51
|
Epidermal Growth Factor Like-domain 7 and miR-126 are abnormally expressed in diffuse Systemic Sclerosis fibroblasts. Sci Rep 2019; 9:4589. [PMID: 30872612 PMCID: PMC6418261 DOI: 10.1038/s41598-019-39485-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/12/2018] [Indexed: 11/10/2022] Open
Abstract
Systemic sclerosis (SSc) is characterized by microangiopathy with impaired reparative angiogenesis and fibrosis. Epidermal Growth Factor Like-domain 7 (EGFL7), firstly described in endothelial cells plays a pivotal role in angiogenesis. Fibroblasts (FBs) are involved in vascular remodeling, under physiological and pathological conditions. In this study, we investigated: (i) the expression of EGFL7 and its miR-126 in patients affected by diffuse cutaneous SSc (dcSSc); (ii) the ability of Transforming Growth Factor-beta (TGF-β) to modulate EGFL7 expression; (iii) the ability of EGFL7 to modulate COL1A1 expression and proliferation/migration, and (iv) the functional role of EGFL7 on angiogenesis. Patients were divided in 2 subsets: patients fulfilling the classification criteria in less than one year from Raynaud’s Phenomenon onset (Early Onset Subset–EOS), and all the others (Long Standing Subset–LSS). We show that EGFL7 expression is increased in EOS dcSSc skin and cultured FBs. EGFL7 is inducible by TGF-β on Healthy Controls (HC) FBs but not in SSc-FBs. EGFL7 decreases COL1A1 expression in EOS SSc-FBs while EGFL7 silencing up-regulates COL1A1 expression. EGFL7 promotes migration/invasion of EOS SSc-FBs but not proliferation. Finally, SSc-FBs, partially inhibit angiogenesis in organotypic coculture assays, and this is reversed by treatment with human recombinant (rh)EGFL7. We conclude that EGFL7 and its specific microRNA miR-126 may be involved in the pathogenesis of SSc vasculopathy and fibrosis.
Collapse
|
52
|
Sharma A, Lacko LA, Argueta LB, Glendinning MD, Stuhlmann H. miR-126 regulates glycogen trophoblast proliferation and DNA methylation in the murine placenta. Dev Biol 2019; 449:21-34. [PMID: 30771304 DOI: 10.1016/j.ydbio.2019.01.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 01/18/2019] [Accepted: 01/18/2019] [Indexed: 12/21/2022]
Abstract
A functional placenta develops through a delicate interplay of its vascular and trophoblast compartments. We have identified a previously unknown expression domain for the endothelial-specific microRNA miR-126 in trophoblasts of murine and human placentas. Here, we determine the role of miR-126 in placental development using a mouse model with a targeted deletion of miR-126. In addition to vascular defects observed only in the embryo, loss of miR-126 function in the placenta leads to junctional zone hyperplasia at E15.5 at the expense of the labyrinth, reduced placental volume for nutrient exchange and intra-uterine growth restriction of the embryos. Junctional zone hyperplasia results from increased numbers of proliferating glycogen trophoblast (GlyT) progenitors at E13.5 that give rise to an expanded glycogen trophoblast population at E15.5. Transcriptomic profile of miR-126-/- placentas revealed dysregulation of a large number of GlyT (Prl6a1, Prl7c1, Pcdh12) and trophoblast-specific genes (Tpbpa, Tpbpb, Prld1) and genes with known roles in placental development. We show that miR-126-/- placentas, but not miR-126-/- embryos, display aberrant expression of imprinted genes with important roles in glycogen trophoblasts and junctional zone development, including Igf2, H19, Cdkn1c and Phlda2, during mid-gestation. We also show that miR126-/- placentas display global hypermethylation, including at several imprint control centers. Our findings uncover a novel role for miR-126 in regulating extra-embryonic energy stores, expression of imprinted genes and DNA methylation in the placenta.
Collapse
Affiliation(s)
- Abhijeet Sharma
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, United States
| | - Lauretta A Lacko
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, United States; Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, United States
| | - Lissenya B Argueta
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, United States
| | - Michael D Glendinning
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, United States
| | - Heidi Stuhlmann
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, United States.
| |
Collapse
|
53
|
Zhou Q, Yu B, Anderson C, Huang ZP, Hanus J, Zhang W, Han Y, Bhattacharjee PS, Srinivasan S, Zhang K, Wang DZ, Wang S. LncEGFL7OS regulates human angiogenesis by interacting with MAX at the EGFL7/miR-126 locus. eLife 2019; 8:e40470. [PMID: 30741632 PMCID: PMC6370342 DOI: 10.7554/elife.40470] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/11/2019] [Indexed: 01/02/2023] Open
Abstract
In an effort to identify human endothelial cell (EC)-enriched lncRNAs,~500 lncRNAs were shown to be highly restricted in primary human ECs. Among them, lncEGFL7OS, located in the opposite strand of the EGFL7/miR-126 gene, is regulated by ETS factors through a bidirectional promoter in ECs. It is enriched in highly vascularized human tissues, and upregulated in the hearts of dilated cardiomyopathy patients. LncEGFL7OS silencing impairs angiogenesis as shown by EC/fibroblast co-culture, in vitro/in vivo and ex vivo human choroid sprouting angiogenesis assays, while lncEGFL7OS overexpression has the opposite function. Mechanistically, lncEGFL7OS is required for MAPK and AKT pathway activation by regulating EGFL7/miR-126 expression. MAX protein was identified as a lncEGFL7OS-interacting protein that functions to regulate histone acetylation in the EGFL7/miR-126 promoter/enhancer. CRISPR-mediated targeting of EGLF7/miR-126/lncEGFL7OS locus inhibits angiogenesis, inciting therapeutic potential of targeting this locus. Our study establishes lncEGFL7OS as a human/primate-specific EC-restricted lncRNA critical for human angiogenesis.
Collapse
Affiliation(s)
- Qinbo Zhou
- Department of Cell and Molecular BiologyTulane UniversityNew OrleansUnited States
| | - Bo Yu
- Department of Cell and Molecular BiologyTulane UniversityNew OrleansUnited States
| | - Chastain Anderson
- Department of Cell and Molecular BiologyTulane UniversityNew OrleansUnited States
| | - Zhan-Peng Huang
- Department of CardiologyBoston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Jakub Hanus
- Department of Cell and Molecular BiologyTulane UniversityNew OrleansUnited States
| | - Wensheng Zhang
- Department of Computer ScienceXavier UniversityNew OrleansUnited States
| | - Yu Han
- Aab Cardiovascular Research InstituteUniversity of Rochester School of Medicine and DentistryRochesterUnited States
| | | | - Sathish Srinivasan
- Cardiovascular Biology Research ProgramOklahoma Medical Research FoundationOklahomaUnited States
| | - Kun Zhang
- Department of Computer ScienceXavier UniversityNew OrleansUnited States
| | - Da-zhi Wang
- Department of CardiologyBoston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Shusheng Wang
- Department of Cell and Molecular BiologyTulane UniversityNew OrleansUnited States
- Department of OphthalmologyTulane UniversityNew OrleansUnited States
| |
Collapse
|
54
|
Jones Buie JN, Zhou Y, Goodwin AJ, Cook JA, Vournakis J, Demcheva M, Broome AM, Dixit S, Halushka PV, Fan H. Application of Deacetylated Poly-N-Acetyl Glucosamine Nanoparticles for the Delivery of miR-126 for the Treatment of Cecal Ligation and Puncture-Induced Sepsis. Inflammation 2019; 42:170-184. [PMID: 30244405 PMCID: PMC6380957 DOI: 10.1007/s10753-018-0882-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sepsis is an acute inflammatory syndrome in response to infection. In some cases, excessive inflammation from sepsis results in endothelial dysfunction and subsequent increased vascular permeability leading to organ failure. We previously showed that treatment with endothelial progenitor cells, which highly express microRNA-126 (miR-126), improved survival in mice subjected to cecal ligation and puncture (CLP) sepsis. miRNAs are important regulators of gene expression and cell function, play a major role in endothelial homeostasis, and may represent an emerging therapeutic modality. However, delivery of miRNAs to cells in vitro and in vivo is challenging due to rapid degradation by ubiquitous RNases. Herein, we developed a nanoparticle delivery system separately combining deacetylated poly-N-acetyl glucosamine (DEAC-pGlcNAc) polymers with miRNA-126-3p and miRNA-126-5p and testing these combinations in vitro and in vivo. Our results demonstrate that DEAC-pGlcNAc polymers have an appropriate size and zeta potential for cellular uptake and when complexed, DEAC-pGlcNAc protects miRNA from RNase A degradation. Further, DEAC-pGlcNAc efficiently encapsulates miRNAs as evidenced by preventing their migration in an agarose gel. The DEAC-pGlcNAc-miRNA complexes were taken up by multiple cell types and the delivered miRNAs had biological effects on their targets in vitro including pERK and DLK-1. In addition, we found that delivery of DEAC-pGlcNAc alone or DEAC-pGlcNAc:miRNA-126-5p nanoparticles to septic animals significantly improved survival, preserved vascular integrity, and modulated cytokine production. These composite studies support the concept that DEAC-pGlcNAc nanoparticles are an effective platform for delivering miRNAs and that they may provide therapeutic benefit in sepsis.
Collapse
Affiliation(s)
- Joy N Jones Buie
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 173 Ashley Ave., MSC 908, CRI Room 610, Charleston, SC, 29425, USA
| | - Yue Zhou
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 173 Ashley Ave., MSC 908, CRI Room 610, Charleston, SC, 29425, USA
- Department of Biopharmaceutics College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Andrew J Goodwin
- Department of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - James A Cook
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - John Vournakis
- Marine Polymer Technologies, Inc., Burlington, MA, 01803, USA
| | - Marina Demcheva
- Marine Polymer Technologies, Inc., Burlington, MA, 01803, USA
| | - Ann-Marie Broome
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Suraj Dixit
- Department of Cell & Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Perry V Halushka
- Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Pharmacology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 173 Ashley Ave., MSC 908, CRI Room 610, Charleston, SC, 29425, USA.
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
55
|
Fernández-Hernando C, Suárez Y. MicroRNAs in endothelial cell homeostasis and vascular disease. Curr Opin Hematol 2019; 25:227-236. [PMID: 29547400 DOI: 10.1097/moh.0000000000000424] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Since the first discovery of microRNAs (miRNAs) in 1993, the involvement of miRNAs in different aspects of vascular disease has emerged as an important research field. In this review, we summarize the fundamental roles of miRNAs in controlling endothelial cell functions and their implication with several aspects of vascular dysfunction. RECENT FINDINGS MiRNAs have been found to be critical modulators of endothelial homeostasis. The dysregulation of miRNAs has been linked to endothelial dysfunction and the development and progression of vascular disease which and open new opportunities of using miRNAs as potential therapeutic targets for vascular disease. SUMMARY Further determination of miRNA regulatory circuits and defining miRNAs-specific target genes remains key to future miRNA-based therapeutic applications toward vascular disease prevention. Many new and unanticipated roles of miRNAs in the control of endothelial functions will assist clinicians and researchers in developing potential therapeutic applications.
Collapse
Affiliation(s)
- Carlos Fernández-Hernando
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Department of Pathology and the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
56
|
Kontomanolis EN, Fasoulakis Z, Papamanolis V, Koliantzaki S, Dimopoulos G, Kambas NJ. The Impact of microRNAs in Breast Cancer Angiogenesis and Progression. Microrna 2019; 8:101-109. [PMID: 30332982 DOI: 10.2174/2211536607666181017122921] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/22/2018] [Accepted: 10/11/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE The study aims to review the recent data considering the expression profile and the role of microRNAs in breast tumorigenesis, and their impact on -the vital for breast cancer progression- angiogenesis. METHODS PubMed was searched for studies focused on data that associate microRNA with breast cancer, using the terms ''breast", "mammary gland", "neoplasia'', "angiogenesis" and ''microRNA'' between 1997-2018. RESULTS Aberrant expression of several circulating and tissue miRNAs is observed in human breast neoplasms with the deregulation of several miRNAs having a major participation in breast cancer progression. Angiogenesis seems to be directly affected by either overexpression or down regulation of many miRNAs, defining the overall prognostic rates. Many miRNAs differentially expressed in breast cancer that reveal a key role in suppression - progression and metastasis of breast cancer along with the contribution of the EGF, TNF-a and EGF cytokines. Conclusion Angiogenesis has proven to be vital for tumor development and metastasis while microRNAs are proposed to have multiple biological roles, including participation in immunosuppressive, immunomodulatory and recent studies reveal their implication in angiogenesis and its possible use as prognostic factors in cancer Even though larger studies are needed in order to reach safe conclusions, important steps are made that reveal the connection of serum microRNA expression to the angiogenic course of breast cancer, while miRNAs could be potential prognostic factors for the different breast cancer types.
Collapse
Affiliation(s)
- Emmanuel N. Kontomanolis
- Department of Obstetrics & Gynecology, Democritus University in Alexandroupolis, Dragana, Greece
| | - Zacharias Fasoulakis
- Department of Obstetrics & Gynecology, Democritus University in Alexandroupolis, Dragana, Greece
| | | | - Sofia Koliantzaki
- Department of Obstetrics & Gynecology, General Hospital of Korinthos, Corinth, Greece
| | - Georgios Dimopoulos
- Department of Obstetrics & Gynecology, General Hospital of Korinthos, Corinth, Greece
| | - Nikolaos J. Kambas
- Department of Obstetrics & Gynecology, General Hospital of Korinthos, Corinth, Greece
| |
Collapse
|
57
|
Villain G, Lelievre E, Broekelmann T, Gayet O, Havet C, Werkmeister E, Mecham R, Dusetti N, Soncin F, Mattot V. MAGP
‐1 and fibronectin control
EGFL
7 functions by driving its deposition into distinct endothelial extracellular matrix locations. FEBS J 2018; 285:4394-4412. [DOI: 10.1111/febs.14680] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/31/2018] [Accepted: 10/16/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Gaëlle Villain
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| | - Etienne Lelievre
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| | - Tom Broekelmann
- Department of Cell Biology and Physiology Washington University School of Medicine St. Louis MO USA
| | - Odile Gayet
- Centre de Recherche en Cancérologie de Marseille (CRCM) INSERM U1068 CNRS UMR 7258 Aix‐Marseille Université and Institut Paoli‐Calmettes, Parc Scientifique et Technologique de Luminy France
| | - Chantal Havet
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| | - Elisabeth Werkmeister
- Cellular Microbiology and Physics of Infection Group – Center for Infection and Immunity of Lille CNRS UMR8204 Inserm U1019 CHU Lille Institut Pasteur de Lille Univ. Lille. France
| | - Robert Mecham
- Department of Cell Biology and Physiology Washington University School of Medicine St. Louis MO USA
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM) INSERM U1068 CNRS UMR 7258 Aix‐Marseille Université and Institut Paoli‐Calmettes, Parc Scientifique et Technologique de Luminy France
| | - Fabrice Soncin
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| | - Virginie Mattot
- CNRS Institut Pasteur de Lille UMR 8161 – M3T – Mechanisms of Tumorigenesis and Target Therapies Univ. Lille France
| |
Collapse
|
58
|
Teijeiro V, Yang D, Majumdar S, González F, Rickert RW, Xu C, Koche R, Verma N, Lai EC, Huangfu D. DICER1 Is Essential for Self-Renewal of Human Embryonic Stem Cells. Stem Cell Reports 2018; 11:616-625. [PMID: 30146489 PMCID: PMC6135725 DOI: 10.1016/j.stemcr.2018.07.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 07/27/2018] [Accepted: 07/27/2018] [Indexed: 02/01/2023] Open
Abstract
MicroRNAs (miRNAs) are the effectors of a conserved gene-silencing system with broad roles in post-transcriptional regulation. Due to functional overlaps, assigning specific functions to individual miRNAs has been challenging. DICER1 cleaves pre-miRNA hairpins into mature miRNAs, and previously Dicer1 knockout mouse embryonic stem cells have been generated to study miRNA function in early mouse development. Here we report an essential requirement of DICER1 for the self-renewal of human embryonic stem cells (hESCs). Utilizing a conditional knockout approach, we found that DICER1 deletion led to increased death receptor-mediated apoptosis and failure of hESC self-renewal. We further devised a targeted miRNA screening strategy and uncovered essential pro-survival roles of members of the mir-302-367 and mir-371-373 clusters that bear the seed sequence AAGUGC. This platform is uniquely suitable for dissecting the roles of individual miRNAs in hESC self-renewal and differentiation, which may help us better understand the early development of human embryos. Conditional DICER1 knockouts enable the study of its essential requirement in hESCs DICER1 deletion leads to increased extrinsic (receptor-mediated) apoptosis miRNAs rescue the DICER1 knockout apoptotic phenotype This work provides a platform for interrogating miRNA function in hESCs
Collapse
Affiliation(s)
- Virginia Teijeiro
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10065, USA
| | - Dapeng Yang
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Sonali Majumdar
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Federico González
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Robert W Rickert
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Chunlong Xu
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Richard Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Nipun Verma
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Tri-Institutional M.D.-Ph.D. Program, 1300 York Avenue, New York, NY 10065, USA
| | - Eric C Lai
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Danwei Huangfu
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
59
|
Zhang L, Zhang Y, Zhao Y, Wang Y, Ding H, Xue S, Li P. Circulating miRNAs as biomarkers for early diagnosis of coronary artery disease. Expert Opin Ther Pat 2018; 28:591-601. [PMID: 30064285 DOI: 10.1080/13543776.2018.1503650] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Coronary artery disease (CAD) contributes to a huge number of human death worldwide. The early diagnosis can arrest the development of CAD and effectively lower the mortality rate. Recently, circulating miRNAs emerged as CAD biomarkers. AREAS COVERED Many efforts were paid to explore early diagnostic biomarkers of CAD. Some proteins have been used as diagnostic golden standard. However, the diagnostic and prognostic value of them is limited. MicroRNAs (miRNAs), a class of small noncoding RNAs, have been illustrated to regulate gene expression. The dysfunction of miRNAs is associated with CAD. MiRNAs presenting stably in body fluids are called circulating miRNAs. The altered expression of specific circulating miRNAs has been discovered in CAD and reported to affect the pathogenesis of CAD. We reviewed the recent data about circulating miRNAs regarding their potential roles in diagnosis, prognosis and therapeutic strategies for CAD. Additionally, we also summarized the current knowledge about circulating miRNA formation and detection. EXPERT OPINION Compared with traditional diagnostic tools, circulating miRNAs have many strongpoints, suggesting that circulating miRNAs can serve as promising biomarkers for the early diagnosis and prognosis of CAD.
Collapse
Affiliation(s)
- Lei Zhang
- a Institute for Translational Medicine, Qingdao University , Qingdao , China
| | - Yuan Zhang
- a Institute for Translational Medicine, Qingdao University , Qingdao , China
| | - Yanfang Zhao
- a Institute for Translational Medicine, Qingdao University , Qingdao , China
| | - Yu Wang
- a Institute for Translational Medicine, Qingdao University , Qingdao , China
| | - Han Ding
- a Institute for Translational Medicine, Qingdao University , Qingdao , China
| | - Sheng Xue
- a Institute for Translational Medicine, Qingdao University , Qingdao , China
| | - Peifeng Li
- a Institute for Translational Medicine, Qingdao University , Qingdao , China
| |
Collapse
|
60
|
Endothelial Cell Aging: How miRNAs Contribute? J Clin Med 2018; 7:jcm7070170. [PMID: 29996516 PMCID: PMC6068727 DOI: 10.3390/jcm7070170] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
Endothelial cells (ECs) form monolayers and line the interior surfaces of blood vessels in the entire body. In most mammalian systems, the capacity of endothelial cells to divide is limited and endothelial cells are prone to be senescent. Aging of ECs and resultant endothelial dysfunction lead to a variety of vascular diseases such as atherosclerosis, diabetes mellites, hypertension, and ischemic injury. However, the mechanism by which ECs get old and become senescent and the impact of endothelial senescence on the vascular function are not fully understood. Recent research has unveiled the crucial roles of miRNAs, which are small non-coding RNAs, in regulating endothelial cellular functions, including nitric oxide production, vascular inflammation, and anti-thromboformation. In this review, how senescent-related miRNAs are involved in controlling the functions of ECs will be discussed.
Collapse
|
61
|
Rouigari M, Dehbashi M, Ghaedi K, Pourhossein M. Targetome Analysis Revealed Involvement of MiR-126 in Neurotrophin Signaling Pathway: A Possible Role in Prevention of Glioma Development. CELL JOURNAL 2018; 20:150-156. [PMID: 29633591 PMCID: PMC5893285 DOI: 10.22074/cellj.2018.4901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/14/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVES For the first time, we used molecular signaling pathway enrichment analysis to determine possible involvement of miR-126 and IRS-1 in neurotrophin pathway. MATERIALS AND METHODS In this prospective study, Validated and predicted targets (targetome) of miR-126 were collected following searching miRtarbase (http://mirtarbase.mbc.nctu.edu.tw/) and miRWalk 2.0 databases, respectively. Then, approximate expression of miR-126 targeting in Glioma tissue was examined using UniGene database (http://www.ncbi. nlm.nih.gov/unigene). In silico molecular pathway enrichment analysis was carried out by DAVID 6.7 database (http://david. abcc.ncifcrf.gov/) to explore which signaling pathway is related to miR-126 targeting and how miR-126 attributes to glioma development. RESULTS MiR-126 exerts a variety of functions in cancer pathogenesis via suppression of expression of target gene including PI3K, KRAS, EGFL7, IRS-1 and VEGF. Our bioinformatic studies implementing DAVID database, showed the involvement of miR-126 target genes in several signaling pathways including cancer pathogenesis, neurotrophin functions, Glioma formation, insulin function, focal adhesion production, chemokine synthesis and secretion and regulation of the actin cytoskeleton. CONCLUSIONS Taken together, we concluded that miR-126 enhances the formation of glioma cancer stem cell probably via down regulation of IRS-1 in neurotrophin signaling pathway.
Collapse
Affiliation(s)
- Maedeh Rouigari
- Isfahan Neuroscience Research Center (INRC), Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Moein Dehbashi
- Isfahan Neuroscience Research Center (INRC), Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
- Genetics Division, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Kamran Ghaedi
- Cell and Molecular Biology Division, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Meraj Pourhossein
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences Isfahan, Iran
- Department of Food Science and Technology, Food Security Research Center, School of Nutrition and Food Science, Isfahan, Iran.
| |
Collapse
|
62
|
Hong G, Kuek V, Shi J, Zhou L, Han X, He W, Tickner J, Qiu H, Wei Q, Xu J. EGFL7: Master regulator of cancer pathogenesis, angiogenesis and an emerging mediator of bone homeostasis. J Cell Physiol 2018; 233:8526-8537. [PMID: 29923200 DOI: 10.1002/jcp.26792] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/30/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Guoju Hong
- National Key Discipline and Orthopedic Laboratory Guangzhou University of Chinese Medicine Guangzhou Guangdong China
- Division of Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia Perth WA Australia
| | - Vincent Kuek
- Division of Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia Perth WA Australia
| | - Jiaxi Shi
- First Clinical College Guangzhou University of Chinese Medicine Guangzhou Guangdong China
| | - Lin Zhou
- Department of Rheumatology The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Xiaorui Han
- Department of Radiography Guangzhou First People's Hospital The Second Affiliated Hospital of South China University of Technology Guangzhou Guangdong China
| | - Wei He
- National Key Discipline and Orthopedic Laboratory Guangzhou University of Chinese Medicine Guangzhou Guangdong China
- Orthopedic Department The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou Guangdong China
| | - Jennifer Tickner
- Division of Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia Perth WA Australia
| | - Heng Qiu
- Division of Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia Perth WA Australia
| | - Qiushi Wei
- National Key Discipline and Orthopedic Laboratory Guangzhou University of Chinese Medicine Guangzhou Guangdong China
- Orthopedic Department The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou Guangdong China
| | - Jiake Xu
- National Key Discipline and Orthopedic Laboratory Guangzhou University of Chinese Medicine Guangzhou Guangdong China
- Division of Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia Perth WA Australia
| |
Collapse
|
63
|
Development of the renal vasculature. Semin Cell Dev Biol 2018; 91:132-146. [PMID: 29879472 DOI: 10.1016/j.semcdb.2018.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022]
Abstract
The kidney vasculature has a unique and complex architecture that is central for the kidney to exert its multiple and essential physiological functions with the ultimate goal of maintaining homeostasis. An appropriate development and coordinated assembly of the different vascular cell types and their association with the corresponding nephrons is crucial for the generation of a functioning kidney. In this review we provide an overview of the renal vascular anatomy, histology, and current knowledge of the embryological origin and molecular pathways involved in its development. Understanding the cellular and molecular mechanisms involved in renal vascular development is the first step to advance the field of regenerative medicine.
Collapse
|
64
|
Yang Y, Pan JJ, Zhou XG, Zhou XY, Cheng R. Differentially expressed miRNAs in premature infants with retinopathy-a bioinformatics analysis. Int J Ophthalmol 2018; 11:773-779. [PMID: 29862174 DOI: 10.18240/ijo.2018.05.09] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 01/29/2018] [Indexed: 12/16/2022] Open
Abstract
AIM To reveal the role of miRNAs in retinopathy of prematurity (ROP) by bioinformatics analysis. METHODS The raw data of this study came from the researches of Wang et al and Zhao et al who analyzed the microRNA (miRNA) expression profile between ROP and controls. Based on the identified differentially expressed miRNAs, the related target genes, lncRNA and circRNA were predicted. Then we performed functional enrichment analysis to further analyze the functions of target genes. RESULTS Hsa-miRNA-128-3p and hsa-miRNA-9-5p showed significantly different expression in both studies. LncRNA of POLDIP2, GAS5, NEFL and UHRF1, circRNA of ZNF280C_hsa_circ_001211 and SIAE_hsa_circ_002083, tar-get gene of QKI showed meaningful differential expression in ROP. Enrichment analysis showed that TGF-β signaling pathway, PI3K-Akt signaling pathway and MAPK signaling pathway might play important roles in the prog-ress of ROP. CONCLUSION This research may provide a comprehensive bioinformatics analysis of differentially expressed miRNAs which are possibly involved in ROP.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neonatology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Jing-Jing Pan
- Department of Neonatology, Jiangsu Provincial People's Hospital Affiliated to Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Xiao-Guang Zhou
- Department of Neonatology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Xiao-Yu Zhou
- Department of Neonatology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Rui Cheng
- Department of Neonatology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
65
|
Teoh JP, Bayoumi AS, Aonuma T, Xu Y, Johnson JA, Su H, Weintraub NL, Tang Y, Kim IM. β-arrestin-biased agonism of β-adrenergic receptor regulates Dicer-mediated microRNA maturation to promote cardioprotective signaling. J Mol Cell Cardiol 2018; 118:225-236. [PMID: 29627294 DOI: 10.1016/j.yjmcc.2018.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/28/2018] [Accepted: 04/02/2018] [Indexed: 12/20/2022]
Abstract
RATIONALE MicroRNAs (miRs) are small, non-coding RNAs that function to post-transcriptionally regulate target genes. First transcribed as primary miR transcripts (pri-miRs), they are enzymatically processed by Drosha into premature miRs (pre-miRs) and further cleaved by Dicer into mature miRs. Initially discovered to desensitize β-adrenergic receptor (βAR) signaling, β-arrestins are now well-appreciated to modulate multiple pathways independent of G protein signaling, a concept known as biased signaling. Using the β-arrestin-biased βAR ligand carvedilol, we previously showed that β-arrestin1 (not β-arrestin2)-biased β1AR (not β2AR) cardioprotective signaling stimulates Drosha-mediated processing of six miRs by forming a multi-protein nuclear complex, which includes β-arrestin1, the Drosha microprocessor complex and a single-stranded RNA binding protein hnRNPA1. OBJECTIVE Here, we investigate whether β-arrestin-mediated βAR signaling induced by carvedilol could regulate Dicer-mediated miR maturation in the cytoplasm and whether this novel mechanism promotes cardioprotective signaling. METHODS AND RESULTS In mouse hearts, carvedilol indeed upregulates three mature miRs, but not their pre-miRs and pri-miRs, in a β-arrestin 1- or 2-dependent manner. Interestingly, carvedilol-mediated activation of miR-466g or miR-532-5p, and miR-674 is dependent on β2ARs and β1ARs, respectively. Mechanistically, β-arrestin 1 or 2 regulates maturation of three newly identified βAR/β-arrestin-responsive miRs (β-miRs) by associating with the Dicer maturation RNase III enzyme on three pre-miRs of β-miRs. Myocardial cell approaches uncover that despite their distinct roles in different cell types, β-miRs act as gatekeepers of cardiac cell functions by repressing deleterious targets. CONCLUSIONS Our findings indicate a novel role for βAR-mediated β-arrestin signaling activated by carvedilol in Dicer-mediated miR maturation, which may be linked to its protective mechanisms.
Collapse
Affiliation(s)
- Jian-Peng Teoh
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Ahmed S Bayoumi
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Tatsuya Aonuma
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Yanyan Xu
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - John A Johnson
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912, USA
| | - Huabo Su
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Medicine, Augusta University, Augusta, GA 30912, USA
| | - Yaoliang Tang
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Medicine, Augusta University, Augusta, GA 30912, USA
| | - Il-Man Kim
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
66
|
Abstract
MicroRNAs (miRNAs) are ∼22 nt RNAs that direct posttranscriptional repression of mRNA targets in diverse eukaryotic lineages. In humans and other mammals, these small RNAs help sculpt the expression of most mRNAs. This article reviews advances in our understanding of the defining features of metazoan miRNAs and their biogenesis, genomics, and evolution. It then reviews how metazoan miRNAs are regulated, how they recognize and cause repression of their targets, and the biological functions of this repression, with a compilation of knockout phenotypes that shows that important biological functions have been identified for most of the broadly conserved miRNAs of mammals.
Collapse
Affiliation(s)
- David P Bartel
- Howard Hughes Medical Institute and Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
67
|
Mir-126 is a conserved modulator of lymphatic development. Dev Biol 2018; 437:120-130. [PMID: 29550364 DOI: 10.1016/j.ydbio.2018.03.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 03/05/2018] [Indexed: 12/20/2022]
Abstract
Organ homeostasis relies upon cellular and molecular processes that restore tissue structure and function in a timely fashion. Lymphatic vessels help maintain fluid equilibrium by returning interstitial fluid that evades venous uptake back to the circulation. Despite its important role in tissue homeostasis, cancer metastasis, and close developmental origins with the blood vasculature, the number of molecular players known to control lymphatic system development is relatively low. Here we show, using genetic approaches in zebrafish and mice, that the endothelial specific microRNA mir-126, previously implicated in vascular integrity, regulates lymphatic development. In zebrafish, in contrast to mir-126 morphants, double mutants (mir-126a-/-; mir-126b-/-, hereafter mir-126-/-) do not exhibit defects in vascular integrity but develop lymphatic hypoplasia; mir-126-/- animals fail to develop complete trunk and facial lymphatics, display severe edema and die as larvae. Notably, following MIR-126 inhibition, human Lymphatic Endothelial Cells (hLECs) respond poorly to VEGFA and VEGFC. In this context, we identify a concomitant reduction in Vascular Endothelial Growth Factor Receptor-2 (VEGFR2) and Vascular Endothelial Growth Factor Receptor-3 (VEGFR3, also known as FLT4) expression upon MIR-126 inhibition. In vivo, we further show that flt4+/- zebrafish embryos exhibit lymphatic defects after mild miR-126 knockdown. Similarly, loss of Mir-126 in Flt4+/- mice results in embryonic edema and lethality. Thus, our results indicate that miR-126 modulation of Vegfr signaling is essential for lymphatic system development in fish and mammals.
Collapse
|
68
|
Zhang B, Nguyen LXT, Li L, Zhao D, Kumar B, Wu H, Lin A, Pellicano F, Hopcroft L, Su YL, Copland M, Holyoake TL, Kuo CJ, Bhatia R, Snyder DS, Ali H, Stein AS, Brewer C, Wang H, McDonald T, Swiderski P, Troadec E, Chen CC, Dorrance A, Pullarkat V, Yuan YC, Perrotti D, Carlesso N, Forman SJ, Kortylewski M, Kuo YH, Marcucci G. Bone marrow niche trafficking of miR-126 controls the self-renewal of leukemia stem cells in chronic myelogenous leukemia. Nat Med 2018; 24:450-462. [PMID: 29505034 PMCID: PMC5965294 DOI: 10.1038/nm.4499] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/22/2018] [Indexed: 12/16/2022]
Abstract
Leukemia stem cells (LSCs) in individuals with chronic myelogenous leukemia (CML) (hereafter referred to as CML LSCs) are responsible for initiating and maintaining clonal hematopoiesis. These cells persist in the bone marrow (BM) despite effective inhibition of BCR-ABL kinase activity by tyrosine kinase inhibitors (TKIs). Here we show that although the microRNA (miRNA) miR-126 supported the quiescence, self-renewal and engraftment capacity of CML LSCs, miR-126 levels were lower in CML LSCs than in long-term hematopoietic stem cells (LT-HSCs) from healthy individuals. Downregulation of miR-126 levels in CML LSCs was due to phosphorylation of Sprouty-related EVH1-domain-containing 1 (SPRED1) by BCR-ABL, which led to inhibition of the RAN-exportin-5-RCC1 complex that mediates miRNA maturation. Endothelial cells (ECs) in the BM supply miR-126 to CML LSCs to support quiescence and leukemia growth, as shown using mouse models of CML in which Mir126a (encoding miR-126) was conditionally knocked out in ECs and/or LSCs. Inhibition of BCR-ABL by TKI treatment caused an undesired increase in endogenous miR-126 levels, which enhanced LSC quiescence and persistence. Mir126a knockout in LSCs and/or ECs, or treatment with a miR-126 inhibitor that targets miR-126 expression in both LSCs and ECs, enhanced the in vivo anti-leukemic effects of TKI treatment and strongly diminished LSC leukemia-initiating capacity, providing a new strategy for the elimination of LSCs in individuals with CML.
Collapse
Affiliation(s)
- Bin Zhang
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Le Xuan Truong Nguyen
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ling Li
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Dandan Zhao
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Bijender Kumar
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Herman Wu
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Allen Lin
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Francesca Pellicano
- Paul O' Gorman Leukemia Research Centre, College of Medical, Veterinary and Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Lisa Hopcroft
- Paul O' Gorman Leukemia Research Centre, College of Medical, Veterinary and Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Yu-Lin Su
- Department of Immuno-oncology, City of Hope Medical Center, Duarte, California, USA
| | - Mhairi Copland
- Paul O' Gorman Leukemia Research Centre, College of Medical, Veterinary and Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Tessa L Holyoake
- Paul O' Gorman Leukemia Research Centre, College of Medical, Veterinary and Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Calvin J Kuo
- Stanford University School of Medicine, Stanford, California, USA
| | - Ravi Bhatia
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David S Snyder
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Haris Ali
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Anthony S Stein
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Casey Brewer
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Huafeng Wang
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA.,Department of Hematology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tinisha McDonald
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Piotr Swiderski
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Estelle Troadec
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Ching-Cheng Chen
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Adrienne Dorrance
- Division of Hematology, Department of Internal Medicine, Ohio State University, Columbus, Ohio, USA
| | - Vinod Pullarkat
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Yate-Ching Yuan
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Danilo Perrotti
- Department of Medicine, Biochemistry and Molecular Biology and the Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine Baltimore, Baltimore, Maryland, USA.,Deparment of Hematology, Hammersmith Hospital, Imperial College London, London, UK
| | - Nadia Carlesso
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Stephen J Forman
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Marcin Kortylewski
- Department of Immuno-oncology, City of Hope Medical Center, Duarte, California, USA
| | - Ya-Huei Kuo
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California, USA
| |
Collapse
|
69
|
Expression, regulation and function of miR-126 in the mouse choroid vasculature. Exp Eye Res 2018; 170:169-176. [PMID: 29501382 PMCID: PMC5993209 DOI: 10.1016/j.exer.2018.02.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/20/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022]
Abstract
MicroRNA miR-126 has been shown to be required for proper angiogenesis in several models. However, its expression, regulation and function in the mouse choroid remain unclear. Our previous data has shown that miR-126 expression is enriched in the endothelial cells (ECs) of the mouse choroid. Here we report that a 5.5 kb Egfl7/miR-126 promoter drives the expression of miR-126 in the choroid ECs during choroidal vascular development. The expression of miR-126 in the ECs is regulated by flow stress likely through Krüppel-like transcriptional factors. miR-126−/− mice show mildly delayed choroidal vascular development, but adult knockout mice develop periphery choroidal vascular lesions. This study suggests that miR-126 is largely dispensable for mouse choroidal development but required for maintaining choroidal vasculature integrity.
Collapse
|
70
|
Abstract
Extracellular matrix (ECM) proteins secreted by blood-brain barrier (BBB) endothelial cells (ECs) are implicated in cell trafficking. We discovered that the expression of ECM epidermal growth factor-like protein 7 (EGFL7) is increased in the CNS vasculature of patients with multiple sclerosis (MS), and in mice with experimental autoimmune encephalomyelitis (EAE). Perivascular CD4 T lymphocytes colocalize with ECM-bound EGFL7 in MS lesions. Human and mouse activated T cells upregulate EGFL7 ligand αvβ3 integrin and can adhere to EGFL7 through integrin αvβ3. EGFL7-knockout (KO) mice show earlier onset of EAE and increased brain and spinal cord parenchymal infiltration of T lymphocytes. Importantly, EC-restricted EGFL7-KO is associated with a similar EAE worsening. Finally, treatment with recombinant EGFL7 improves EAE, reduces MCAM expression, and tightens the BBB in mouse. Our data demonstrate that EGFL7 can limit CNS immune infiltration and may represent a novel therapeutic avenue in MS. Endothelial cells release extracellular matrix components that regulate inflammation. Here the authors demonstrate that the extracellular matrix component epidermal growth factor-like protein 7 regulates inflammation in experimental autoimmune encephalomyelitis in the mouse.
Collapse
|
71
|
Kovacova J, Poprach A, Buchler T, Cho WC, Slaby O. MicroRNAs as predictive biomarkers of response to tyrosine kinase inhibitor therapy in metastatic renal cell carcinoma. ACTA ACUST UNITED AC 2018; 56:1426-1431. [DOI: 10.1515/cclm-2017-0861] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/08/2018] [Indexed: 01/07/2023]
Abstract
Abstract
Renal cell carcinoma (RCC) accounts for 2%–3% of all malignant tumours. The first-choice treatment in metastatic RCC (mRCC) patients is tyrosine kinase inhibitors (TKIs). Although TKIs may prolong survival of the treated patients who are not primary resistant, almost all of them will eventually develop secondary resistance to the treatment after a progression-free period. To predict treatment response, thus, we need efficient biomarkers for rational indication of TKIs in mRCC. MicroRNAs (miRNAs) not only play important roles in the pathogenesis of many cancers, including RCC but also have been shown to serve as promising diagnostic, prognostic and predictive biomarkers in various cancers. However, the potential of miRNAs to predict response to therapy with TKIs in mRCC has not yet gained sufficient attention. Because personalisation of the TKIs indication in mRCC presents an important unmet medical need, we summarise research on this topic and give an overall insight on the current knowledge in this field.
Collapse
Affiliation(s)
- Julia Kovacova
- Central European Institute of Technology , Masaryk University , Brno , Czech Republic
| | - Alexandr Poprach
- Department of Comprehensive Cancer Care , Masaryk Memorial Cancer Institute , Brno , Czech Republic
| | - Tomas Buchler
- Department of Oncology , Thomayer Hospital and Charles University First Faculty of Medicine , Prague , Czech Republic
| | - William C. Cho
- Department of Clinical Oncology , Queen Elizabeth Hospital , Kowloon , Hong Kong
| | - Ondrej Slaby
- Central European Institute of Technology , Masaryk University , Brno , Czech Republic
- Department of Comprehensive Cancer Care , Masaryk Memorial Cancer Institute , Brno , Czech Republic
| |
Collapse
|
72
|
Luo J, Zhu C, Wang H, Yu L, Zhou J. MicroRNA-126 affects ovarian cancer cell differentiation and invasion by modulating expression of vascular endothelial growth factor. Oncol Lett 2018; 15:5803-5808. [PMID: 29552211 DOI: 10.3892/ol.2018.8025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 11/02/2017] [Indexed: 11/06/2022] Open
Abstract
Primary ovarian cancer is the main cause of gynecological cancer-associated mortality. However, the mechanism behind the spread of ovarian cancer requires elucidation. The present study aimed to investigate the effects of microRNA-126 (miR-126) on differentiation and invasion, and its mechanism in primary ovarian cancer. Ovarian cancer SKOV3 cells transfected with LV3-has-miR-126 mimics and LV3-has-miR-126 inhibitor were produced; it was revealedthatLV-miR-126 mimics could induce cell cycle arrest at G1 phase, suppress cell invasion through Matrigel-coated membranes and downregulate the expression of vascular endothelial growth factor (VEGF). Furthermore, LV-has-miR-126 inhibitor-transfected cells could increase the number of cells in S phase, induce cell invasion and upregulate the expression of VEGF. The present study, to the best of our knowledge, is the first to report that miR-126 may serve tumor suppressor roles by inducing G1 cell cycle arrest and suppressing invasion in ovarian cancer cells, at least in part by targeting VEGF expression.
Collapse
Affiliation(s)
- Jianqiao Luo
- Department of Gynecology, Shaoxing Shangyu Women and Children Hospital, Shaoxing, Zhejiang 312000, P.R. China.,Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Caidan Zhu
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Hongya Wang
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Li Yu
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
73
|
Xie H, Wang F, Chen X, Ying H. miR‑126 is essential for endothelial phenotype expression during endothelial differentiation in adipose‑derived stem cells. Mol Med Rep 2018; 17:442-446. [PMID: 29115445 DOI: 10.3892/mmr.2017.7915] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 02/27/2017] [Indexed: 11/05/2022] Open
Abstract
The endothelial differentiation of stem cells serves an essential role in vascular development, function and disease. Autograft adipose‑derived stem cells (ADSCs) may be a novel source of cell for use in the study of angiogenesis. microRNA‑126 (miR‑126) has been extensively studied in endothelial cells. However, the effect of miR‑126 on the endothelial differentiation of ADSCs has not been demonstrated. In the present study, it was observed that the expression of miR‑126 was markedly increased during the endothelial differentiation of ADSCs in a time‑dependent manner. The effect of miR‑126 on ADSC differentiation was confirmed by employing up‑ and down‑regulation strategies. The expression of endothelial markers was decreased by miR‑126 inhibitor transfection during endothelial differentiation. The results of the present study suggest that miR‑126 is essential for endothelial phenotype expression, and may promote the progress of endothelial differentiation in ADSCs, providing a novel strategy for modulating vascular formation and function.
Collapse
Affiliation(s)
- Han Xie
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, P.R. China
| | - Fang Wang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiao Chen
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, P.R. China
| | - Hao Ying
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, P.R. China
| |
Collapse
|
74
|
Natarelli L, Schober A. MicroRNAs and the response to injury in atherosclerosis. Hamostaseologie 2017; 35:142-50. [DOI: 10.5482/hamo-14-10-0051] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/13/2015] [Indexed: 11/05/2022] Open
Abstract
SummaryEndothelial cells (ECs) at arterial branching points are physiologically subjected to chronic damage by disturbed blood flow, which triggers a vascular wound healing response. Additional damage by hyperlipid -aemia perturbs this delicate balance of en-dothelial injury and regeneration, and the progressive accumulation of noxious modified lipoproteins leads to macrophage death. Several miRNAs such as miR-92a and miR-712, which modulate EC proliferation and inflammation, are up-regulated by disturbed flow in ECs, and contribute to atherosclerosis. In addition, reduced endothelial levels of miR-126–5p limit the regenerative capacity of ECs, which becomes apparent by insufficient endothelial repair under hyperlipidemic stress. In macrophages, miR-342–5p induces the expression of miR-155 during the progression of atherosclerosis, which promotes inflammatory gene expression and inhibits efferocytosis by targeting Bcl6, thus contributing to necrotic core formation. Deciphering the complex cell- and context-specific effects of miRNAs during vascular wound healing appears essential for the development of miRNA-based therapies of atherosclerosis.
Collapse
|
75
|
Neale JPH, Pearson JT, Katare R, Schwenke DO. Ghrelin, MicroRNAs, and Critical Limb Ischemia: Hungering for a Novel Treatment Option. Front Endocrinol (Lausanne) 2017; 8:350. [PMID: 29326658 PMCID: PMC5733488 DOI: 10.3389/fendo.2017.00350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/29/2017] [Indexed: 12/15/2022] Open
Abstract
Critical limb ischemia (CLI) is the most severe manifestation of peripheral artery disease. It is characterized by chronic pain at rest, skin ulcerations, and gangrene tissue loss. CLI is a highly morbid condition, resulting in a severely diminished quality of life and a significant risk of mortality. The primary goal of therapy for CLI is to restore blood flow to the affected limb, which is only possible by surgery, but is inadvisable in up to 50% of patients. This subset of patients who are not candidates for revascularisation are referred to as "no-option" patients and are the focus of investigation for novel therapeutic strategies. Angiogenesis, arteriogenesis and vasculogenesis are the processes whereby new blood vessel networks form from the pre-existing vasculature and primordial cells, respectively. In therapeutic angiogenesis, exogenous stimulants are administered to promote angiogenesis and augment limb perfusion, offering a potential treatment option for "no option" patients. However, to date, very few clinical trials of therapeutic angiogenesis in patients with CLI have reported clinically significant results, and it remains a major challenge. Ghrelin, a 28-amino acid peptide, is emerging as a potential novel therapeutic for CLI. In pre-clinical models, exogenous ghrelin has been shown to induce therapeutic angiogenesis, promote muscle regeneration, and reduce oxidative stress via the modulation of microRNAs (miRs). miRs are endogenous, small, non-coding ribonucleic acids of ~20-22 nucleotides which regulate gene expression at the post-transcriptional level by either translational inhibition or by messenger ribonucleic acid cleavage. This review focuses on the mounting evidence for the use of ghrelin as a novel therapeutic for CLI, and highlights the miRs which orchestrate these physiological events.
Collapse
Affiliation(s)
- Joshua P. H. Neale
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| | - James T. Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Rajesh Katare
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| | - Daryl O. Schwenke
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
76
|
Johnson MG, Konicke K, Kristianto J, Gustavson A, Garbo R, Wang X, Yuan B, Blank RD. Endothelin signaling regulates mineralization and posttranscriptionally regulates SOST in TMOb cells via miR 126-3p. Physiol Rep 2017; 5:5/4/e13088. [PMID: 28235973 PMCID: PMC5328763 DOI: 10.14814/phy2.13088] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 11/24/2022] Open
Abstract
Previously, our laboratory identified ECE‐1, encoding endothelin‐converting enzyme‐1 (ECE‐1), as a positional candidate for a pleiotropic quantitative trait locus affecting femoral size, shape, and biomechanical performance. We hypothesized that endothelin‐1 (ET‐1) signaling promotes osteogenesis. Exposure of immortalized mouse osteoblast (TMOb) cells to big ET‐1 increased mineralization. Following big ET‐1 treatment, we measured the secretion of insulin‐like‐growth factor‐1 (IGF1), dickkopf‐homolog‐1 protein 1 (DKK1), and sclerostin (SOST). In each case, big ET‐1 signaling changed secretion in a manner that favored increased osteogenic activity. Treatment with ECE‐1, endothelin receptor A (EDNRA), or WNT receptor antagonists inhibited the big ET‐1‐mediated increase in mineralization. In the presence of big ET‐1, message levels of Runx2, Igf1, Dkk1, and Sost are uncoupled from protein production, suggesting posttranscriptional regulation. To evaluate the role of big ET‐1 in normal bone physiology, we inhibited EDNRA signaling during mineralization in the absence of exogenous ET‐1. EDNRA blockade reduced mineralization, decreased IGF1, and increased DKK1 and SOST secretion, responses opposite to those induced by exogenous big ET‐1. Pharmacological and siRNA knockdown to inhibit ECE‐1 reduced mineralization and IGF1 secretion with decreasing DKK1 and decreasing or stable SOST secretion, suggesting a further, unknown role of ECE‐1 in osteoblast maturation. Previously we identified miR 126‐3p as a potential ET‐1‐responsive regulator of SOST in murine cells. Overexpression of miR126‐3p increased mineralization in TMOb cells and decreased SOST secretion. Osteoblasts express the ET‐1 signaling pathway and ET‐1 signaling is necessary for normal osteoblast differentiation and mineralization, acting through regulation of miRs that target osteogenic molecules.
Collapse
Affiliation(s)
- Michael G Johnson
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin .,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kathryn Konicke
- Medical Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin.,Division of Endocrinology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jasmin Kristianto
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Anne Gustavson
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rachel Garbo
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Xiaohu Wang
- Medical Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin.,Division of Endocrinology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Baozhi Yuan
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Robert D Blank
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Medical Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin.,Division of Endocrinology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
77
|
Lykoudi A, Kolialexi A, Lambrou GI, Braoudaki M, Siristatidis C, Papaioanou GK, Tzetis M, Mavrou A, Papantoniou N. Dysregulated placental microRNAs in Early and Late onset Preeclampsia. Placenta 2017; 61:24-32. [PMID: 29277268 DOI: 10.1016/j.placenta.2017.11.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 12/24/2022]
Abstract
INTRODUCTION To determine the miRNA expression profile in placentas complicated by Preeclampsia (PE) and compare it to uncomplicated pregnancies. METHODS Sixteen placentas from women with PE, [11 with early onset PE (EOPE) and 5 with late onset PE (LOPE)], as well as 8 placentas from uncomplicated pregnancies were analyzed using miRNA microarrays. For statistical analyses the MATLAB® simulation environment was applied. The over-expression of miR-518a-5p was verified using Quantitative Real-Time Polymerase Chain Reaction. RESULTS Forty four miRNAs were found dysregulated in PE complicated placentas. Statistical analysis revealed that miR-431, miR-518a-5p and miR-124* were over-expressed in EOPE complicated placentas as compared to controls, whereas miR-544 and miR-3942 were down-regulated in EOPE. When comparing the miRNA expression profile in cases with PE and PE-growth restricted fetuses (FGR), miR-431 and miR-518a-5p were found over-expressed in pregnancies complicated by FGR. DISCUSSION Since specific miRNAs can differentiate EOPE and LOPE from uncomplicated placentas, they may be considered as putative PE-specific biomarkers. MiR-518a-5p emerged as a potential diagnostic indicator for EOPE cases as well as for PE-FGR complicated placentas, indicating a potential link to the severity of the disease.
Collapse
Affiliation(s)
- Alexandra Lykoudi
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Athens, Greece; Department of Medical Genetics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Aggeliki Kolialexi
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Athens, Greece; Department of Medical Genetics, National and Kapodistrian University of Athens Medical School, Athens, Greece.
| | - George I Lambrou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Thivon & Levadeias, 11527, Athens, Greece
| | - Maria Braoudaki
- Department of Medical Genetics, National and Kapodistrian University of Athens Medical School, Athens, Greece; School of Life and Medical Sciences, University of Hertfordshire, Hatfield, AL10 9AB, United Kingdom
| | - Charalampos Siristatidis
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - George Konstantinos Papaioanou
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Maria Tzetis
- Department of Medical Genetics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ariadni Mavrou
- Department of Medical Genetics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Nikolas Papantoniou
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
78
|
MicroRNA-126a-5p enhances myocardial ischemia-reperfusion injury through suppressing Hspb8 expression. Oncotarget 2017; 8:94172-94187. [PMID: 29212219 PMCID: PMC5706865 DOI: 10.18632/oncotarget.21613] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/23/2017] [Indexed: 12/17/2022] Open
Abstract
Previously, we found several genes are involved in myocardial ischemia-reperfusion (M-I/R) injury. In this report, we first developed a mouse model of M-I/R injury and demonstrated microRNA-126a-5p was associated with the M-I/R injury by using high-throughput microRNA expression analysis. We further investigated the expression and function of microRNA-126a-5p during mouse M-I/R injury. We observed high expression of microRNA-126a-5p in the M-I/R mice and increased levels of LDH and CK-MB (damage markers) in the serum. H2O2 and hypoxia/reoxygenation (H/R) treatment significantly increased the expression of microRNA-126a-5p in H9C2 cells in concentration- and time-dependent manners. Moreover, microRNA-126a-5p overexpression in H9C2 cells inhibited cell viability but increased LDH release and caspase 3 activity. Cardiac function analysis based on the measurements of hemodynamic parameters showed that microRNA-126a-5p expression ablation in M-I/R injured mice led to the reversal of the symptoms caused by M-I/R injury. Transesophageal echocardiography also revealed that the values of LVIDd and LVIDs were decreased while the values of LVFS% and LVEF% were increased in M-I/R injured mice after treatment with microRNA-126a-5p inhibitor, compared with the M-I/R injured mice treated with the control. Bioinformatic analysis demonstrated that Hspb8, a protective protein in myocardium, was the target of microRNA-126a-5p. Thus, these findings indicated that microRNA-126a-5p was up-regulated in mouse M-I/R model and promoted M-I/R injury in vivo through suppressing the expression of Hspb8, which may shed light on the development of potential therapeutic target for M-I/R injury.
Collapse
|
79
|
Paikari A, D Belair C, Saw D, Blelloch R. The eutheria-specific miR-290 cluster modulates placental growth and maternal-fetal transport. Development 2017; 144:3731-3743. [PMID: 28935707 DOI: 10.1242/dev.151654] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
The vertebrate-specific ESCC microRNA family arises from two genetic loci in mammals: miR-290/miR-371 and miR-302. The miR-302 locus is found broadly among vertebrates, whereas the miR-290/miR-371 locus is unique to eutheria, suggesting a role in placental development. Here, we evaluate that role. A knock-in reporter for the mouse miR-290 cluster is expressed throughout the embryo until gastrulation, when it becomes specifically expressed in extraembryonic tissues and the germline. In the placenta, expression is limited to the trophoblast lineage, where it remains highly expressed until birth. Deletion of the miR-290 cluster gene (Mirc5) results in reduced trophoblast progenitor cell proliferation and a reduced DNA content in endoreduplicating trophoblast giant cells. The resulting placenta is reduced in size. In addition, the vascular labyrinth is disorganized, with thickening of the maternal-fetal blood barrier and an associated reduction in diffusion. Multiple mRNA targets of the miR-290 cluster microRNAs are upregulated. These data uncover a crucial function for the miR-290 cluster in the regulation of a network of genes required for placental development, suggesting a central role for these microRNAs in the evolution of placental mammals.
Collapse
Affiliation(s)
- Alireza Paikari
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Urology, University of California, San Francisco, CA 94143, USA
| | - Cassandra D Belair
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Urology, University of California, San Francisco, CA 94143, USA
| | - Daniel Saw
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Urology, University of California, San Francisco, CA 94143, USA
| | - Robert Blelloch
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA .,Department of Urology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
80
|
Singh K, Pal D, Sinha M, Ghatak S, Gnyawali SC, Khanna S, Roy S, Sen CK. Epigenetic Modification of MicroRNA-200b Contributes to Diabetic Vasculopathy. Mol Ther 2017; 25:2689-2704. [PMID: 29037594 DOI: 10.1016/j.ymthe.2017.09.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/01/2017] [Accepted: 09/07/2017] [Indexed: 12/26/2022] Open
Abstract
Hyperglycemia (HG) induces genome-wide cytosine demethylation. Our previous work recognized miR-200b as a critical angiomiR, which must be transiently downregulated to initiate wound angiogenesis. Under HG, miR-200b downregulation is not responsive to injury. Here, we demonstrate that HG may drive vasculopathy by epigenetic modification of a miR promoter. In human microvascular endothelial cells (HMECs), HG also lowered DNA methyltransferases (DNMT-1 and DNMT-3A) and compromised endothelial function as manifested by diminished endothelial nitric oxide (eNOS), lowered LDL uptake, impaired Matrigel tube formation, lower NO production, and compromised VE-cadherin expression. Bisulfite-sequencing documented HG-induced miR-200b promoter hypomethylation in HMECs and diabetic wound-site endothelial cells. In HMECs, HG compromised endothelial function. Methyl donor S-adenosyl-L-methionine (SAM) corrected miR-200b promoter hypomethylaton and rescued endothelial function. In vivo, wound-site administration of SAM to diabetic mice improved wound perfusion by limiting the pathogenic rise of miR-200b. Quantitative stable isotope labeling by amino acids in cell culture (SILAC) proteomics and ingenuity pathway analysis identified HG-induced proteins and principal clusters in HMECs sensitive to the genetic inhibition of miR-200b. This work presents the first evidence of the miR-200b promoter methylation as a critical determinant of diabetic wound angiogenesis.
Collapse
Affiliation(s)
- Kanhaiya Singh
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Durba Pal
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Mithun Sinha
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Subhadip Ghatak
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Surya C Gnyawali
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Savita Khanna
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Sashwati Roy
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Chandan K Sen
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
81
|
Farías JG, Molina VM, Carrasco RA, Zepeda AB, Figueroa E, Letelier P, Castillo RL. Antioxidant Therapeutic Strategies for Cardiovascular Conditions Associated with Oxidative Stress. Nutrients 2017; 9:nu9090966. [PMID: 28862654 PMCID: PMC5622726 DOI: 10.3390/nu9090966] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress (OS) refers to the imbalance between the generation of reactive oxygen species (ROS) and the ability to scavenge these ROS by endogenous antioxidant systems, where ROS overwhelms the antioxidant capacity. Excessive presence of ROS results in irreversible damage to cell membranes, DNA, and other cellular structures by oxidizing lipids, proteins, and nucleic acids. Oxidative stress plays a crucial role in the pathogenesis of cardiovascular diseases related to hypoxia, cardiotoxicity and ischemia-reperfusion. Here, we describe the participation of OS in the pathophysiology of cardiovascular conditions such as myocardial infarction, anthracycline cardiotoxicity and congenital heart disease. This review focuses on the different clinical events where redox factors and OS are related to cardiovascular pathophysiology, giving to support for novel pharmacological therapies such as omega 3 fatty acids, non-selective betablockers and microRNAs.
Collapse
Affiliation(s)
- Jorge G Farías
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
| | - Víctor M Molina
- Unidad de Cuidados Intensivos, Hospital de Niños Roberto del Río, Santiago 7500922, Chile.
- Unidad de Cuidados Intensivos Pediátricos, Hospital Clínico Pontificia Universidad Católica de Chile, Santiago 7500922, Chile.
| | - Rodrigo A Carrasco
- Laboratorio de Investigación Biomédica, Departamento de Medicina Interna, Hospital del Salvador, Santiago 7500922, Chile.
- Departamento de Cardiología, Clínica Alemana, Santiago 7500922, Chile.
| | - Andrea B Zepeda
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
| | - Elías Figueroa
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
- Núcleo de Investigación en Producción Alimentaria, BIOACUI, Escuela de Acuicultura, Universidad Católica de Temuco, Temuco 4780000, Chile.
| | - Pablo Letelier
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
- School of Health Sciences, Universidad Católica de Temuco, Temuco 4780000, Chile.
| | - Rodrigo L Castillo
- Laboratorio de Investigación Biomédica, Departamento de Medicina Interna, Hospital del Salvador, Santiago 7500922, Chile.
- Programa de Fisiopatología Oriente, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile.
| |
Collapse
|
82
|
Fiala O, Pitule P, Hosek P, Liska V, Sorejs O, Bruha J, Vycital O, Buchler T, Poprach A, Topolcan O, Finek J. The association of miR-126-3p, miR-126-5p and miR-664-3p expression profiles with outcomes of patients with metastatic colorectal cancer treated with bevacizumab. Tumour Biol 2017; 39:1010428317709283. [PMID: 28714375 DOI: 10.1177/1010428317709283] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs regulate the expression of genes involved in several important cancer-related processes including cell adhesion, proliferation, and tumour angiogenesis. Bevacizumab is routinely used in the treatment of patients with metastatic colorectal cancer, but, so far, no reliable biomarker predicting response to bevacizumab has been established. The aim of our retrospective study was to evaluate the association of miR-126-3p, miR-126-5p and miR-664-3p tumour expression levels with outcomes of patients with metastatic colorectal cancer treated with bevacizumab. The study included 63 patients. For the assessment of microRNA expression, gene-specific TaqMan assays were used. The median progression-free survival and overall survival for patients with low tumour expression of miR-126-3p were 8.8 and 20.6 months versus 13.5 months and median overall survival was not reached for patients with high expression ( p = 0.0064 and p = 0.0027), respectively. The median progression-free survival and overall survival for patients with low tumour expression of miR-126-5p were 9.0 and 22.2 months versus 12.0 and 23.4 months for patients with high expression ( p = 0.2113 and 0.6858), respectively. The median progression-free survival and overall survival for patients with low tumour expression of miR-664-3p were 9.1 and 22.5 months versus 8.8 and 23.4 months for patients with high expression ( p = 0.2542 and p = 0.1922), respectively. The multivariable Cox proportional hazards model revealed that miR-126-3p expression was significantly associated with progression-free survival (hazard ratio = 0.28, p = 0.0053) and also with overall survival (hazard ratio = 0.18, p = 0.0046). In conclusion, the results of this study suggest that the expression of miR-126-3p in the tumour tissue was associated with outcome of metastatic colorectal cancer patients treated with bevacizumab.
Collapse
Affiliation(s)
- Ondrej Fiala
- 1 Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic.,2 Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Pitule
- 2 Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Petr Hosek
- 2 Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Vaclav Liska
- 2 Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.,3 Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondrej Sorejs
- 1 Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jan Bruha
- 3 Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondrej Vycital
- 3 Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Tomas Buchler
- 4 Department of Oncology and First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Alexandr Poprach
- 5 Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Ondrej Topolcan
- 6 Department of Nuclear Medicine, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jindrich Finek
- 1 Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
83
|
Mulholland EJ, Dunne N, McCarthy HO. MicroRNA as Therapeutic Targets for Chronic Wound Healing. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 8:46-55. [PMID: 28918046 PMCID: PMC5485763 DOI: 10.1016/j.omtn.2017.06.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 06/06/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022]
Abstract
Wound healing is a highly complex biological process composed of three overlapping phases: inflammation, proliferation, and remodeling. Impairments at any one or more of these stages can lead to compromised healing. MicroRNAs (miRs) are non-coding RNAs that act as post-transcriptional regulators of multiple proteins and associated pathways. Thus, identification of the appropriate miR involved in the different phases of wound healing could reveal an effective third-generation genetic therapy in chronic wound care. Several miRs have been shown to be upregulated or downregulated during the wound healing process. This article examines the biological processes involved in wound healing, the miR involved at each stage, and how expression levels are modulated in the chronic wound environment. Key miRs are highlighted as possible therapeutic targets, either through underexpression or overexpression, and the healing benefits are interrogated. These are prime miR candidates that could be considered as a gene therapy option for patients suffering from chronic wounds. The success of miR as a gene therapy, however, is reliant on the development of an appropriate delivery system that must be designed to overcome both extracellular and intracellular barriers.
Collapse
Affiliation(s)
- Eoghan J Mulholland
- School of Pharmacy, Queen's University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Nicholas Dunne
- School of Pharmacy, Queen's University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.
| | - Helen O McCarthy
- School of Pharmacy, Queen's University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
84
|
Biswas A, Pan X, Meyer M, Khanna S, Roy S, Pearson G, Kirschner R, Witman P, Faith EF, Sen CK, Gordillo GM. Urinary Excretion of MicroRNA-126 Is a Biomarker for Hemangioma Proliferation. Plast Reconstr Surg 2017; 139:1277e-1284e. [PMID: 28538565 PMCID: PMC5963954 DOI: 10.1097/prs.0000000000003349] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Hemangiomas are unique endothelial cell tumors that involute spontaneously, which makes interpreting their response to therapies difficult. The objective of this work was to identify a potential biomarker in the urine of children with infantile hemangiomas that would facilitate testing new therapies. METHODS A prospective longitudinal study in children with hemangiomas and age-matched healthy controls was performed to determine whether microRNA-126, which is highly abundant in fetal endothelial cells, was more abundant in the urine of affected children. Prospective ultrasound measurements of hemangioma size and blood flow velocity were obtained as secondary endpoints to document longitudinal changes in untreated hemangiomas. RESULTS Urinary microRNA-126 levels were significantly elevated in children with proliferating hemangiomas, and relative levels of urinary microRNA abundance correlated with hemangioma size. Hemangiomas had elevated levels of microRNA abundance compared with healthy controls. Ultrasound data revealed that hemangioma proliferation typically stopped between 6 and 9 months of age. When hemangioma proliferation stopped, urinary microRNA-126 levels in children with hemangiomas dropped to levels observed in healthy age-matched controls. CONCLUSIONS These are the first reported results to identify a potential microRNA biomarker in the urine of children with hemangiomas. Measurement of urinary levels of microRNA-126 could potentially be used to monitor hemangioma response to therapies. CLINICAL QUESTION/LEVEL OF EVIDENCE Diagnostic, II.
Collapse
Affiliation(s)
- Ayan Biswas
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Xueliang Pan
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Melissa Meyer
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Savita Khanna
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Sashwati Roy
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Gregory Pearson
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Richard Kirschner
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Patricia Witman
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Esteban Fernandez Faith
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Chandan K Sen
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Gayle M Gordillo
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| |
Collapse
|
85
|
Lacko LA, Hurtado R, Hinds S, Poulos MG, Butler JM, Stuhlmann H. Altered feto-placental vascularization, feto-placental malperfusion and fetal growth restriction in mice with Egfl7 loss of function. Development 2017; 144:2469-2479. [PMID: 28526753 DOI: 10.1242/dev.147025] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/16/2017] [Indexed: 01/16/2023]
Abstract
EGFL7 is a secreted angiogenic factor produced by embryonic endothelial cells. To understand its role in placental development, we established a novel Egfl7 knockout mouse. The mutant mice have gross defects in chorioallantoic branching morphogenesis and placental vascular patterning. Microangiography and 3D imaging revealed patchy perfusion of Egfl7-/- placentas marked by impeded blood conductance through sites of narrowed vessels. Consistent with poor feto-placental perfusion, Egfl7 knockout resulted in reduced placental weight and fetal growth restriction. The placentas also showed abnormal fetal vessel patterning and over 50% reduction in fetal blood space. In vitro, placental endothelial cells were deficient in migration, cord formation and sprouting. Expression of genes involved in branching morphogenesis, Gcm1, Syna and Synb, and in patterning of the extracellular matrix, Mmrn1, were temporally dysregulated in the placentas. Egfl7 knockout did not affect expression of the microRNA embedded within intron 7. Collectively, these data reveal that Egfl7 is crucial for placental vascularization and embryonic growth, and may provide insight into etiological factors underlying placental pathologies associated with intrauterine growth restriction, which is a significant cause of infant morbidity and mortality.
Collapse
Affiliation(s)
- Lauretta A Lacko
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA
| | - Romulo Hurtado
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA
| | - Samantha Hinds
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA
| | - Michael G Poulos
- Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA
| | - Jason M Butler
- Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA
| | - Heidi Stuhlmann
- Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA
| |
Collapse
|
86
|
Jiang R, Zhang C, Liu G, Gu R, Wu H. Retracted
: MicroRNA‐126 Inhibits Proliferation, Migration, Invasion, and EMT in Osteosarcoma by Targeting ZEB1. J Cell Biochem 2017; 118:3765-3774. [DOI: 10.1002/jcb.26024] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/31/2017] [Indexed: 02/02/2023]
Affiliation(s)
- Rui Jiang
- Department of OrthopedicsChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Chao Zhang
- Department of OphthalmologyThe Second Hospital of Jilin UniversityChangchunChina
| | - Guangyao Liu
- Department of OrthopedicsChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Rui Gu
- Department of OrthopedicsChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Han Wu
- Department of OrthopedicsChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
87
|
van Beijnum JR, Giovannetti E, Poel D, Nowak-Sliwinska P, Griffioen AW. miRNAs: micro-managers of anticancer combination therapies. Angiogenesis 2017; 20:269-285. [PMID: 28474282 PMCID: PMC5519663 DOI: 10.1007/s10456-017-9545-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/11/2017] [Indexed: 12/15/2022]
Abstract
Angiogenesis is one of the hallmarks of cancer progression and as such has been considered a target of therapeutic interest. However, single targeted agents have not fully lived up to the initial promise of anti-angiogenic therapy. Therefore, it has been suggested that combining therapies and agents will be the way forward in the oncology field. In recent years, microRNAs (miRNAs) have received considerable attention as drivers of tumor development and progression, either acting as tumor suppressors or as oncogenes (so-called oncomiRs), as well as in the process of tumor angiogenesis (angiomiRs). Not only from a functional, but also from a therapeutic view, miRNAs are attractive tools. Thus far, several mimics and antagonists of miRNAs have entered clinical development. Here, we review the provenance and promise of miRNAs as targets as well as therapeutics to contribute to anti-angiogenesis-based (combination) treatment of cancer.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VUMC - Cancer Center Amsterdam, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Laboratory Medical Oncology, Department of Medical Oncology, VUMC - Cancer Center Amsterdam, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - Dennis Poel
- Angiogenesis Laboratory, Department of Medical Oncology, VUMC - Cancer Center Amsterdam, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | | | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VUMC - Cancer Center Amsterdam, VU University Medical Center (VUmc), Amsterdam, The Netherlands.
| |
Collapse
|
88
|
MicroRNA miR-126-5p Enhances the Inflammatory Responses of Monocytes to Lipopolysaccharide Stimulation by Suppressing Cylindromatosis in Chronic HIV-1 Infection. J Virol 2017; 91:JVI.02048-16. [PMID: 28250134 DOI: 10.1128/jvi.02048-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/24/2017] [Indexed: 02/02/2023] Open
Abstract
Persistent immune activation during chronic human immunodeficiency virus type 1 (HIV-1) infection facilitates immune dysfunction and thereby fuels disease progression. The translocation of bacterial derivatives into blood and the hyperinflammatory responsiveness of monocytes have been considered important causative factors for persistent immune activation. Whether microRNAs (miRNAs) are involved in regulating monocyte-mediated inflammatory responses during chronic HIV-1 infection remains elusive. In this study, we show that miR-126-5p functions as a positive regulator of monocyte-mediated inflammatory responses. Significantly increased miRNA miR-126-5p and decreased cylindromatosis (CYLD) were observed in primary monocytes from chronic HIV-1 patients. Inhibition of miR-126-5p in monocytes from chronic HIV-1 patients attenuated the responsiveness of these cells to lipopolysaccharide (LPS) stimulation. Gain-of-function assays confirmed that miR-126-5p could downregulate CYLD, which in turn caused an upregulation of phosphorylation of JNK protein (pJNK) and enhanced inflammatory responses of monocytes to LPS stimulation. Overall, miR-126-5p upregulates the responsiveness of monocytes to LPS stimulation in chronic HIV-1 infection, and the suppression of miR-126-5p and the promotion of CYLD expression in primary monocytes may represent a practical immune intervention strategy to contain persistent inflammation in chronic HIV-1 infection.IMPORTANCE Monocyte-mediated hyperinflammatory responses during chronic HIV-1 infection are important causative factors driving AIDS progression; however, the underlying mechanism has not been fully addressed. We demonstrated that miR-126-5p, one of the most upregulated miRNAs during chronic HIV-1 infection, could enhance the inflammatory responses of monocytes to LPS by suppressing the inhibitory protein CYLD and thereby unleashing the expression of pJNK in the LPS/Toll-like receptor 4/mitogen-activated protein kinase pathway. This observation reveals a new mechanism for HIV-1 pathogenesis, which could be targeted by immune intervention.
Collapse
|
89
|
Gong C, Fang J, Li G, Liu HH, Liu ZS. Effects of microRNA-126 on cell proliferation, apoptosis and tumor angiogenesis via the down-regulating ERK signaling pathway by targeting EGFL7 in hepatocellular carcinoma. Oncotarget 2017; 8:52527-52542. [PMID: 28881749 PMCID: PMC5581048 DOI: 10.18632/oncotarget.17283] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/24/2017] [Indexed: 12/31/2022] Open
Abstract
This study intends to explore the effects of microRNA-126 (miR-126) on cell proliferation, apoptosis, and tumor angiogenesis in hepatocellular carcinoma (HCC) by regulating epidermal growth factor-like domain 7 (EGFL7) through extracellular signal-regulated kinase (ERK) signaling. HCC tissues and adjacent normal tissues were obtained from 184 HCC patients. HCC cells were separately transfected with recombinant plasmids. Western blotting and qRT-PCR were applied to detect miR-126 and EGFL7, ERK, Fas/FasL, Bcl-2, Caspase mRNA and protein levels. CCK8 and TUNEL were performed to determinate cell proliferation and apoptosis. Flow cytometry was used to analyze cell cycle distribution. Rats model of HCC was constructed, and tumor weight and the number of new blood vessels were recorded after 3 weeks of tumor transplantation. Compared with the adjacent normal tissues, HCC tissues exhibited lower miR-126 expression, and higher EGFL7, and ERK mRNA and protein levels. Overexpression of miR-126 in HCC cell lines suppressed EGFL7, ERK, Bcl-2, and P-ERK, and increased apoptotic-associated proteins Fas/FasL and Caspase-3, and it inhibited cell proliferation and induced cell apoptosis. Overexpression of miR-126 in nude mice resulted in reduced tumor weight and less new blood vessels in tumors. The inhibition of miR-126 decreased cell apoptosis, and enhanced cell proliferation and tumor angiogenesis. This study demonstrates that miR-126 might decrease cell proliferation, induce apoptosis, and inhibit tumor angiogenesis in HCC by inhibiting EGFL7 via down-regulating the ERK signaling pathway.
Collapse
Affiliation(s)
- Cheng Gong
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Jing Fang
- Department of Oncology, Wuhan Pu-Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, P.R. China
| | - Guang Li
- Department of Oncology, Wuhan Pu-Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, P.R. China
| | - Han-Han Liu
- Department of Pathology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, P.R. China
| | - Zhi-Su Liu
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| |
Collapse
|
90
|
Yang WZ, Yang J, Xue LP, Xiao LB, Li Y. MiR-126 overexpression inhibits high glucose-induced migration and tube formation of rhesus macaque choroid-retinal endothelial cells by obstructing VEGFA and PIK3R2. J Diabetes Complications 2017; 31:653-663. [PMID: 28131600 DOI: 10.1016/j.jdiacomp.2016.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 11/27/2016] [Accepted: 12/19/2016] [Indexed: 12/26/2022]
Abstract
AIM The aims of this study are to investigate the relative regulation between miR-126 and VEGF/PI3K/AKT signaling pathway in retinal vascular endothelial cells. METHODS Rhesus macaque choroid-retinal endothelial cell line (RF/6A) cells were cultured in high glucose to imitate the conditions occurring in DR. First, we detected the expression of miR-126, VEGFA and PIK3R2 in RF/6A cells on the condition of high glucose by q-PCR and western blot. Then, after addition of miR-126 mimics and miR-126 inhibitor, we investigated the function of miR-126 in RF/6A cells by scratch wound, Transwell migration and tube formation assays, and the effect of miR-126 on the expression of VEGFA, PIK3R2 and AKT. Moreover, bioinformatics analysis and luciferase array were used to confirm the direct or specific regulation of miR-126 to VEGFA or PIK3R2. RESULTS Here, first, we found that high glucose could induce the decrease of miR-126 and the increase of VEGFA and PIK3R2 in RF/6A. Then, by scratch wound, Transwell migration and tube formation assays, we found that miR-126 overexpression could inhibit the migration and sprouting of RF/6A cells induced by high glucose, while knockdown of miR-126 led to the opposite results. Moreover, overexpression of miR-126 inhibited the increased expression of VEGFA, PIK3R2, SDF-1α, VCAM-1, and SPRED1, and the activation of AKT1 induced by high glucose and miR-126 inhibitor caused the opposite results which were determined by q-PCR and western blot. In addition, by luciferase assay, we found that miR-126 could directly negatively regulate VEGFA and PIK3R2. CONCLUSION Our results suggest that miR-126 overexpression inhibits the migration and sprouting of RF/6A cells induced by high glucose which might possibly be by blocking VEGFA and PIK3R2 in the VEGF/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Wen-Zhi Yang
- Department of Ophthalmology, Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, China
| | - Jin Yang
- Department of Osteology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Li-Ping Xue
- Department of Ophthalmology, Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, China
| | - Li-Bo Xiao
- Department of Ophthalmology, Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, China
| | - Yan Li
- Department of Ophthalmology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| |
Collapse
|
91
|
Huleihel L, Scarritt ME, Badylak SF. The Influence of Extracellular RNA on Cell Behavior in Health, Disease and Regeneration. CURRENT PATHOBIOLOGY REPORTS 2017; 5:13-22. [PMID: 28944104 PMCID: PMC5604481 DOI: 10.1007/s40139-017-0121-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW An overview of the role of extracellular RNAs (exRNA) in the regulation of homeostasis, disease progression, and regeneration is provided herein. Several exRNAs have been identified as potential biomarkers for disease and disease progression. In addition, the potential of exRNAs as a therapeutic modality is discussed. RECENT FINDINGS Fibrotic diseases of the lung, liver, and heart, among other organs share a number of identical exRNAs which play key roles in disease pathogenesis. Though regeneration is limited to only a few tissues in humans, small RNAs (e.g. microRNA) have been shown to be involved in the regenerative process of tissues such as liver and bone. The regulation of healing versus disease appears to be balanced by small RNAs. Because small RNAs are critical to health, they are being investigated as drug targets in multiple ongoing clinical trials. Preclinical studies suggest that promoting or blocking specific small RNAs can provide a novel therapeutic approach. SUMMARY exRNA can be utilized for both detection and treatment of disease. Natural and synthetic RNA carriers are being investigated as delivery methods for small RNA molecules. Current and future investigations are likely to lead to expanded applications for exRNAs.
Collapse
Affiliation(s)
- Luai Huleihel
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Michelle E. Scarritt
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
92
|
Vimalraj S, Sumantran VN, Chatterjee S. MicroRNAs: Impaired vasculogenesis in metal induced teratogenicity. Reprod Toxicol 2017; 70:30-48. [PMID: 28249814 DOI: 10.1016/j.reprotox.2017.02.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 02/07/2023]
Abstract
Certain metals have been known for their toxic effects on embryos and fetal development. The vasculature in early pregnancy is extremely dynamic and plays an important role in organogenesis. Nascent blood vessels in early embryonic life are considered to be a primary and delicate target for many teratogens since the nascent blood islands follow a tightly controlled program to form vascular plexus around and inside the embryo for resourcing optimal ingredients for its development. The state of the distribution of toxic metals, their transport mechanisms and the molecular events by which they notch extra-embryonic and embryonic vasculatures are illustrated. In addition, pharmacological aspects of toxic metal induced teratogenicity have also been portrayed. The work reviewed state of the current knowledge of specific role of microRNAs (miRNAs) that are differentially expressed in response to toxic metals, and how they interfere with the vasculogenesis that manifests into embryonic anomalies.
Collapse
Affiliation(s)
- Selvaraj Vimalraj
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India.
| | | | - Suvro Chatterjee
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India; Department of Biotechnology, Anna University, Chennai, India.
| |
Collapse
|
93
|
Kong F, Zhou J, Zhou W, Guo Y, Li G, Yang L. Protective role of microRNA-126 in intracerebral hemorrhage. Mol Med Rep 2017; 15:1419-1425. [PMID: 28112373 DOI: 10.3892/mmr.2017.6134] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/01/2016] [Indexed: 11/05/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a disease associated with high mortality and morbidity. MicroRNAs (miRNAs) are important regulators of translation and have been reported to be associated with the pathogenesis of numerous cerebrovascular diseases, including ICH. The present study explored the role of miRNA (miR)‑126 in ICH. Adult male Wistar rats were randomly assigned to ICH model and sham groups. ICH was induced by intracerebral injection of collagenase. The mRNA expression levels of miR‑126 in the two groups were determined. The miR‑126 lentivirus expression vector pWPXL‑miR‑126 or negative control vector was then constructed and delivered via intraparenchymal injection. Following transduction, behavioral testing (rotarod and limb placement tests), relative hemorrhagic lesion size, apoptotic cells and protein levels of vascular endothelial growth factor (VEGF)‑A and caspase‑3 were determined. The relative expression levels of miR‑126 were significantly decreased in the ICH group compared to the sham group (P=0.026). Overexpression of miR‑126 significantly improved the relative duration of stay on the rotarod at day 2 (P=0.029) and 3 (P=0.033), and statistically reduced the deficit score (P=0.036), the relative size of hemorrhagic lesion (P=0.019) and the number of apoptotic cortical neurons (P=0.024) compared with the sham group. Additionally, the protein levels of VEGF‑A were significantly elevated, however levels of caspase‑3 were downregulated by overexpression of miR‑126 compared with the negative control group. MiR‑126 therefore exhibits a protective role in ICH. Overexpression of miR‑126 protects against ICH, and may be involved in the process of angiogenesis and exhibit an anti-apoptotic effect.
Collapse
Affiliation(s)
- Fangen Kong
- Department of Neurosurgery, Fifth Affiliated Hospital of Sun Yat‑Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Jianhui Zhou
- Department of Clinical Laboratory, Fifth Affiliated Hospital of Sun Yat‑Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Wenying Zhou
- Department of Central Laboratory, Fifth Affiliated Hospital of Sun Yat‑Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Yuanqing Guo
- Department of Orthopaedics, Fifth Affiliated Hospital of Sun Yat‑Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Guowei Li
- Department of Orthopaedics, Fifth Affiliated Hospital of Sun Yat‑Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Lukun Yang
- Department of Anesthesiology, Fifth Affiliated Hospital of Sun Yat‑Sen University, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
94
|
Chen J, Cui C, Yang X, Xu J, Venkat P, Zacharek A, Yu P, Chopp M. MiR-126 Affects Brain-Heart Interaction after Cerebral Ischemic Stroke. Transl Stroke Res 2017; 8:374-385. [PMID: 28101763 DOI: 10.1007/s12975-017-0520-z] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 12/31/2022]
Abstract
Cardiovascular diseases are approximately three times higher in patients with neurological deficits than in patients without neurological deficits. MicroRNA-126 (MiR-126) facilitates vascular remodeling and decreases fibrosis and is emerging as an important factor in the pathogenesis of cardiovascular diseases and cerebral stroke. In this study, we tested the hypothesis that decreased miR-126 after ischemic stroke may play an important role in regulating cardiac function. Wild-type (WT), specific conditional-knockout endothelial cell miR-126 (miR-126EC-/-), and miR-126 knockout control (miR-126fl/fl) mice were subjected to distal middle cerebral artery occlusion (dMCAo) (n = 10/group). Cardiac hemodynamics and function were measured using transthoracic Doppler echocardiography. Mice were sacrificed at 28 days after dMCAo. WT mice subjected to stroke exhibited significantly decreased cardiac ejection fraction and increased myocyte hypertrophy, fibrosis as well as increased heart inflammation, infiltrating macrophages, and oxidative stress compared to non-stroke animals. Stroke significantly decreased serum and heart miR-126 expression and increased miR-126 target genes, vascular cell adhesion protein-1, and monocyte chemotactic protein-1 gene, and protein expression in the heart compared to non-stroke mice. MiR-126EC-/- mice exhibited significantly decreased cardiac function and increased cardiomyocyte hypertrophy, fibrosis, and inflammatory factor expression after stroke compared to miR-126fl/fl stroke mice. Exosomes derived from endothelial cells of miR-126EC-/- (miR-126EC-/-EC-Exo) mice exhibited significantly decreased miR-126 expression than exosomes derived from miR-126fl/fl (miR-126fl/fl-EC-Exo) mice. Treatment of cardiomyocytes subjected to oxygen glucose deprivation with miR-126fl/fl-EC-Exo exhibited significantly decreased hypertrophy than with miR-126EC-/-EC-Exo treatment. Ischemic stroke directly induces cardiac dysfunction. Decreasing miR-126 expression may contribute to cardiac dysfunction after stroke in mice.
Collapse
Affiliation(s)
- Jieli Chen
- Henry Ford Hospital Neurology, Detroit, MI, 48202, USA. .,Gerontology Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | | | - Xiaoping Yang
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Jiang Xu
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI, 48202, USA
| | | | - Alex Zacharek
- Henry Ford Hospital Neurology, Detroit, MI, 48202, USA
| | - Peng Yu
- Henry Ford Hospital Neurology, Detroit, MI, 48202, USA
| | - Michael Chopp
- Henry Ford Hospital Neurology, Detroit, MI, 48202, USA.,Department of Physics, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
95
|
Codagnone M, Recchiuti A, Lanuti P, Pierdomenico AM, Cianci E, Patruno S, Mari VC, Simiele F, Di Tomo P, Pandolfi A, Romano M. Lipoxin A 4 stimulates endothelial miR-126-5p expression and its transfer via microvesicles. FASEB J 2017; 31:1856-1866. [PMID: 28100645 DOI: 10.1096/fj.201600952r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/03/2017] [Indexed: 01/01/2023]
Abstract
The proresolution lipid mediator lipoxin (LX)A4 bestows protective bioactions on endothelial cells. We examined the impact of LXA4 on transcellular endothelial signaling via microRNA (miR)-containing microvesicles. We report LXA4 inhibition of MV release by TNF-α-treated HUVECs, associated with the down-regulation of 18 miR in endothelial microvesicles (EMVs) and the up-regulation of miR-126-5p, both in HUVECs and in EMVs. LXA4 up-regulated miR-126-5p by ∼5-fold in HUVECs and promoted a release of microvesicles (LXA4-EMVs) that enhanced miR-126-5p by ∼7-fold in recipient HUVECs. In these cells, LXA4-EMVs abrogated the up-regulation of VCAM-1, induced in recipient HUVECs by EMVs released by untreated or TNF-α-treated HUVECs. LXA4-EMVs also reduced by ∼40% the expression of SPRED1, which we validated as an miR-126-5p target, whereas they stimulated monolayer repair in an in vitro wound assay. This effect was lost when the EMVs were depleted of miR-126-5p. These results provide evidence that changes in miR expression and microvesicle packaging and transfer represent a mechanism of action of LXA4, which may be relevant in vascular biology and inflammation.-Codagnone, M., Recchiuti, A., Lanuti, P., Pierdomenico, A. M., Cianci, E., Patruno, S., Mari, V. C., Simiele, F., Di Tomo, P., Pandolfi, A., Romano, M. Lipoxin A4 stimulates endothelial miR-126-5p expression and its transfer via microvesicles.
Collapse
Affiliation(s)
- Marilina Codagnone
- Department of Medical, Oral, and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy.,Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| | - Antonio Recchiuti
- Department of Medical, Oral, and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy.,Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| | - Paola Lanuti
- Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and.,Department of Medicine and Aging Science, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Anna Maria Pierdomenico
- Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and.,Department of Medicine and Aging Science, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Eleonora Cianci
- Department of Medical, Oral, and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy.,Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| | - Sara Patruno
- Department of Medical, Oral, and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy.,Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| | - Veronica Cecilia Mari
- Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| | - Felice Simiele
- Department of Medical, Oral, and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy.,Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| | - Pamela Di Tomo
- Department of Medical, Oral, and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy.,Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| | - Assunta Pandolfi
- Department of Medical, Oral, and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy.,Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| | - Mario Romano
- Department of Medical, Oral, and Biotechnological Sciences, G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; .,Center on Aging Science and Translational Medicine (CeSI-MeT), G. D'Annunzio University of Chieti-Pescara, Chieti, Italy; and
| |
Collapse
|
96
|
Rawal S, Munasinghe PE, Shindikar A, Paulin J, Cameron V, Manning P, Williams MJA, Jones GT, Bunton R, Galvin I, Katare R. Down-regulation of proangiogenic microRNA-126 and microRNA-132 are early modulators of diabetic cardiac microangiopathy. Cardiovasc Res 2017; 113:90-101. [PMID: 28065883 DOI: 10.1093/cvr/cvw235] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 07/15/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023] Open
Abstract
AIM Microangiopathy due to endothelial dysfunction is a major contributing factor to the development of diabetes-induced cardiovascular disease (CVD). Dysregulation of endothelial-specific microRNAs (miRs) is correlated with impaired angiogenesis and cell survival. We investigated the profile of two angiomiRs, miR-126, and miR-132, in the plasma of type 2 diabetic individuals without any known history of CVD as well as in the cardiac tissues collected from diabetics undergoing cardiac surgery. METHODS AND RESULTS The presence of diabetes alone significantly decreased both angiomiRs in the plasma and the myocardium. The down-regulation of angiomiRs was also associated with reduced capillaries and arterioles and increased endothelial cell apoptosis, the hallmark of microangiopathy. Importantly, a time course study in a type 2 diabetic mouse model confirmed that the down-regulation of angiomiRs preceded endothelial apoptosis as well as alterations in the density of the microvasculature. Finally, therapeutic overexpression of both angiomiRs in diabetic aortic rings and human umbilical vein endothelial cells exposed to high glucose (HG) abrogated the deleterious effects of diabetes and HG on cell survival and proliferation and restored their angiogenic potential. CONCLUSIONS These novel findings demonstrate that the down-regulation of angiomiRs is a major underlying mechanism for the development of microangiopathy in diabetic hearts. Therefore, therapeutic restoration of angiomiRs could become a potential approach to combat the cardiovascular complications of diabetes.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Physiology-HeartOtago, Otago School of Medical Sciences
| | | | - Amol Shindikar
- Department of Physiology-HeartOtago, Otago School of Medical Sciences
| | - Jono Paulin
- Department of Physiology-HeartOtago, Otago School of Medical Sciences
| | | | | | | | | | - Richard Bunton
- Cardiothoracic Surgery, Dunedin School of Medicine, University of Otago
| | - Ivor Galvin
- Cardiothoracic Surgery, Dunedin School of Medicine, University of Otago
| | - Rajesh Katare
- Department of Physiology-HeartOtago, Otago School of Medical Sciences;
| |
Collapse
|
97
|
Wang Y, Wu S, Yang Y, Peng F, Li Q, Tian P, Xiang E, Liang H, Wang B, Zhou X, Huang H, Zhou X. Differentially expressed miRNAs in oxygen‑induced retinopathy newborn mouse models. Mol Med Rep 2016; 15:146-152. [PMID: 27922698 PMCID: PMC5355681 DOI: 10.3892/mmr.2016.5993] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 09/08/2016] [Indexed: 12/26/2022] Open
Abstract
The present study aimed to identify microRNAs (miRNAs) involved in regulating retinal neovascularization and retinopathy of prematurity (ROP). A total of 80 healthy C57BL/6 neonatal mice were randomly divided into the oxygen-induced retinopathy (OIR) group (n=40), in which 7-day-old mice were maintained in 75% oxygen conditions for 5 days, or the control group (n=40). Following collection of retinal tissue, retinal angiography and hematoxylin and eosin (H&E) staining were performed. Total RNA was also extracted from retinal tissue, and miRNA microarrays and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were performed to identify differentially expressed miRNAs in the two groups. Retinal angiography and H&E staining revealed damage to retinas in the OIR group. Compared with the control group, 67 miRNAs were differentially expressed in the OIR group, of which 34 were upregulated and 33 were downregulated. Of these differentially expressed miRNAs, 32 exhibited a fold change ≥2, of which 21 were upregulated and 11 were downregulated. The results of RT-qPCR for miR-130a-3p and miR-5107-5p were in accordance with those of the miRNA microarray. The newly identified miRNAs may be important in the development of ROP, and may provide a basis for future research into the mechanisms of ROP.
Collapse
Affiliation(s)
- Yunpeng Wang
- Department of Neonatology, Nanshan People's Hospital, Affiliated to Guangdong Medical University, Shenzhen, Guangdong 518052, P.R. China
| | - Suying Wu
- Department of Neonatology, University Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Yang Yang
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Fen Peng
- Department of Neonatology, University Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Qintao Li
- Department of Neonatology, University Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Peng Tian
- Department of Neonatology, University Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Erying Xiang
- Department of Neonatology, University Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Honglu Liang
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Beibei Wang
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaoyu Zhou
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Hua Huang
- Department of Neonatology, University Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Xiaoguang Zhou
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
98
|
Araldi E, Suárez Y. MicroRNAs as regulators of endothelial cell functions in cardiometabolic diseases. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1861:2094-2103. [PMID: 26825686 PMCID: PMC5039046 DOI: 10.1016/j.bbalip.2016.01.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/18/2016] [Accepted: 01/20/2016] [Indexed: 12/13/2022]
Abstract
Endothelial cells (ECs) provide nutrients and oxygen essential for tissue homeostasis. Metabolic imbalances and other environmental stimuli, like cytokines or low shear stress, trigger endothelial inflammation, increase permeability, compromise vascular tone, promote cell proliferation, and ultimately cause cell death. These factors contribute to EC dysfunction, which is crucial in the development of cardiometabolic diseases. microRNAs (miRNAs) are small non-coding RNAs that have important functions in the regulation of ECs. In the present review, we discuss the role of miRNAs in various aspects of EC pathology in cardiometabolic diseases like atherosclerosis, type 2 diabetes, obesity, and the metabolic syndrome, and in complication of those pathologies, like ischemia. We also discuss the potential therapeutic applications of miRNAs in promoting angiogenesis and neovascularization in tissues where the endothelium is damaged in different cardiometabolic diseases. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Elisa Araldi
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Department of Pathology and the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yajaira Suárez
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Department of Pathology and the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
99
|
Li M, Ke QF, Tao SC, Guo SC, Rui BY, Guo YP. Fabrication of hydroxyapatite/chitosan composite hydrogels loaded with exosomes derived from miR-126-3p overexpressed synovial mesenchymal stem cells for diabetic chronic wound healing. J Mater Chem B 2016; 4:6830-6841. [PMID: 32263577 DOI: 10.1039/c6tb01560c] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The exploration of an effective diabetic chronic wound healing process still remains a great challenge. Herein, we used gene overexpression technology to obtain synovial mesenchymal stem cells (SMSCs) and the miR-126-3p highly expressed SMSCs (SMSCs-126). The exosomes derived from miR-126-3p overexpressed SMSCs (SMSCs-126-Exos) with a particle size of 85 nm were encapsulated in hydroxyapatite/chitosan (HAP-CS) composite hydrogels (HAP-CS-SMSCs-126-Exos) as wound dressings. The SMSCs-126-Exos, CS and low-crystallinity HAP nanorods with a length of 200 nm and a diameter of 50 nm are uniformly dispersed within the whole composite hydrogel. The HAP-CS-SMSCs-126-Exos possess the controlled release property of SMSCs-126-Exos for at least 6 days. The released SMSCs-126-Exos nanoparticles stimulate the proliferation and migration of human dermal fibroblasts and human dermal microvascular endothelial cells (HMEC-1). At the same time, the migration and capillary-network formation of HMEC-1 are promoted through the activation of MAPK/ERK and PI3K/AKT. In vivo tests demonstrate that the HAP-CS-SMSCs-126-Exos successfully promote wound surface re-epithelialization, accelerate angiogenesis, and expedite collagen maturity due to the presence of HAP, CS and SMSCs-126-Exos. Therefore, the HAP-CS-SMSCs-126-Exos possess great potential application for diabetic chronic wound healing, and especially provide the possibility of using exosomes derived from modified cells as a new approach to bring wonderful functionality and controllability in future chronic wound therapy.
Collapse
Affiliation(s)
- Min Li
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China.
| | | | | | | | | | | |
Collapse
|
100
|
Azevedo-Pouly ACP, Sutaria DS, Jiang J, Elgamal OA, Amari F, Allard D, Grippo PJ, Coppola V, Schmittgen TD. miR-216 and miR-217 expression is reduced in transgenic mouse models of pancreatic adenocarcinoma, knockout of miR-216/miR-217 host gene is embryonic lethal. Funct Integr Genomics 2016; 17:203-212. [PMID: 27541609 DOI: 10.1007/s10142-016-0512-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/20/2016] [Accepted: 08/02/2016] [Indexed: 02/07/2023]
Abstract
Mice harboring a G12D activating Kras mutation are among the most heavily studied models in the field of pancreatic adenocarcinoma (PDAC) research. miRNAs are differentially expressed in PDAC from patients and mouse models of PDAC. To better understand the relationship that Kras activation has on miRNA expression, we profiled the expression of 629 miRNAs in RNA isolated from the pancreas of control, young, and old P48+/Cre;LSL-KRASG12D as well as PDX-1-Cre;LSL-KRASG12D mice. One hundred of the differentially expressed miRNAs had increased expression in the advanced disease (old) P48+/Cre;LSL-KRASG12D compared to wild-type mice. Interestingly, the expression of three miRNAs, miR-216a, miR-216b, and miR-217, located within a ∼30-kbp region on 11qA3.3, decreased with age (and phenotype severity) in these mice. miR-216/-217 expression was also evaluated in another acinar-specific ELa-KrasG12D mouse model and was downregulated as well. As miR-216/-217 are acinar enriched, reduced in human PDAC and target KRAS, we hypothesized that they may maintain acinar differentiation or represent tumor suppressive miRNAs. To test this hypothesis, we deleted a 27.9-kbp region of 11qA3.3 containing the miR-216/-217 host gene in the mouse's germ line. We report that germ line deletion of this cluster is embryonic lethal in the mouse. We estimate that lethality occurs shortly after E9.5. qPCR analysis of the miR-216b and miR-217 expression in the heterozygous animals showed no difference in expression, suggesting haplosufficiency by some type of compensatory mechanism. We present the differential miRNA expression in KrasG12D transgenic mice and report lethality from deletion of the miR-216/-217 host gene in the mouse's germ line.
Collapse
Affiliation(s)
- Ana Clara P Azevedo-Pouly
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Jinmai Jiang
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Ola A Elgamal
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Foued Amari
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Paul J Grippo
- Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Vincenzo Coppola
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Thomas D Schmittgen
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|