D. Histone deacetylation meets miRNA: epigenetics and post-transcriptional regulation in cancer and chronic diseases. Expert Opin Biol Ther 2015; 15:651-64. [PMID: 25766312 DOI: 10.1517/14712598.2015.1025047] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Epigenetic regulation via DNA methylation, histone acetylation, as well as by microRNAs (miRNAs) is currently in the scientific focus due to its role in carcinogenesis and its involvement in initiation, progression and metastasis. While many target genes of DNA methylation, histone acetylation and miRNAs are known, even less information exists as to how these mechanisms cooperate and how they may regulate each other in a specific pathological context. For further development of therapeutic approaches, this review presents the current status of the crosstalk of histone acetylation and miRNAs in human carcinogenesis and chronic diseases. AREAS COVERED This article reviews information from comprehensive PubMed searches to evaluate relevant literature with a focus on possible association between histone acetylation, miRNAs and their targets. Our analysis identified specific miRNAs which collaborate with histone deacetylases (HDACs) and cooperatively regulate several relevant target genes. EXPERT OPINION Fourteen miRNAs could be linked to the expression of eight HDACs influencing the α-(1,6)-fucosyltransferase, polycystin-2 and the fibroblast-growth-factor 2 pathways. Focusing on the complex linkage of miRNA and HDAC expression could give deeper insights in new 'druggable' targets and might provide possible novel therapeutic approaches in future.
Collapse
Affiliation(s)
- Stefan Swierczynski
- Paracelsus Medical University, Salzburger Landeskliniken, Department of Surgery , Salzburg , Austria
| | | | | | | | | | | |
Collapse
|
79
|
Fraineau S, Palii CG, Allan DS, Brand M. Epigenetic regulation of endothelial-cell-mediated vascular repair. FEBS J 2015; 282:1605-29. [PMID: 25546332 DOI: 10.1111/febs.13183] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 01/16/2023]
Abstract
Maintenance of vascular integrity is essential for the prevention of vascular disease and for recovery following cardiovascular, cerebrovascular and peripheral vascular events including limb ischemia, heart attack and stroke. Endothelial stem/progenitor cells have recently gained considerable interest due to their potential use in stem cell therapies to mediate revascularization after ischemic injury. Therefore, there is an urgent need to understand fundamental mechanisms regulating vascular repair in specific cell types to develop new beneficial therapeutic interventions. In this review, we highlight recent studies demonstrating that epigenetic mechanisms (including post-translational modifications of DNA and histones as well as non-coding RNA-mediated processes) play essential roles in the regulation of endothelial stem/progenitor cell functions through modifying chromatin structure. Furthermore, we discuss the potential of using small molecules that modulate the activities of epigenetic enzymes to enhance the vascular repair function of endothelial cells and offer insight on potential strategies that may accelerate clinical applications.
Collapse
Affiliation(s)
- Sylvain Fraineau
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Canada; Ottawa Institute of Systems Biology, Canada
| | | | | | | |
Collapse
|
80
|
Volný O, Kašičková L, Coufalová D, Cimflová P, Novák J. microRNAs in Cerebrovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 888:155-95. [PMID: 26663183 DOI: 10.1007/978-3-319-22671-2_9] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cardiovascular diseases are major causes of morbidity and mortality in developed countries. Cerebrovascular diseases, especially stroke, represent major burden of disability and economy impact. Major advances in primary and secondary prevention and therapy are needed in order to tackle this public health problem. Our better understanding of pathophysiology is essential in order to develop novel diagnostic and therapeutic tools and strategies. microRNAs are a family of important post-transcriptional regulators of gene expression and their involvement in the pathophysiology of cerebrovascular diseases has already been reported. Moreover, microRNAs may represent above-mentioned potential diagnostic and therapeutic tools in clinical practice. Within this chapter, we briefly describe basic epidemiology, aetiology and clinical manifestation of following cerebrovascular diseases: extracranial carotid atherosclerosis, acute stroke, intracranial aneurysms and cerebral arterio-venous malformations. Further, in each chapter, the current knowledge about the involvement of specific microRNAs and their potential use in clinical practice will be summarized. More specifically, within the subchapter "miRNAs in carotid atherosclerosis", general information about miRNA involvement in atherosclerosis will be described (miR-126, miR-17-92, miR-155 and others) with special emphasis put on miRNAs affecting carotid plaque progression and stability (e.g. miR-145, miR-146 or miR-217). In the subchapter "miRNAs in acute stroke", we will provide insight into recent knowledge from animal and human studies concerning miRNA profiling in acute stroke and their expression dynamics in brain tissue and extracellular fluids (roles of, e.g. let-7 family, miR-21, miR-29 family, miR-124, miR-145, miR-181 family, miR-210 and miR-223). Subchapters dealing with "miRNAs and AV malformations" and "miRNAs and intracranial aneurysms" will focus on miR-21, miR-26, miR-29 family and miR-143/145.
Collapse
Affiliation(s)
- Ondřej Volný
- Department of Neurology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekarska 53, Brno, 656 91, Czech Republic. .,Department of Anatomy, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic. .,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, Brno, 656 91, Czech Republic.
| | - Linda Kašičková
- Department of Neurology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekarska 53, Brno, 656 91, Czech Republic. .,Department of Anatomy, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
| | - Dominika Coufalová
- Department of Anatomy, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic. .,International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, Brno, 656 91, Czech Republic.
| | - Petra Cimflová
- Department of Radiology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekarska 53, Brno, 656 91, Czech Republic.
| | - Jan Novák
- 2nd Department of Internal Medicine, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekarska 53, Brno, 656 91, Czech Republic. .,Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic. .,Department of Physiology, Faculty of Medicine, Masaryk University, Brno, 62500, Czech Republic.
| |
Collapse
|
81
|
Azghandi S, Prell C, van der Laan SW, Schneider M, Malik R, Berer K, Gerdes N, Pasterkamp G, Weber C, Haffner C, Dichgans M. Deficiency of the stroke relevant HDAC9 gene attenuates atherosclerosis in accord with allele-specific effects at 7p21.1. Stroke 2014; 46:197-202. [PMID: 25388417 DOI: 10.1161/strokeaha.114.007213] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Recent genome-wide association studies identified the histone deacetylase 9 (HDAC9) gene region as a major risk locus for large-vessel stroke and coronary artery disease. However, the mechanisms linking variants at this locus to vascular risk are poorly understood. In this study, we investigated the candidacy and directionality of HDAC9 in atherosclerosis and analyzed associations between risk alleles at 7p21.1 and plaque characteristics. METHODS Allele-dependent expression of HDAC9 was analyzed in human peripheral blood mononuclear cells of healthy donors. Effects of HDAC9 deficiency on atherosclerotic plaques were investigated in 18- and 28-week-old ApoE(-/-) mice by histology and immunohistochemistry. We further performed detailed plaque phenotyping and genotyping of rs2107595, the lead single-nucleotide polymorphism for large-vessel stroke, in carotid endarterectomy samples of 1858 subjects from the Athero-Express study. RESULTS Gene expression studies in peripheral blood mononuclear cells revealed increased mRNA levels of HDAC9 but not of neighboring genes (TWIST1/FERD3L) in risk allele carriers of rs2107595. Compared with HDAC9(+/+)ApoE(-/-) mice, HDAC9(-/-)ApoE(-/-) mice exhibited markedly reduced lesion sizes throughout atherosclerotic aortas and significantly less advanced lesions. The proportion of Mac3-positive macrophages was higher in plaques from HDAC9(-/-)ApoE(-/-) mice, but this was largely because of a lower proportion of advanced lesions. Analysis of human atherosclerotic plaques revealed no association between rs2107595 and specific plaque characteristics. CONCLUSIONS Our results suggest that HDAC9 represents the disease-relevant gene at the stroke and coronary artery disease risk locus on 7p21.1, and that risk alleles in this region mediate their effects through increased HDAC9 expression. Targeted inhibition of HDAC9 might be a viable strategy to prevent atherosclerosis.
Collapse
Affiliation(s)
- Sepiede Azghandi
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Caroline Prell
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Sander W van der Laan
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Manuela Schneider
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Rainer Malik
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Kerstin Berer
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Norbert Gerdes
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Gerard Pasterkamp
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Christian Weber
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Christof Haffner
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Martin Dichgans
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (S.A., C.P., M.S., R.M., C.H., M.D.); Divisions Heart and Lungs and Laboratories and Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands (S.W.v.d.L., G.P.); Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany (K.B.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (N.G., C.W.); and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.).
| |
Collapse
|
82
|
Eom GH, Kook H. Posttranslational modifications of histone deacetylases: Implications for cardiovascular diseases. Pharmacol Ther 2014; 143:168-80. [DOI: 10.1016/j.pharmthera.2014.02.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 02/25/2014] [Indexed: 02/08/2023]
|
83
|
Cao Q, Rong S, Repa JJ, St Clair R, Parks JS, Mishra N. Histone deacetylase 9 represses cholesterol efflux and alternatively activated macrophages in atherosclerosis development. Arterioscler Thromb Vasc Biol 2014; 34:1871-9. [PMID: 25035344 DOI: 10.1161/atvbaha.114.303393] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Recent genome-wide association studies revealed that a genetic variant in the loci corresponding to histone deacetylase 9 (HDAC9) is associated with large vessel stroke. HDAC9 expression was upregulated in human atherosclerotic plaques in different arteries. The molecular mechanisms how HDAC9 might increase atherosclerosis is not clear. APPROACH AND RESULTS In this study, we show that systemic and bone marrow cell deletion of HDAC9 decreased atherosclerosis in LDLr(-/-) (low density lipoprotein receptor) mice with minimal effect on plasma lipid concentrations. HDAC9 deletion resulted upregulation of lipid homeostatic genes, downregulation of inflammatory genes, and polarization toward an M2 phenotype via increased accumulation of total acetylated H3 and H3K9 at the promoters of ABCA1 (ATP-binding cassette transporter), ABCG1, and PPAR-γ (peroxisome proliferator-activated receptor) in macrophages. CONCLUSIONS We conclude that macrophage HDAC9 upregulation is atherogenic via suppression of cholesterol efflux and generation of alternatively activated macrophages in atherosclerosis.
Collapse
Affiliation(s)
- Qiang Cao
- From the Departments of Internal Medicine/Rheumatology and Immunology (Q.C., N.M.), Pathology/Lipid Sciences (S.R., R.S.C., J.S.P.), and Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; and Departments of Internal Medicine and Physiology, The University of Texas Southwestern Medical Center, Dallas (J.J.R.)
| | - Shunxing Rong
- From the Departments of Internal Medicine/Rheumatology and Immunology (Q.C., N.M.), Pathology/Lipid Sciences (S.R., R.S.C., J.S.P.), and Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; and Departments of Internal Medicine and Physiology, The University of Texas Southwestern Medical Center, Dallas (J.J.R.)
| | - Joyce J Repa
- From the Departments of Internal Medicine/Rheumatology and Immunology (Q.C., N.M.), Pathology/Lipid Sciences (S.R., R.S.C., J.S.P.), and Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; and Departments of Internal Medicine and Physiology, The University of Texas Southwestern Medical Center, Dallas (J.J.R.)
| | - Richard St Clair
- From the Departments of Internal Medicine/Rheumatology and Immunology (Q.C., N.M.), Pathology/Lipid Sciences (S.R., R.S.C., J.S.P.), and Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; and Departments of Internal Medicine and Physiology, The University of Texas Southwestern Medical Center, Dallas (J.J.R.)
| | - John S Parks
- From the Departments of Internal Medicine/Rheumatology and Immunology (Q.C., N.M.), Pathology/Lipid Sciences (S.R., R.S.C., J.S.P.), and Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; and Departments of Internal Medicine and Physiology, The University of Texas Southwestern Medical Center, Dallas (J.J.R.)
| | - Nilamadhab Mishra
- From the Departments of Internal Medicine/Rheumatology and Immunology (Q.C., N.M.), Pathology/Lipid Sciences (S.R., R.S.C., J.S.P.), and Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; and Departments of Internal Medicine and Physiology, The University of Texas Southwestern Medical Center, Dallas (J.J.R.).
| |
Collapse
|
84
|
Aung HH, Tsoukalas A, Rutledge JC, Tagkopoulos I. A systems biology analysis of brain microvascular endothelial cell lipotoxicity. BMC SYSTEMS BIOLOGY 2014; 8:80. [PMID: 24993133 PMCID: PMC4112729 DOI: 10.1186/1752-0509-8-80] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/23/2014] [Indexed: 02/08/2023]
Abstract
Background Neurovascular inflammation is associated with a number of neurological diseases including vascular dementia and Alzheimer’s disease, which are increasingly important causes of morbidity and mortality around the world. Lipotoxicity is a metabolic disorder that results from accumulation of lipids, particularly fatty acids, in non-adipose tissue leading to cellular dysfunction, lipid droplet formation, and cell death. Results Our studies indicate for the first time that the neurovascular circulation also can manifest lipotoxicity, which could have major effects on cognitive function. The penetration of integrative systems biology approaches is limited in this area of research, which reduces our capacity to gain an objective insight into the signal transduction and regulation dynamics at a systems level. To address this question, we treated human microvascular endothelial cells with triglyceride-rich lipoprotein (TGRL) lipolysis products and then we used genome-wide transcriptional profiling to obtain transcript abundances over four conditions. We then identified regulatory genes and their targets that have been differentially expressed through analysis of the datasets with various statistical methods. We created a functional gene network by exploiting co-expression observations through a guilt-by-association assumption. Concomitantly, we used various network inference algorithms to identify putative regulatory interactions and we integrated all predictions to construct a consensus gene regulatory network that is TGRL lipolysis product specific. Conclusion System biology analysis has led to the validation of putative lipid-related targets and the discovery of several genes that may be implicated in lipotoxic-related brain microvascular endothelial cell responses. Here, we report that activating transcription factors 3 (ATF3) is a principal regulator of TGRL lipolysis products-induced gene expression in human brain microvascular endothelial cell.
Collapse
Affiliation(s)
| | | | | | - Ilias Tagkopoulos
- UC Davis Genome Center, University of California, Davis, CA 95616, USA.
| |
Collapse
|
85
|
Daniel JM, Penzkofer D, Teske R, Dutzmann J, Koch A, Bielenberg W, Bonauer A, Boon RA, Fischer A, Bauersachs J, van Rooij E, Dimmeler S, Sedding DG. Inhibition of miR-92a improves re-endothelialization and prevents neointima formation following vascular injury. Cardiovasc Res 2014; 103:564-72. [PMID: 25020912 PMCID: PMC4145012 DOI: 10.1093/cvr/cvu162] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Aims MicroRNA (miR)-92a is an important regulator of endothelial proliferation and angiogenesis after ischaemia, but the effects of miR-92a on re-endothelialization and neointimal lesion formation after vascular injury remain elusive. We tested the effects of lowering miR-92a levels using specific locked nucleic acid (LNA)-based antimiRs as well as endothelial-specific knock out of miR-92a on re-endothelialization and neointimal formation after wire-induced injury of the femoral artery in mice. Methods and results MiR-92a was significantly up-regulated in neointimal lesions following wire-induced injury. Pre-miR-92a overexpression resulted in repression of the direct miR-92a target genes integrin α5 and sirtuin1, and reduced eNOS expression in vitro. MiR-92a impaired proliferation and migration of endothelial cells but not smooth muscle cells. In vivo, systemic inhibition of miR-92a expression with LNA-modified antisense molecules resulted in a significant acceleration of re-endothelialization of the denuded vessel area. Genetic deletion of miR-92a in Tie2-expressing cells, representing mainly endothelial cells, enhanced re-endothelialization, whereas no phenotype was observed in mice lacking miR-92a expression in haematopoietic cells. The enhanced endothelial recovery was associated with reduced accumulation of leucocytes and inhibition of neointimal formation 21 days after injury and led to the de-repression of the miR-92a targets integrin α5 and sirtuin1. Conclusion Our data indicate that inhibition of endothelial miR-92a attenuates neointimal lesion formation by accelerating re-endothelialization and thus represents a putative novel mechanism to enhance the functional recovery following vascular injury.
Collapse
Affiliation(s)
- Jan-Marcus Daniel
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover D-30625, Germany Department of Cardiology, University Hospital Giessen & Marburg, Giessen, Germany
| | - Daniela Penzkofer
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Rebecca Teske
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover D-30625, Germany Department of Cardiology, University Hospital Giessen & Marburg, Giessen, Germany
| | - Jochen Dutzmann
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover D-30625, Germany
| | - Alexander Koch
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover D-30625, Germany Department of Cardiology, University Hospital Giessen & Marburg, Giessen, Germany
| | - Wiebke Bielenberg
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover D-30625, Germany Department of Cardiology, University Hospital Giessen & Marburg, Giessen, Germany
| | - Angelika Bonauer
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Reinier A Boon
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Ariane Fischer
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover D-30625, Germany
| | | | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Daniel G Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover D-30625, Germany Department of Cardiology, University Hospital Giessen & Marburg, Giessen, Germany
| |
Collapse
|
86
|
Zhang P, Li C, Shao Y, Chen X, Li Y, Su X, Li T. Identification and characterization of miR-92a and its targets modulating Vibrio splendidus challenged Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2014; 38:383-388. [PMID: 24747055 DOI: 10.1016/j.fsi.2014.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 03/18/2014] [Accepted: 04/08/2014] [Indexed: 06/03/2023]
Abstract
miR-92a is a kind of disease related fine-tuning regulator which is not only related with tumorigenesis and tumor development but also participates in host-pathogen interaction in vertebrates. In present study, the potential targets of miR-92a in Apostichopus japonicus coelomocytes were screened by high-throughout sequencing and PCR approaches. Total of 10 annotated candidates were identified by hybrid PCR, and 23 were verified from RNA-seq, in which SMURF, PCBP and MEGF were found in both methods. The expression patterns of miR-92a and some putative targets were further characterized by qPCR at cell and individual levels. Vibrio splendidus and LPS exposure could significantly increase the expression level of sea cucumber miR-92a at all examined time points. Accordingly, strictly negative correlation expression profiles were detected in two candidates genes of MEGF and SMURF, suggesting that these two genes showed higher possibilities to be the targets of miR-92a in sea cucumber. Overall, the present work will enhance our understanding in the context of miR-92a modulating the interaction of sea cucumber upon pathogen challenged.
Collapse
Affiliation(s)
- Pengjuan Zhang
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province 315211, PR China
| | - Chenghua Li
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province 315211, PR China.
| | - Yina Shao
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province 315211, PR China
| | - Xiaochong Chen
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province 315211, PR China
| | - Ye Li
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province 315211, PR China
| | - Xiurong Su
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province 315211, PR China
| | - Taiwu Li
- Ningbo City College of Vocational Technology, Ningbo 315100, PR China
| |
Collapse
|
87
|
Michalik KM, You X, Manavski Y, Doddaballapur A, Zörnig M, Braun T, John D, Ponomareva Y, Chen W, Uchida S, Boon RA, Dimmeler S. Long noncoding RNA MALAT1 regulates endothelial cell function and vessel growth. Circ Res 2014; 114:1389-97. [PMID: 24602777 DOI: 10.1161/circresaha.114.303265] [Citation(s) in RCA: 755] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
RATIONALE The human genome harbors a large number of sequences encoding for RNAs that are not translated but control cellular functions by distinct mechanisms. The expression and function of the longer transcripts namely the long noncoding RNAs in the vasculature are largely unknown. OBJECTIVE Here, we characterized the expression of long noncoding RNAs in human endothelial cells and elucidated the function of the highly expressed metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). METHODS AND RESULTS Endothelial cells of different origin express relative high levels of the conserved long noncoding RNAs MALAT1, taurine upregulated gene 1 (TUG1), maternally expressed 3 (MEG3), linc00657, and linc00493. MALAT1 was significantly increased by hypoxia and controls a phenotypic switch in endothelial cells. Silencing of MALAT1 by small interfering RNAs or GapmeRs induced a promigratory response and increased basal sprouting and migration, whereas proliferation of endothelial cells was inhibited. When angiogenesis was further stimulated by vascular endothelial growth factor, MALAT1 small interfering RNAs induced discontinuous sprouts indicative of defective proliferation of stalk cells. In vivo studies confirmed that genetic ablation of MALAT1 inhibited proliferation of endothelial cells and reduced neonatal retina vascularization. Pharmacological inhibition of MALAT1 by GapmeRs reduced blood flow recovery and capillary density after hindlimb ischemia. Gene expression profiling followed by confirmatory quantitative reverse transcriptase-polymerase chain reaction demonstrated that silencing of MALAT1 impaired the expression of various cell cycle regulators. CONCLUSIONS Silencing of MALAT1 tips the balance from a proliferative to a migratory endothelial cell phenotype in vitro, and its genetic deletion or pharmacological inhibition reduces vascular growth in vivo.
Collapse
Affiliation(s)
- Katharina M Michalik
- From the Institute of Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany (K.M.M., Y.M., A.D., D.J., Y.P., S.U., R.A.B., S.D.); Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin-Buch, Germany (X.Y., W.C.); Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Tumor Therapy, Frankfurt, Germany (M.Z.); Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (T.B., S.U., S.D.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Shiva Shankar TV, Willems L. Epigenetic modulators mitigate angiogenesis through a complex transcriptomic network. Vascul Pharmacol 2014; 60:57-66. [PMID: 24445350 DOI: 10.1016/j.vph.2014.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/18/2013] [Accepted: 01/08/2014] [Indexed: 12/19/2022]
Abstract
In this review, we summarize the knowledge pertaining to the role of epigenetics in the regulation of angiogenesis. In particular, we show that lysine acetylation and cytosine methylation are important transcriptional regulators of angiogenic genes in endothelial cells. Lysine acetylation and cytosine methylation inhibitors idiosyncratically tune the transcriptome and affect expression of key modulators of angiogenesis such as VEGF and eNOS. Transcriptomic profiling also reveals a series of novel genes that are concomitantly affected by epigenetic modulators. The reversibility and overall tolerability of currently available epigenetic inhibitors open up the prospect of therapeutic intervention in pathologies where angiogenesis is exacerbated. This type of multitargeted strategy has the major advantage of overcoming the compensatory feedback mechanisms that characterize single anti-angiogenic factors.
Collapse
Affiliation(s)
- T V Shiva Shankar
- Molecular and Cellular Epigenetics (GIGA-Cancer) and Molecular Biology (GxABT), University of Liège (ULg), Liège, Belgium
| | - L Willems
- Molecular and Cellular Epigenetics (GIGA-Cancer) and Molecular Biology (GxABT), University of Liège (ULg), Liège, Belgium.
| |
Collapse
|
89
|
Li M, Guan X, Sun Y, Mi J, Shu X, Liu F, Li C. miR-92a family and their target genes in tumorigenesis and metastasis. Exp Cell Res 2014; 323:1-6. [PMID: 24394541 DOI: 10.1016/j.yexcr.2013.12.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/18/2013] [Indexed: 01/01/2023]
Abstract
The miR-92a family, including miR-25, miR-92a-1, miR-92a-2 and miR-363, arises from three different paralog clusters miR-17-92, miR-106a-363, and miR-106b-25 that are highly conservative in the process of evolution, and it was thought as a group of microRNAs (miRNAs) correlated with endothelial cells. Aberrant expression of miR-92a family was detected in multiple cancers, and the disturbance of miR-92a family was related with tumorigenesis and tumor development. In this review, the progress on the relationship between miR-92a family and their target genes and malignant tumors will be summarized.
Collapse
Affiliation(s)
- Molin Li
- Department of Pathophysiology, Basic Medical Science of Dalian Medical University, Dalian 116044, China; Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Dalian 116044, China.
| | - Xingfang Guan
- Department of Pathophysiology, Basic Medical Science of Dalian Medical University, Dalian 116044, China
| | - Yuqiang Sun
- Department of Pathophysiology, Basic Medical Science of Dalian Medical University, Dalian 116044, China
| | - Jun Mi
- Institute of Cancer Stem Cell, Dalian Medical University Cancer Center, Dalian 116044, China
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University Cancer Center, Dalian 116044, China
| | - Fang Liu
- Department of Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Chuangang Li
- Department of Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China.
| |
Collapse
|
90
|
Lee SE, Park YS. Gene expression profiling of human umbilical vein endothelial cells exposed to myricetin. BIOCHIP JOURNAL 2013. [DOI: 10.1007/s13206-013-7404-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
91
|
Flowers E, Aouizerat BE. MicroRNA associated with dyslipidemia and coronary disease in humans. Physiol Genomics 2013; 45:1199-205. [PMID: 24170031 DOI: 10.1152/physiolgenomics.00106.2013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
MicroRNAs are structural components of an epigenetic mechanism of posttranscriptional regulation of messenger RNA translation. Recently, there has been significant interest in the application of microRNA as a blood-based biomarker of underlying physiological conditions. Dyslipidemia is a complex, heterogeneous condition conferring substantially increased risk for cardiovascular disease. The purpose of this review is to describe the current body of knowledge on the role of microRNA regulation of lipoprotein metabolism in humans and to discuss relevant methodological and study design considerations. We highlight the potential roles for microRNA in gene-environment interactions.
Collapse
Affiliation(s)
- Elena Flowers
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, California; and
| | | |
Collapse
|
92
|
McAlinden A, Varghese N, Wirthlin L, Chang LW. Differentially expressed microRNAs in chondrocytes from distinct regions of developing human cartilage. PLoS One 2013; 8:e75012. [PMID: 24040378 PMCID: PMC3767648 DOI: 10.1371/journal.pone.0075012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/11/2013] [Indexed: 12/21/2022] Open
Abstract
There is compelling in vivo evidence from reports on human genetic mutations and transgenic mice that some microRNAs (miRNAs) play an important functional role in regulating skeletal development and growth. A number of published in vitro studies also point toward a role for miRNAs in controlling chondrocyte gene expression and differentiation. However, information on miRNAs that may regulate a specific phase of chondrocyte differentiation (i.e. production of progenitor, differentiated or hypertrophic chondrocytes) is lacking. To attempt to bridge this knowledge gap, we have investigated miRNA expression patterns in human embryonic cartilage tissue. Specifically, a developmental time point was selected, prior to endochondral ossification in the embryonic limb, to permit analysis of three distinct populations of chondrocytes. The location of chondroprogenitor cells, differentiated chondrocytes and hypertrophic chondrocytes in gestational day 54-56 human embryonic limb tissue sections was confirmed both histologically and by specific collagen expression patterns. Laser capture microdissection was utilized to separate the three chondrocyte populations and a miRNA profiling study was carried out using TaqMan® OpenArray® Human MicroRNA Panels (Applied Biosystems®). Here we report on abundantly expressed miRNAs in human embryonic cartilage tissue and, more importantly, we have identified miRNAs that are significantly differentially expressed between precursor, differentiated and hypertrophic chondrocytes by 2-fold or more. Some of the miRNAs identified in this study have been described in other aspects of cartilage or bone biology, while others have not yet been reported in chondrocytes. Finally, a bioinformatics approach was applied to begin to decipher developmental cellular pathways that may be regulated by groups of differentially expressed miRNAs during distinct stages of chondrogenesis. Data obtained from this work will serve as an important resource of information for the field of cartilage biology and will enhance our understanding of miRNA-driven mechanisms regulating cartilage and endochondral bone development, regeneration and repair.
Collapse
Affiliation(s)
- Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University, St Louis, Missouri, United States of America
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, United States of America
| | - Nobish Varghese
- Department of Pathology and Immunology, Washington University, St Louis, Missouri, United States of America
| | - Louisa Wirthlin
- Department of Orthopaedic Surgery, Washington University, St Louis, Missouri, United States of America
| | - Li-Wei Chang
- Department of Pathology and Immunology, Washington University, St Louis, Missouri, United States of America
| |
Collapse
|