1
|
Li J, Yao H, Zhao F, An J, Wang Q, Mu J, Liu Z, Zou MH, Xie Z. Pycard deficiency inhibits microRNA maturation and prevents neointima formation by promoting chaperone-mediated autophagic degradation of AGO2/argonaute 2 in adipose tissue. Autophagy 2024; 20:629-644. [PMID: 37963060 PMCID: PMC10936599 DOI: 10.1080/15548627.2023.2277610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/10/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023] Open
Abstract
PYCARD (PYD and CARD domain containing), a pivotal adaptor protein in inflammasome assembly and activation, contributes to innate immunity, and plays an essential role in the pathogenesis of atherosclerosis and restenosis. However, its roles in microRNA biogenesis remain unknown. Therefore, this study aimed to investigate the roles of PYCARD in miRNA biogenesis and neointima formation using pycard knockout (pycard-/-) mice. Deficiency of Pycard reduced circulating miRNA profile and inhibited Mir17 seed family maturation. The systemic pycard knockout also selectively reduced the expression of AGO2 (argonaute RISC catalytic subunit 2), an important enzyme in regulating miRNA biogenesis, by promoting chaperone-mediated autophagy (CMA)-mediated degradation of AGO2, specifically in adipose tissue. Mechanistically, pycard knockout increased PRMT8 (protein arginine N-methyltransferase 8) expression in adipose tissue, which enhanced AGO2 methylation, and subsequently promoted its binding to HSPA8 (heat shock protein family A (Hsp70) member 8) that targeted AGO2 for lysosome degradation through chaperone-mediated autophagy. Finally, the reduction of AGO2 and Mir17 family expression prevented vascular injury-induced neointima formation in Pycard-deficient conditions. Overexpression of AGO2 or administration of mimic of Mir106b (a major member of the Mir17 family) prevented Pycard deficiency-mediated inhibition of neointima formation in response to vascular injury. These data demonstrate that PYCARD inhibits CMA-mediated degradation of AGO2, which promotes microRNA maturation, thereby playing a critical role in regulating neointima formation in response to vascular injury independently of inflammasome activity and suggest that modulating PYCARD expression and function may represent a powerful therapeutic strategy for neointima formation.Abbreviations: 6-AN: 6-aminonicotinamide; ACTB: actin, beta; aDMA: asymmetric dimethylarginine; AGO2: argonaute RISC catalytic subunit 2; CAL: carotid artery ligation; CALCOCO2: calcium binding and coiled-coil domain 2; CMA: chaperone-mediated autophagy; CTSB: cathepsin B; CTSD: cathepsin D; DGCR8: DGCR8 microprocessor complex subunit; DOCK2: dedicator of cyto-kinesis 2; EpiAdi: epididymal adipose tissue; HSPA8: heat shock protein family A (Hsp70) member 8; IHC: immunohistochemical; ISR: in-stent restenosis; KO: knockout; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; miRNA: microRNA; NLRP3: NLR family pyrin domain containing 3; N/L: ammonium chloride combined with leupeptin; PRMT: protein arginine methyltransferase; PVAT: peri-vascular adipose tissues; PYCARD: PYD and CARD domain containing; sDMA: symmetric dimethylarginine; ULK1: unc-51 like kinase 1; VSMCs: vascular smooth muscle cells; WT: wild-type.
Collapse
Affiliation(s)
- Jian Li
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Hongmin Yao
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Fujie Zhao
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Junqing An
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Qilong Wang
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Jing Mu
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Zhixue Liu
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Ming-Hui Zou
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Zhonglin Xie
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| |
Collapse
|
2
|
Poller W, Sahoo S, Hajjar R, Landmesser U, Krichevsky AM. Exploration of the Noncoding Genome for Human-Specific Therapeutic Targets-Recent Insights at Molecular and Cellular Level. Cells 2023; 12:2660. [PMID: 37998395 PMCID: PMC10670380 DOI: 10.3390/cells12222660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
While it is well known that 98-99% of the human genome does not encode proteins, but are nevertheless transcriptionally active and give rise to a broad spectrum of noncoding RNAs [ncRNAs] with complex regulatory and structural functions, specific functions have so far been assigned to only a tiny fraction of all known transcripts. On the other hand, the striking observation of an overwhelmingly growing fraction of ncRNAs, in contrast to an only modest increase in the number of protein-coding genes, during evolution from simple organisms to humans, strongly suggests critical but so far essentially unexplored roles of the noncoding genome for human health and disease pathogenesis. Research into the vast realm of the noncoding genome during the past decades thus lead to a profoundly enhanced appreciation of the multi-level complexity of the human genome. Here, we address a few of the many huge remaining knowledge gaps and consider some newly emerging questions and concepts of research. We attempt to provide an up-to-date assessment of recent insights obtained by molecular and cell biological methods, and by the application of systems biology approaches. Specifically, we discuss current data regarding two topics of high current interest: (1) By which mechanisms could evolutionary recent ncRNAs with critical regulatory functions in a broad spectrum of cell types (neural, immune, cardiovascular) constitute novel therapeutic targets in human diseases? (2) Since noncoding genome evolution is causally linked to brain evolution, and given the profound interactions between brain and immune system, could human-specific brain-expressed ncRNAs play a direct or indirect (immune-mediated) role in human diseases? Synergistic with remarkable recent progress regarding delivery, efficacy, and safety of nucleic acid-based therapies, the ongoing large-scale exploration of the noncoding genome for human-specific therapeutic targets is encouraging to proceed with the development and clinical evaluation of novel therapeutic pathways suggested by these research fields.
Collapse
Affiliation(s)
- Wolfgang Poller
- Department for Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum Charité (DHZC), Charité-Universitätsmedizin Berlin, 12200 Berlin, Germany;
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Site Berlin, 10785 Berlin, Germany
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA;
| | - Roger Hajjar
- Gene & Cell Therapy Institute, Mass General Brigham, 65 Landsdowne St, Suite 143, Cambridge, MA 02139, USA;
| | - Ulf Landmesser
- Department for Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum Charité (DHZC), Charité-Universitätsmedizin Berlin, 12200 Berlin, Germany;
- German Center for Cardiovascular Research (DZHK), Site Berlin, 10785 Berlin, Germany
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Anna M. Krichevsky
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
3
|
Nappi F, Avtaar Singh SS, Jitendra V, Alzamil A, Schoell T. The Roles of microRNAs in the Cardiovascular System. Int J Mol Sci 2023; 24:14277. [PMID: 37762578 PMCID: PMC10531750 DOI: 10.3390/ijms241814277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The discovery of miRNAs and their role in disease represent a significant breakthrough that has stimulated and propelled research on miRNAs as targets for diagnosis and therapy. Cardiovascular disease is an area where the restrictions of early diagnosis and conventional pharmacotherapy are evident and deserve attention. Therefore, miRNA-based drugs have significant potential for development. Research and its application can make considerable progress, as seen in preclinical and clinical trials. The use of miRNAs is still experimental but has a promising role in diagnosing and predicting a variety of acute coronary syndrome presentations. Its use, either alone or in combination with currently available biomarkers, might be adopted soon, particularly if there is diagnostic ambiguity. In this review, we examine the current understanding of miRNAs as possible targets for diagnosis and treatment in the cardiovascular system. We report on recent advances in recognising and characterising miRNAs with a focus on clinical translation. The latest challenges and perspectives towards clinical application are discussed.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (T.S.)
| | | | - Vikram Jitendra
- Department of Cardiothoracic Surgery, Aberdeen Royal Infirmary, Aberdeen AB25 2ZN, UK;
| | - Almothana Alzamil
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (T.S.)
| | - Thibaut Schoell
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.A.); (T.S.)
| |
Collapse
|
4
|
Zhao B, Lv Y. A biomechanical view of epigenetic tumor regulation. J Biol Phys 2023; 49:283-307. [PMID: 37004697 PMCID: PMC10397176 DOI: 10.1007/s10867-023-09633-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/12/2023] [Indexed: 04/04/2023] Open
Abstract
The occurrence and development of tumors depend on a complex regulation by not only biochemical cues, but also biomechanical factors in tumor microenvironment. With the development of epigenetic theory, the regulation of biomechanical stimulation on tumor progress genetically is not enough to fully illustrate the mechanism of tumorigenesis. However, biomechanical regulation on tumor progress epigenetically is still in its infancy. Therefore, it is particularly important to integrate the existing relevant researches and develop the potential exploration. This work sorted out the existing researches on the regulation of tumor by biomechanical factors through epigenetic means, which contains summarizing the tumor epigenetic regulatory mode by biomechanical factors, exhibiting the influence of epigenetic regulation under mechanical stimulation, illustrating its existing applications, and prospecting the potential. This review aims to display the relevant knowledge through integrating the existing studies on epigenetic regulation in tumorigenesis under mechanical stimulation so as to provide theoretical basis and new ideas for potential follow-up research and clinical applications. Mechanical factors under physiological conditions stimulate the tumor progress through epigenetic ways, and new strategies are expected to be found with the development of epidrugs and related delivery systems.
Collapse
Affiliation(s)
- Boyuan Zhao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, No. 1 Sunshine Avenue, Jiangxia District, Wuhan, Hubei Province, 430200, People's Republic of China.
| |
Collapse
|
5
|
Berger AG, Deiss-Yehiely E, Vo C, McCoy MG, Almofty S, Feinberg MW, Hammond PT. Electrostatically assembled wound dressings deliver pro-angiogenic anti-miRs preferentially to endothelial cells. Biomaterials 2023; 300:122188. [PMID: 37329684 PMCID: PMC10424785 DOI: 10.1016/j.biomaterials.2023.122188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/19/2023]
Abstract
Chronic non-healing wounds occur frequently in individuals affected by diabetes, yet standard-of-care treatment leaves many patients inadequately treated or with recurring wounds. MicroRNA (miR) expression is dysregulated in diabetic wounds and drives an anti-angiogenic phenotype, but miRs can be inhibited with short, chemically-modified RNA oligonucleotides (anti-miRs). Clinical translation of anti-miRs is hindered by delivery challenges such as rapid clearance and uptake by off-target cells, requiring repeated injections, excessively large doses, and bolus dosing mismatched to the dynamics of the wound healing process. To address these limitations, we engineered electrostatically assembled wound dressings that locally release anti-miR-92a, as miR-92a is implicated in angiogenesis and wound repair. In vitro, anti-miR-92a released from these dressings was taken up by cells and inhibited its target. An in vivo cellular biodistribution study in murine diabetic wounds revealed that endothelial cells, which play a critical role in angiogenesis, exhibit higher uptake of anti-miR eluted from coated dressings than other cell types involved in the wound healing process. In a proof-of-concept efficacy study in the same wound model, anti-miR targeting anti-angiogenic miR-92a de-repressed target genes, increased gross wound closure, and induced a sex-dependent increase in vascularization. Overall, this proof-of-concept study demonstrates a facile, translational materials approach for modulating gene expression in ulcer endothelial cells to promote angiogenesis and wound healing. Furthermore, we highlight the importance of probing cellular interactions between the drug delivery system and the target cells to drive therapeutic efficacy.
Collapse
Affiliation(s)
- Adam G Berger
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Elad Deiss-Yehiely
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Chau Vo
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael G McCoy
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Almofty
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Mark W Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
6
|
Dlouha D, Blaha M, Huckova P, Lanska V, Hubacek JA, Blaha V. Long-Term LDL-Apheresis Treatment and Dynamics of Circulating miRNAs in Patients with Severe Familial Hypercholesterolemia. Genes (Basel) 2023; 14:1571. [PMID: 37628623 PMCID: PMC10454435 DOI: 10.3390/genes14081571] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Lipoprotein apheresis (LA) is a therapeutic option for patients with severe hypercholesterolemia who have persistently elevated LDL-C levels despite attempts at drug therapy. MicroRNAs (miRNAs), important posttranscriptional gene regulators, are involved in the pathogenesis of atherosclerosis. Our study aimed to monitor the dynamics of twenty preselected circulating miRNAs in patients under long-term apheresis treatment. Plasma samples from 12 FH patients (men = 50%, age = 55.3 ± 12.2 years; mean LA overall treatment time = 13.1 ± 7.8 years) were collected before each apheresis therapy every sixth month over the course of four years of treatment. Eight complete follow-up (FU) samples were measured in each patient. Dynamic changes in the relative quantity of 6 miRNAs (miR-92a, miR-21, miR-126, miR-122, miR-26a, and miR-185; all p < 0.04) during FU were identified. Overall apheresis treatment time influenced circulating miR-146a levels (p < 0.04). In LDLR mutation homozygotes (N = 5), compared to heterozygotes (N = 7), we found higher plasma levels of miR-181, miR-126, miR-155, and miR-92a (all p < 0.03). Treatment with PCSK9 inhibitors (N = 6) affected the plasma levels of 7 miRNAs (miR-126, miR-122, miR-26a, miR-155, miR-125a, miR-92a, and miR-27a; all p < 0.04). Long-term monitoring has shown that LA in patients with severe familial hypercholesterolemia influences plasma circulating miRNAs involved in endothelial dysfunction, cholesterol homeostasis, inflammation, and plaque development. The longer the treatment using LA, the better the miRNA milieu depicting the potential cardiovascular risk.
Collapse
Affiliation(s)
- Dana Dlouha
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (P.H.); (J.A.H.)
| | - Milan Blaha
- 4th Department of Internal Medicine—Hematology, University Hospital Hradec Králové, 50005 Hradec Králové, Czech Republic;
- Faculty of Medicine in Hradec Králové, Charles University, 50003 Hradec Králové, Czech Republic;
| | - Pavlina Huckova
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (P.H.); (J.A.H.)
| | - Vera Lanska
- Statistical Unit, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic;
| | - Jaroslav Alois Hubacek
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (P.H.); (J.A.H.)
- 1st Faculty of Medicine, Charles University, 12108 Prague, Czech Republic
| | - Vladimir Blaha
- Faculty of Medicine in Hradec Králové, Charles University, 50003 Hradec Králové, Czech Republic;
- 3rd Department of Internal Medicine—Metabolism and Gerontology, University Hospital Hradec Králové, 50005 Hradec Králové, Czech Republic
| |
Collapse
|
7
|
Martín-Bórnez M, Falcón D, Morrugares R, Siegfried G, Khatib AM, Rosado JA, Galeano-Otero I, Smani T. New Insights into the Reparative Angiogenesis after Myocardial Infarction. Int J Mol Sci 2023; 24:12298. [PMID: 37569674 PMCID: PMC10418963 DOI: 10.3390/ijms241512298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/23/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Myocardial infarction (MI) causes massive loss of cardiac myocytes and injury to the coronary microcirculation, overwhelming the limited capacity of cardiac regeneration. Cardiac repair after MI is finely organized by complex series of procedures involving a robust angiogenic response that begins in the peri-infarcted border area of the infarcted heart, concluding with fibroblast proliferation and scar formation. Efficient neovascularization after MI limits hypertrophied myocytes and scar extent by the reduction in collagen deposition and sustains the improvement in cardiac function. Compelling evidence from animal models and classical in vitro angiogenic approaches demonstrate that a plethora of well-orchestrated signaling pathways involving Notch, Wnt, PI3K, and the modulation of intracellular Ca2+ concentration through ion channels, regulate angiogenesis from existing endothelial cells (ECs) and endothelial progenitor cells (EPCs) in the infarcted heart. Moreover, cardiac repair after MI involves cell-to-cell communication by paracrine/autocrine signals, mainly through the delivery of extracellular vesicles hosting pro-angiogenic proteins and non-coding RNAs, as microRNAs (miRNAs). This review highlights some general insights into signaling pathways activated under MI, focusing on the role of Ca2+ influx, Notch activated pathway, and miRNAs in EC activation and angiogenesis after MI.
Collapse
Affiliation(s)
- Marta Martín-Bórnez
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Débora Falcón
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Rosario Morrugares
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
- Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, 14071 Córdoba, Spain
| | - Geraldine Siegfried
- RyTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615 Pessac, France (A.-M.K.)
| | - Abdel-Majid Khatib
- RyTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615 Pessac, France (A.-M.K.)
| | - Juan A. Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003 Caceres, Spain;
| | - Isabel Galeano-Otero
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain; (M.M.-B.); (D.F.); (R.M.)
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
| |
Collapse
|
8
|
Future regenerative medicine developments and their therapeutic applications. Biomed Pharmacother 2023; 158:114131. [PMID: 36538861 DOI: 10.1016/j.biopha.2022.114131] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Although the currently available pharmacological assays can cure most pathological disorders, they have limited therapeutic value in relieving certain disorders like myocardial infarct, peripheral vascular disease, amputated limbs, or organ failure (e.g. renal failure). Pilot studies to overcome such problems using regenerative medicine (RM) delivered promising data. Comprehensive investigations of RM in zebrafish or reptilians are necessary for better understanding. However, the precise mechanisms remain poorly understood despite the tremendous amount of data obtained using the zebrafish model investigating the exact mechanisms behind their regenerative capability. Indeed, understanding such mechanisms and their application to humans can save millions of lives from dying due to potentially life-threatening events. Recent studies have launched a revolution in replacing damaged human organs via different approaches in the last few decades. The newly established branch of medicine (known as Regenerative Medicine aims to enhance natural repair mechanisms. This can be done through the application of several advanced broad-spectrum technologies such as organ transplantation, tissue engineering, and application of Scaffolds technology (support vascularization using an extracellular matrix), stem cell therapy, miRNA treatment, development of 3D mini-organs (organoids), and the construction of artificial tissues using nanomedicine and 3D bio-printers. Moreover, in the next few decades, revolutionary approaches in regenerative medicine will be applied based on artificial intelligence and wireless data exchange, soft intelligence biomaterials, nanorobotics, and even living robotics capable of self-repair. The present work presents a comprehensive overview that summarizes the new and future advances in the field of RM.
Collapse
|
9
|
Samak M, Kues A, Kaltenborn D, Klösener L, Mietsch M, Germena G, Hinkel R. Dysregulation of Krüppel-like Factor 2 and Myocyte Enhancer Factor 2D Drive Cardiac Microvascular Inflammation and Dysfunction in Diabetes. Int J Mol Sci 2023; 24:2482. [PMID: 36768805 PMCID: PMC9916909 DOI: 10.3390/ijms24032482] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Cardiovascular complications are the main cause of morbidity and mortality from diabetes. Herein, vascular inflammation is a major pathological manifestation. We previously characterized the cardiac microvascular inflammatory phenotype in diabetic patients and highlighted micro-RNA 92a (miR-92a) as a driver of endothelial dysfunction. In this article, we further dissect the molecular underlying of these findings by addressing anti-inflammatory Krüppel-like factors 2 and 4 (KLF2 and KLF4). We show that KLF2 dysregulation in diabetes correlates with greater monocyte adhesion as well as migratory defects in cardiac microvascular endothelial cells. We also describe, for the first time, a role for myocyte enhancer factor 2D (MEF2D) in cardiac microvascular dysfunction in diabetes. We show that both KLFs 2 and 4, as well as MEF2D, are dysregulated in human and porcine models of diabetes. Furthermore, we prove a direct interaction between miR-92a and all three targets. Altogether, our data strongly qualify miR-92a as a potential therapeutic target for diabetes-associated cardiovascular disease.
Collapse
Affiliation(s)
- Mostafa Samak
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Andreas Kues
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
| | - Diana Kaltenborn
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
| | - Lina Klösener
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine, 30173 Hannover, Germany
| | - Matthias Mietsch
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Giulia Germena
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, Leibniz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Institute for Animal Hygiene, Animal Welfare and Farm Animal Behaviour, University of Veterinary Medicine, 30173 Hannover, Germany
| |
Collapse
|
10
|
Shaharyar MA, Bhowmik R, Al-Abbasi FA, AlGhamdi SA, Alghamdi AM, Sarkar A, Kazmi I, Karmakar S. Vaccine Formulation Strategies and Challenges Involved in RNA Delivery for Modulating Biomarkers of Cardiovascular Diseases: A Race from Laboratory to Market. Vaccines (Basel) 2023; 11:vaccines11020241. [PMID: 36851119 PMCID: PMC9963957 DOI: 10.3390/vaccines11020241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
It has been demonstrated that noncoding RNAs have significant physiological and pathological roles. Modulation of noncoding RNAs may offer therapeutic approaches as per recent findings. Small RNAs, mostly long noncoding RNAs, siRNA, and microRNAs make up noncoding RNAs. Inhibiting or promoting protein breakdown by binding to 3' untranslated regions of target mRNA, microRNAs post-transcriptionally control the pattern of gene expression. Contrarily, long non-coding RNAs perform a wider range of tasks, including serving as molecular scaffolding, decoys, and epigenetic regulators. This article provides instances of long noncoding RNAs and microRNAs that may be a biomarker of CVD (cardiovascular disease). In this paper we highlight various RNA-based vaccine formulation strategies designed to target these biomarkers-that are either currently in the research pipeline or are in the global pharmaceutical market-along with the physiological hurdles that need to be overcome.
Collapse
Affiliation(s)
- Md. Adil Shaharyar
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Rudranil Bhowmik
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shareefa A. AlGhamdi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amira M. Alghamdi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Arnab Sarkar
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (I.K.); (S.K.); Tel.: +966-543970731 (I.K.); +91-8017136385 (S.K.)
| | - Sanmoy Karmakar
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
- Correspondence: (I.K.); (S.K.); Tel.: +966-543970731 (I.K.); +91-8017136385 (S.K.)
| |
Collapse
|
11
|
Tang Z, Song J, Yu Z, Cui K, Ruan Y, Liu Y, Wang T, Wang S, Liu J, Yang J. Inhibition of MicroRNA-92a Improved Erectile Dysfunction in Streptozotocin-Induced Diabetic Rats via Suppressing Oxidative Stress and Endothelial Dysfunction. World J Mens Health 2023; 41:142-154. [PMID: 35274504 PMCID: PMC9826907 DOI: 10.5534/wjmh.210177] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/17/2021] [Accepted: 12/08/2021] [Indexed: 01/21/2023] Open
Abstract
PURPOSE To determine whether microRNA could be a therapy target of erectile dysfunction (ED) and the underlying mechanisms. MATERIALS AND METHODS Eight-week-old fasting male SD rats were intraperitoneally injected with streptozotocin to construct diabetic rat models. Diabetic ED rats were treated with miRNA-92a inhibitor. The cavernous nerves were electrically stimulated to measure the intracavernous pressure and mean arterial pressure of rats in each group. After the detection, the penile cavernous tissues are properly stored for subsequent experiments. Rat aortic endothelial cells were used in in vitro studies. RESULTS The expression of miR-92a was significantly increased in the corpus cavernosum of Streptozocin (STZ)-induced diabetic rats and injection of miR-92a antagomir into the corpus cavernosum of diabetic rats significantly increased eNOS/NO/cGMP signaling pathway activities, cavernous endothelial cell proliferation, endothelial cell-cell junction protein expression and decreased the levels of oxidative stress. These changes restored erectile function in STZ-induced diabetic rats. Moreover, in vitro study demonstrated that the miR-92a expression increased significantly in endothelial cells treated with high glucose, inhibiting AMPK/eNOS and AMPK/Nrf2/HO-1 signaling pathways in rat aortic endothelial cells via targeting Prkaa2, causing endothelial dysfunction and overactive oxidative stress, miR-92a inhibitor can improve the above parameters. CONCLUSIONS miRNA-92a inhibitor could exert an inhibition role on oxidative stress and endothelial dysfunction to improve diabetic ED effectively.
Collapse
Affiliation(s)
- Zhe Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingyu Song
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhe Yu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Cui
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yajun Ruan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
12
|
Innate Immunity in Cardiovascular Diseases-Identification of Novel Molecular Players and Targets. J Clin Med 2023; 12:jcm12010335. [PMID: 36615135 PMCID: PMC9821340 DOI: 10.3390/jcm12010335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
During the past few years, unexpected developments have driven studies in the field of clinical immunology. One driver of immense impact was the outbreak of a pandemic caused by the novel virus SARS-CoV-2. Excellent recent reviews address diverse aspects of immunological re-search into cardiovascular diseases. Here, we specifically focus on selected studies taking advantage of advanced state-of-the-art molecular genetic methods ranging from genome-wide epi/transcriptome mapping and variant scanning to optogenetics and chemogenetics. First, we discuss the emerging clinical relevance of advanced diagnostics for cardiovascular diseases, including those associated with COVID-19-with a focus on the role of inflammation in cardiomyopathies and arrhythmias. Second, we consider newly identified immunological interactions at organ and system levels which affect cardiovascular pathogenesis. Thus, studies into immune influences arising from the intestinal system are moving towards therapeutic exploitation. Further, powerful new research tools have enabled novel insight into brain-immune system interactions at unprecedented resolution. This latter line of investigation emphasizes the strength of influence of emotional stress-acting through defined brain regions-upon viral and cardiovascular disorders. Several challenges need to be overcome before the full impact of these far-reaching new findings will hit the clinical arena.
Collapse
|
13
|
The expression profiling of serum miR-92a, miR-134 and miR-375 in acute ischemic stroke. Future Sci OA 2022; 8:FSO829. [PMID: 36874371 PMCID: PMC9979103 DOI: 10.2144/fsoa-2022-0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/17/2023] [Indexed: 02/22/2023] Open
Abstract
Aim To investigate the expression profile and diagnostic potentials of serum miR-92a, 134, and 375 in acute ischemic stroke (AIS) patients. Materials & methods Serum miRs-92a, 134, and 375 expression profiles were estimated by qRT-PCR for 70 AIS patients, age-matched with 25 control subjects. Their diagnostic potential was estimated by ROC analysis. Results Down-expression of miR-92a and miR-375 was found (56; 96.5%; -1.86 ± 1.36; and 53; 91.4%; -1.63 ± 1.38, respectively), while miR-134 showed a predominant upregulation (46; 79.3%; 0.853 ± 1.34). The diagnostic accuracy was the highest for miR-92a and miR-375 (area under the curve = 0.9183 and 0.898, respectively), with greater specificity for miR-375 (Sp = 96%). Conclusion Serum miR-92a and miR-375 could be promising early detective biomarkers of AIS.
Collapse
|
14
|
Shah AM, Giacca M. Small non-coding RNA therapeutics for cardiovascular disease. Eur Heart J 2022; 43:4548-4561. [PMID: 36106499 PMCID: PMC9659475 DOI: 10.1093/eurheartj/ehac463] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/29/2022] [Accepted: 08/11/2022] [Indexed: 01/07/2023] Open
Abstract
Novel bio-therapeutic agents that harness the properties of small, non-coding nucleic acids hold great promise for clinical applications. These include antisense oligonucleotides that inhibit messenger RNAs, microRNAs (miRNAs), or long non-coding RNAs; positive effectors of the miRNA pathway (short interfering RNAs and miRNA mimics); or small RNAs that target proteins (i.e. aptamers). These new therapies also offer exciting opportunities for cardiovascular diseases and promise to move the field towards more precise approaches based on disease mechanisms. There have been substantial advances in developing chemical modifications to improve the in vivo pharmacological properties of antisense oligonucleotides and reduce their immunogenicity. Carrier methods (e.g. RNA conjugates, polymers, and lipoplexes) that enhance cellular uptake of RNA therapeutics and stability against degradation by intracellular nucleases are also transforming the field. A number of small non-coding RNA therapies for cardiovascular indications are now approved. Moreover, there is a large pipeline of therapies in clinical development and an even larger list of putative therapies emerging from pre-clinical studies. Progress in this area is reviewed herein along with the hurdles that need to be overcome to allow a broader clinical translation.
Collapse
Affiliation(s)
- Ajay M Shah
- King’s College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Mauro Giacca
- King’s College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
15
|
Cui Y, Liu R, Hong Y, Wang Y, Zhu Y, Wen T, Lu J, Mao S, Wang X, Pan J, Luo Y. MicroRNA-92a-3p Regulates Retinal Angiogenesis by Targeting SGK3 in Vascular Endothelial Cells. Invest Ophthalmol Vis Sci 2022; 63:19. [PMID: 36269185 PMCID: PMC9617502 DOI: 10.1167/iovs.63.11.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose The purpose of this study was to investigate the effects and mechanism of microRNA (miR)-92a-3p in retinal angiogenesis in vitro and in vivo. Methods The expression of miR-92a-3p was verified by real-time quantitative polymerase chain reaction (RT-qPCR). Agomir-92a-3p was intravitreally injected into the right eye on postnatal day 3 (P3), P5, and P8 in the mice, with the agomir-NC injected left eye as the control. At P7, P9, and P12, immunofluorescence was performed to examine the retinal superficial vascular plexus, deep vascular plexus, proliferation, and apoptosis in retinal vascular endothelial cells (ECs). Human retinal microvascular endothelial cells (HRMECs) were treated with mimic-NC and mimic-92a-3p, then the tube formation, cell migration, and wound healing assays were used to detect the effect of miR-92a-3p on retinal angiogenesis in vitro. Agomir-92a-3p was also intravitreally injected into the right eye of oxygen-induced retinopathy (OIR) mice at P12, with the agomir-NC injected left eye as the control, the neovascularization was observed by retinal flatmount staining with isolectin B4 at P17. Bioinformatics and high-throughput sequencing were performed to identify potential target genes of miR-92a-3p. RT-qPCR and Western blot were carried out to detect the expression of SGK3, p-GSK3β, GSK3β, Bcl-xL, and cleaved caspase-3 in the HRMECs and mouse retinas. Results The overexpression of miR-92a-3p inhibited the development of retinal superficial vascular plexus and deep vascular plexus, decreased the expression of Ki67, and increased the expression of cleaved caspase-3 in isolectin B4-labeled retinal vascular ECs. In vitro, the overexpression of miR-92a-3p markedly suppressed the tube formation, cell migration, and wound healing of cultured ECs. Overexpression of miR-92a-3p inhibited both in vivo and in vitro physiological angiogenesis by downregulating the expression of SGK3, p-GSK3β/GSK3β, and Bcl-xL. In addition, agomir-92a-3p inhibited the pathological retinal neovascularization of OIR mice, by targeting SGK3, p-GSK3β/GSK3β, and Bcl-xL. Conclusions The miR-92a-3p could affect retinal angiogenesis by targeting SGK3 pathway, suggesting that miR-92a-3p may be a potential anti-angiogenic factor for retinal vascular disease.
Collapse
Affiliation(s)
- Yamei Cui
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ruyuan Liu
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yiwen Hong
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yishen Wang
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanjie Zhu
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Wen
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Lu
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shudi Mao
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao Wang
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianying Pan
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Luo
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
miR-100-5p Promotes Epidermal Stem Cell Proliferation through Targeting MTMR3 to Activate PIP3/AKT and ERK Signaling Pathways. Stem Cells Int 2022; 2022:1474273. [PMID: 36045954 PMCID: PMC9421352 DOI: 10.1155/2022/1474273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/22/2022] [Accepted: 08/02/2022] [Indexed: 12/04/2022] Open
Abstract
Skin epidermal stem cells (EpSCs) play a critical role in wound healing and are ideal seed cells for skin tissue engineering. Exosomes from human adipose-derived stem cells (ADSC-Exos) promote human EpSC proliferation, but the underlying mechanism remains unclear. Here, we investigated the effect of miR-100-5p, one of the most abundant miRNAs in ADSC-Exos, on the proliferation of human EpSCs and explored the mechanisms involved. MTT and BrdU incorporation assays showed that miR-100-5p mimic transfection promoted EpSC proliferation in a time-dependent manner. Cell cycle analysis showed that miR-100-5p mimic transfection significantly decreased the percentage of cells in the G1 phase and increased the percentage of cells in the G2/M phase. Myotubularin-related protein 3 (MTMR3), a lipid phosphatase, was identified as a direct target of miR-100-5p. Knockdown of MTMR3 in EpSCs by RNA interference significantly enhanced cell proliferation, decreased the percentage of cells in the G1 phase and increased the percentage of cells in the S phase. Overexpression of MTMR3 reversed the proproliferative effect of miR-100-5p on EpSCs, indicating that miR-100-5p promoted EpSC proliferation by downregulating MTMR3. Mechanistic studies showed that transfection of EpSCs with miR-100-5p mimics elevated the intracellular PIP3 level, induced AKT and ERK phosphorylation, and upregulated cyclin D1, E1, and A2 expression, which could be attenuated by MTMR3 overexpression. Consistently, intradermal injection of ADSC-Exos or miR-100-5p-enriched ADSC-Exos into cultured human skin tissues significantly reduced MTMR3 expression and increased the thickness of the epidermis and the number of EpSCs in the basal layer of the epidermis. The aforementioned effect of miR-100-5p-enriched ADSC-Exos was stronger than that of ADSC-Exos and was reversed by MTMR3 overexpression. Collectively, our findings indicate that miR-100-5p promotes EpSC proliferation through MTMR3-mediated elevation of PIP3 and activation of AKT and ERK. miR-100-5p-enriched ADSC-Exos can be used to treat skin wound and expand EpSCs for generating epidermal autografts and engineered skin equivalents.
Collapse
|
17
|
Sufianov A, Begliarzade S, Kudriashov V, Nafikova R, Ilyasova T, Liang Y. Role of miRNAs in vascular development. Noncoding RNA Res 2022; 8:1-7. [PMID: 36262425 PMCID: PMC9552023 DOI: 10.1016/j.ncrna.2022.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/27/2022] Open
|
18
|
Hu X, Ning X, Zhao Q, Zhang Z, Zhang C, Xie M, Huang W, Cai Y, Xiang Q, Ou C. Islet-1 Mesenchymal Stem Cells-Derived Exosome-Incorporated Angiogenin-1 Hydrogel for Enhanced Acute Myocardial Infarction Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:36289-36303. [PMID: 35920579 DOI: 10.1021/acsami.2c04686] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Although stem cell-derived exosomes have been recognized as new candidates for cell-free treatment in myocardial infarction (MI), the challenge to improve the exosome retention in ischemic tissue remains. Our previous research indicated that islet-1(ISL1) overexpression enhances the paracrine function of mesenchymal stem cells (MSCs) and promotes angiogenesis in a model of MI. In this study, genetically engineered ISL1-MSC-derived exosomes (ISL1-MSCs-Exo) were collected, and the contents were analyzed by exosomal RNA sequencing. Next, we investigated if ISL1-MSCs-Exo could exert therapeutic effects and their incorporation into a new angiogenin-1 hydrogel (Ang-1 gel) could boost the retention of exosomes and further enhance their protective effects. Our results demonstrated that ISL1-MSCs-Exo could play a therapeutic role in vitro and in vivo, which might be due to changed exosomal contents. Ang-1 gel increased the retention and enhanced the anti-apoptosis, proliferation, and angiogenic capacity of ISL1-MSCs-Exo in endothelial cells. Echocardiography revealed that Ang-1 gel significantly augment the therapeutic effects of ISL1-MSCs-Exo for MI. The main mechanism might result from increased retention of ISL1-MSCs-Exo, herein enhanced pro-angiogenetic effects in an ischemic heart. Taken together, our findings indicated that ISL1-MSCs-Exo had endothelium-protective and pro-angiogenic abilities alone and Ang-1 gel could notably retain ISL1-MSCs-Exo at ischemic sites, which improved the survival and angiogenesis of endothelial cells and accelerated the recovery of MI. These results not only shed light on the therapeutic mechanism of ISL1-MSCs-Exo incorporated with Ang-1 gel but also offer a promising therapeutic option for ischemic disease.
Collapse
Affiliation(s)
- Xinyi Hu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Xiaodong Ning
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Qianqian Zhao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhen Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chi Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Manting Xie
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yanbin Cai
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Qiuling Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Caiwen Ou
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Dongguan Hospital of Southern Medical University, Southern Medical University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
19
|
Decoding microRNA drivers in Atherosclerosis. Biosci Rep 2022; 42:231479. [PMID: 35758143 PMCID: PMC9289798 DOI: 10.1042/bsr20212355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/17/2022] [Accepted: 06/26/2022] [Indexed: 11/17/2022] Open
Abstract
An estimated 97% of the human genome consists of non-protein-coding sequences. As our understanding of genome regulation improves, this has led to the characterization of a diverse array of non-coding RNAs (ncRNA). Among these, micro-RNAs (miRNAs) belong to the short ncRNA class (22–25 nucleotides in length), with approximately 2500 miRNA genes encoded within the human genome. From a therapeutic perspective, there is interest in exploiting miRNA as biomarkers of disease progression and response to treatments, as well as miRNA mimics/repressors as novel medicines. miRNA have emerged as an important class of RNA master regulators with important roles identified in the pathogenesis of atherosclerotic cardiovascular disease. Atherosclerosis is characterized by a chronic inflammatory build-up, driven largely by low-density lipoprotein cholesterol accumulation within the artery wall and vascular injury, including endothelial dysfunction, leukocyte recruitment and vascular remodelling. Conventional therapy focuses on lifestyle interventions, blood pressure-lowering medications, high-intensity statin therapy and antiplatelet agents. However, a significant proportion of patients remain at increased risk of cardiovascular disease. This continued cardiovascular risk is referred to as residual risk. Hence, a new drug class targeting atherosclerosis could synergise with existing therapies to optimise outcomes. Here, we review our current understanding of the role of ncRNA, with a focus on miRNA, in the development and progression of atherosclerosis, highlighting novel biological mechanisms and therapeutic avenues.
Collapse
|
20
|
Abstract
The discovery of microRNAs and their role in diseases was a breakthrough that inspired research into microRNAs as drug targets. Cardiovascular diseases are an area in which limitations of conventional pharmacotherapy are highly apparent and where microRNA-based drugs have appreciably progressed into preclinical and clinical testing. In this Review, we summarize the current state of microRNAs as therapeutic targets in the cardiovascular system. We report recent advances in the identification and characterization of microRNAs, their manipulation and clinical translation, and discuss challenges and perspectives toward clinical application.
Collapse
Affiliation(s)
- Bernhard Laggerbauer
- Institute of Pharmacology and Toxicology, Technical University of Munich (TUM), Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich (TUM), Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
21
|
Utilization and Potential of RNA-Based Therapies in Cardiovascular Disease. JACC Basic Transl Sci 2022; 7:956-969. [PMID: 36317129 PMCID: PMC9617127 DOI: 10.1016/j.jacbts.2022.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
Abstract
RNA-based therapeutics have the potential to reach previously “undruggable” pathways in cardiovascular disease RNA-based therapeutics constitute a vast array of technologies, including unique forms, chemistries, and modalities of delivery Rapid development of RNA-based vaccines was made possible by decades of foundational work Specificity and efficacy of targeting and determination of mechanism(s) of action remain a distinct challenge
Cardiovascular disease (CVD) remains the largest cause of mortality worldwide. The development of new effective therapeutics is a major unmet need. The current review focuses broadly on the concept of nucleic acid (NA)–based therapies, considering the use of various forms of NAs, including mRNAs, miRNAs, siRNA, and guide RNAs, the latter specifically for the purpose of CRISPR-Cas directed gene editing. We describe the current state-of-the-art of RNA target discovery and development, the status of RNA therapeutics in the context of CVD, and some of the challenges and hurdles to be overcome.
Collapse
|
22
|
Zhou X, Ye Q, Zheng J, Kuang L, Zhu J, Yan H. IMP3 promotes re-endothelialization after arterial injury via increasing stability of VEGF mRNAhv. J Cell Mol Med 2022; 26:2023-2037. [PMID: 35315195 PMCID: PMC8980943 DOI: 10.1111/jcmm.17225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 01/01/2022] [Accepted: 01/05/2022] [Indexed: 11/30/2022] Open
Abstract
IMP3, an RNA‐binding protein (RBP) that participates in the process of post‐transcriptional modifications of mRNA transcripts, is capable of altering cellular functions, and in some cases, be involved in specific disease progression. We aimed to investigate whether IMP3 has the ability to regulate the functional properties of endothelial cells and re‐endothelialization in response to arterial injury. Wire injury was introduced to the right carotid arteries of wildtype C57/BL6 mice. As a result, IMPs’ expressions were up‐regulated in the induced arterial lesions, and IMP3 was the most up‐regulated RNA among other IMPs. We overexpressed IMP3 before the wire‐injured surgery using adeno‐associated virus AAV2‐IMP3. In vivo studies confirmed that IMP3 overexpression accelerated the progress of re‐endothelialization after arterial injury. In vitro, endothelial cells were transfected with either ad‐IMP3 or Si‐IMP3, cell functional studies showed that IMP3 could promote endothelial cell proliferation and migration, while reducing apoptosis. Mechanistic studies also revealed that IMP3 could enhance VEGF mRNA stability and therefore up‐regulate activities of VEGF/PI3K/Akt signalling pathway. Our data indicated that IMP3 promotes re‐endothelialization after arterial injury and regulates endothelial cell proliferation, migration and apoptosis via increasing stability of VEGF mRNA and activation of VEGF/PI3K/Akt signalling pathway.
Collapse
Affiliation(s)
- Xinmiao Zhou
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qingqing Ye
- Department of Intensive Care Unit, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jinlei Zheng
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Kuang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianhua Zhu
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Yan
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
23
|
De Rosa S, Iaconetti C, Eyileten C, Yasuda M, Albanese M, Polimeni A, Sabatino J, Sorrentino S, Postula M, Indolfi C. Flow-Responsive Noncoding RNAs in the Vascular System: Basic Mechanisms for the Clinician. J Clin Med 2022; 11:jcm11020459. [PMID: 35054151 PMCID: PMC8777617 DOI: 10.3390/jcm11020459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/10/2022] Open
Abstract
The vascular system is largely exposed to the effect of changing flow conditions. Vascular cells can sense flow and its changes. Flow sensing is of pivotal importance for vascular remodeling. In fact, it influences the development and progression of atherosclerosis, controls its location and has a major influx on the development of local complications. Despite its importance, the research community has traditionally paid scarce attention to studying the association between different flow conditions and vascular biology. More recently, a growing body of evidence has been accumulating, revealing that ncRNAs play a key role in the modulation of several biological processes linking flow-sensing to vascular pathophysiology. This review summarizes the most relevant evidence on ncRNAs that are directly or indirectly responsive to flow conditions to the benefit of the clinician, with a focus on the underpinning mechanisms and their potential application as disease biomarkers.
Collapse
Affiliation(s)
- Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
- Correspondence: (S.D.R.); (C.I.)
| | - Claudio Iaconetti
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CePT, Medical University of Warsaw, 02-097 Warsaw, Poland; (C.E.); (M.P.)
| | - Masakazu Yasuda
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Michele Albanese
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Alberto Polimeni
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Jolanda Sabatino
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Sabato Sorrentino
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CePT, Medical University of Warsaw, 02-097 Warsaw, Poland; (C.E.); (M.P.)
| | - Ciro Indolfi
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (C.I.); (M.Y.); (M.A.); (A.P.); (J.S.); (S.S.)
- Mediterranea Cardiocentro, 80122 Naples, Italy
- Correspondence: (S.D.R.); (C.I.)
| |
Collapse
|
24
|
Wang X, Gao B, Feng Y. Recent advances in inhibiting atherosclerosis and restenosis: from pathogenic factors, therapeutic agents to nano-delivery strategies. J Mater Chem B 2022; 10:1685-1708. [DOI: 10.1039/d2tb00003b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Due to dominant atherosclerosis etiology, cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide. In clinical trials, advanced atherosclerotic plaques can be removed by angioplasty and vascular...
Collapse
|
25
|
Zhou Z, Yeh CF, Mellas M, Oh MJ, Zhu J, Li J, Huang RT, Harrison DL, Shentu TP, Wu D, Lueckheide M, Carver L, Chung EJ, Leon L, Yang KC, Tirrell MV, Fang Y. Targeted polyelectrolyte complex micelles treat vascular complications in vivo. Proc Natl Acad Sci U S A 2021; 118:e2114842118. [PMID: 34880134 PMCID: PMC8685925 DOI: 10.1073/pnas.2114842118] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2021] [Indexed: 01/09/2023] Open
Abstract
Vascular disease is a leading cause of morbidity and mortality in the United States and globally. Pathological vascular remodeling, such as atherosclerosis and stenosis, largely develop at arterial sites of curvature, branching, and bifurcation, where disturbed blood flow activates vascular endothelium. Current pharmacological treatments of vascular complications principally target systemic risk factors. Improvements are needed. We previously devised a targeted polyelectrolyte complex micelle to deliver therapeutic nucleotides to inflamed endothelium in vitro by displaying the peptide VHPKQHR targeting vascular cell adhesion molecule 1 (VCAM-1) on the periphery of the micelle. This paper explores whether this targeted nanomedicine strategy effectively treats vascular complications in vivo. Disturbed flow-induced microRNA-92a (miR-92a) has been linked to endothelial dysfunction. We have engineered a transgenic line (miR-92aEC-TG /Apoe-/- ) establishing that selective miR-92a overexpression in adult vascular endothelium causally promotes atherosclerosis in Apoe-/- mice. We tested the therapeutic effectiveness of the VCAM-1-targeting polyelectrolyte complex micelles to deliver miR-92a inhibitors and treat pathological vascular remodeling in vivo. VCAM-1-targeting micelles preferentially delivered miRNA inhibitors to inflamed endothelial cells in vitro and in vivo. The therapeutic effectiveness of anti-miR-92a therapy in treating atherosclerosis and stenosis in Apoe-/- mice is markedly enhanced by the VCAM-1-targeting polyelectrolyte complex micelles. These results demonstrate a proof of concept to devise polyelectrolyte complex micelle-based targeted nanomedicine approaches treating vascular complications in vivo.
Collapse
Affiliation(s)
- Zhengjie Zhou
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Chih-Fan Yeh
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Michael Mellas
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
| | - Myung-Jin Oh
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jiayu Zhu
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jin Li
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Ru-Ting Huang
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Devin L Harrison
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637
| | - Tzu-Pin Shentu
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - David Wu
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Michael Lueckheide
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
| | - Lauryn Carver
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Eun Ji Chung
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
| | - Lorraine Leon
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
| | - Kai-Chien Yang
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Matthew V Tirrell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637;
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637
- Materials Science Division, Argonne National Laboratory, Lemont, IL 60439
| | - Yun Fang
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637;
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637
| |
Collapse
|
26
|
Zhou Y, Wei W, Shen J, Lu L, Lu T, Wang H, Xue X. Alisol A 24-acetate protects oxygen-glucose deprivation-induced brain microvascular endothelial cells against apoptosis through miR-92a-3p inhibition by targeting the B-cell lymphoma-2 gene. PHARMACEUTICAL BIOLOGY 2021; 59:513-524. [PMID: 33905668 PMCID: PMC8081307 DOI: 10.1080/13880209.2021.1912117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
CONTEXT Alisol A 24-acetate has been used to treat vascular diseases. However, the underlying mechanisms still remain unclear. OBJECTIVE The present study evaluated the antiapoptotic effect of alisol A 24-acetate on brain microvascular endothelial cells (BMECs) and explored the underlying mechanisms. MATERIALS AND METHODS BMECs were injured through oxygen -glucose deprivation (OGD) after alisol A 24-acetate treatment. Cell viability and half-maximal inhibitory concentration (IC50) were measured using CCK-8, whereas inflammatory factors and oxidative stress indicators were measured using enzyme linked immunosorbent assay. Cell invasion and wound healing assays were detected. Cell apoptosis was assessed using flow cytometry. B-cell lymphoma-2 (Bcl-2) and Bcl-2 associated X (Bax) expression were analyzed using Western blotting. Dual-luciferase assay was applied to detect target genes of miR-92a-3p. RESULT Alisol A 24-acetate had an IC50 of 98.53 mg/L and inhibited cell viability at concentrations over 50mg/L. OGD induced apoptosis and promoted miR-92a-3p overexpression in BMECs. However, alisol A 24-acetate treatment suppressed inflammation, improved migration and invasion abilities, increased Bcl-2 expression, inhibited Bax expression, and repressed apoptosis and miR92a-3p overexpression in OGD-induced BMECs. MiR-92a-3p overexpression promoted cell apoptosis and suppressed Bcl-2 expression, whereas its inhibitor reversed the tendency. Alisol A 24-acetate treatment relieved the effects of miR-92a-3p overexpression. Dual-luciferase assay confirmed that miR-92a-3p negatively regulated the Bcl-2 expression. CONCLUSIONS These findings suggest that alisol A 24-acetate exerts antiapoptotic effects on OGD-induced BMECs through miR-92a-3p inhibition by targeting the Bcl-2 gene, indicating its potential for BMECs protection and as a novel therapeutic agent for the treatment of cerebrovascular disease.
Collapse
Affiliation(s)
- Yangjie Zhou
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Wei Wei
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Julian Shen
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lu Lu
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Taotao Lu
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hong Wang
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiehua Xue
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- CONTACT Xiehua Xue No. 13, Hudongzhi Road, Gulou District, Fuzhou350122, China
| |
Collapse
|
27
|
Darrah KE, Deiters A. Translational control of gene function through optically regulated nucleic acids. Chem Soc Rev 2021; 50:13253-13267. [PMID: 34739027 DOI: 10.1039/d1cs00257k] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Translation of mRNA into protein is one of the most fundamental processes within biological systems. Gene expression is tightly regulated both in space and time, often involving complex signaling or gene regulatory networks, as most prominently observed in embryo development. Thus, studies of gene function require tools with a matching level of external control. Light is an excellent conditional trigger as it is minimally invasive, can be easily tuned in wavelength and amplitude, and can be applied with excellent spatial and temporal resolution. To this end, modification of established oligonucleotide-based technologies with optical control elements, in the form of photocaging groups and photoswitches, has rendered these tools capable of navigating the dynamic regulatory pathways of mRNA translation in cellular and in vivo models. In this review, we discuss the different optochemical approaches used to generate photoresponsive nucleic acids that activate and deactivate gene expression and function at the translational level.
Collapse
Affiliation(s)
- Kristie E Darrah
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA.
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA.
| |
Collapse
|
28
|
Abhange K, Makler A, Wen Y, Ramnauth N, Mao W, Asghar W, Wan Y. Small extracellular vesicles in cancer. Bioact Mater 2021; 6:3705-3743. [PMID: 33898874 PMCID: PMC8056276 DOI: 10.1016/j.bioactmat.2021.03.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EV) are lipid-bilayer enclosed vesicles in submicron size that are released from cells. A variety of molecules, including proteins, DNA fragments, RNAs, lipids, and metabolites can be selectively encapsulated into EVs and delivered to nearby and distant recipient cells. In tumors, through such intercellular communication, EVs can regulate initiation, growth, metastasis and invasion of tumors. Recent studies have found that EVs exhibit specific expression patterns which mimic the parental cell, providing a fingerprint for early cancer diagnosis and prognosis as well as monitoring responses to treatment. Accordingly, various EV isolation and detection technologies have been developed for research and diagnostic purposes. Moreover, natural and engineered EVs have also been used as drug delivery nanocarriers, cancer vaccines, cell surface modulators, therapeutic agents and therapeutic targets. Overall, EVs are under intense investigation as they hold promise for pathophysiological and translational discoveries. This comprehensive review examines the latest EV research trends over the last five years, encompassing their roles in cancer pathophysiology, diagnostics and therapeutics. This review aims to examine the full spectrum of tumor-EV studies and provide a comprehensive foundation to enhance the field. The topics which are discussed and scrutinized in this review encompass isolation techniques and how these issues need to be overcome for EV-based diagnostics, EVs and their roles in cancer biology, biomarkers for diagnosis and monitoring, EVs as vaccines, therapeutic targets, and EVs as drug delivery systems. We will also examine the challenges involved in EV research and promote a framework for catalyzing scientific discovery and innovation for tumor-EV-focused research.
Collapse
Affiliation(s)
- Komal Abhange
- The Pq Laboratory of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | - Amy Makler
- Micro and Nanotechnology in Medicine, Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Yi Wen
- The Pq Laboratory of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | - Natasha Ramnauth
- Micro and Nanotechnology in Medicine, Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Wenjun Mao
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Waseem Asghar
- Micro and Nanotechnology in Medicine, Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Yuan Wan
- The Pq Laboratory of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY 13902, USA
| |
Collapse
|
29
|
Lu J, Fang Q, Ge X. Role and Mechanism of mir-5189-3p in Deep Vein Thrombosis of Lower Extremities. Ann Vasc Surg 2021; 77:288-295. [PMID: 34416282 DOI: 10.1016/j.avsg.2021.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND This study is to investigate the role and mechanism of mir-5189-3p in deep vein thrombosis (DVT) in lower extremity. METHODS The blood samples were collected from Kazakh patients with DVT in lower extremity and were subjected to microRNA sequencing. Bioinformatics were used to identify mir-5189-3p and its target genes. Dual luciferase reporter assay was used to determine the regulatory effect of mir-5189-3p on JAG1. SD rats were randomly divided into normal control, DVT model, hsa-miR-5189-3p mimics and hsa-miR-5189-3p negative control groups. HE staining was used to observe the pathological changes. TUNEL method was used to observe apoptosis. Western blot was used to detect Bax and Bcl-2 protein expression. Real-time quantitative PCR was used to detect JAG1, Notch1 and Hes1 mRNA. RESULTS The target of Has-miR-5189-3p was JAG1. Co-transfection of miR-5189-3p mimics and pmirGLO/JAG1 wild-type plasmid induced significantly decreased luciferase activity. In hsa-miR-5189-3p mimics and hsa-miR-5189-3p negative control groups, there were more nucleated cells in the thrombus tissues, and the organization degree obviously increased. Signs of blood flow recanalization were observed. The apoptosis of hsa-miR-5189-3p mimics and hsa-miR-5189-3p negative control groups was lower than that in DVT model group. Furthermore, mir-5189-3p mimics significantly increased the mRNA levels of JAG1, Notch1 and Hes1. Additionally, mir-5189-3p mimics significantly increased Bcl-2 while decreased Bax protein. CONCLUSIONS mir-5189-3p could inhibit apoptosis and promote thrombus organization in DVT possibly via Notch signaling pathway. Mir-5189-3p can be used as a potential target for DVT treatment.
Collapse
Affiliation(s)
- Jing Lu
- Xinjiang Medical University, Urumqi, China
| | - Qingbo Fang
- Department of Vascular Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xiaohu Ge
- Department of Vascular Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China.
| |
Collapse
|
30
|
Bibby G, Krasniqi B, Reddy I, Sekar D, Ross K. Capturing the RNA castle: Exploiting MicroRNA inhibition for wound healing. FEBS J 2021; 289:5137-5151. [PMID: 34403569 DOI: 10.1111/febs.16160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/14/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023]
Abstract
The growing pipelines of RNA-based therapies herald new opportunities to deliver better patient outcomes for complex disorders such as chronic nonhealing wounds associated with diabetes. Members of the microRNA (miRNA) family of small noncoding RNAs have emerged as targets for diverse elements of cutaneous wound repair, and both miRNA enhancement with mimics or inhibition with antisense oligonucleotides represent tractable approaches for miRNA-directed wound healing. In this review, we focus on miRNA inhibition strategies to stimulate skin repair given advances in chemical modifications to enhance the performance of antisense miRNA (anti-miRs). We first explore miRNAs whose inhibition in keratinocytes promotes keratinocyte migration, an essential part of re-epithelialisation during wound repair. We then focus on miRNAs that can be targeted for inhibition in endothelial cells to promote neovascularisation for wound healing in the context of diabetic mouse models. The picture that emerges is that direct comparisons of different anti-miRNAs modifications are required to establish the most translationally viable options in the chronic wound environment, that direct comparisons of the impact of inhibition of different miRNAs are needed to quantify and rank their relative efficacies in promoting wound repair, and that a standardised human ex vivo model of the diabetic wound is needed to reduce reliance on mouse models that do not necessarily enhance mechanistic understanding of miRNA-targeted wound healing.
Collapse
Affiliation(s)
- George Bibby
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, UK
| | - Blerta Krasniqi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, UK
| | - Izaak Reddy
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, UK
| | - Durairaj Sekar
- Dental Research Cell and Biomedical Research Unit (DRC-BRULAC), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science (SIMATS), Saveetha University, Chennai, India
| | - Kehinde Ross
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, UK
| |
Collapse
|
31
|
Huang K, Narumi T, Zhang Y, Li Q, Murugesan P, Wu Y, Liu NM, Cai H. Targeting MicroRNA-192-5p, a Downstream Effector of NOXs (NADPH Oxidases), Reverses Endothelial DHFR (Dihydrofolate Reductase) Deficiency to Attenuate Abdominal Aortic Aneurysm Formation. Hypertension 2021; 78:282-293. [PMID: 34176283 DOI: 10.1161/hypertensionaha.120.15070] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kai Huang
- Division of Molecular Medicine, Department of Anesthesiology (K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles.,Division of Cardiology, Department of Medicine ((K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles
| | - Taro Narumi
- Division of Molecular Medicine, Department of Anesthesiology (K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles.,Division of Cardiology, Department of Medicine ((K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology (K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles.,Division of Cardiology, Department of Medicine ((K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles
| | - Qiang Li
- Division of Molecular Medicine, Department of Anesthesiology (K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles.,Division of Cardiology, Department of Medicine ((K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles
| | - Priya Murugesan
- Division of Molecular Medicine, Department of Anesthesiology (K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles.,Division of Cardiology, Department of Medicine ((K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles
| | - Yusi Wu
- Division of Molecular Medicine, Department of Anesthesiology (K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles.,Division of Cardiology, Department of Medicine ((K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles
| | - Norika Mengchia Liu
- Division of Molecular Medicine, Department of Anesthesiology (K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles.,Division of Cardiology, Department of Medicine ((K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology (K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles.,Division of Cardiology, Department of Medicine ((K.H., T.N., Y.Z., Q.L., P.M., Y.W., N.M.L., H.C.), David Geffen School of Medicine, University of California Los Angeles
| |
Collapse
|
32
|
Shen Y, Lu H, Song G. MiR-221-3p and miR-92a-3p enhances smoking-induced inflammation in COPD. J Clin Lab Anal 2021; 35:e23857. [PMID: 34097306 PMCID: PMC8274981 DOI: 10.1002/jcla.23857] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/29/2021] [Accepted: 04/10/2021] [Indexed: 12/23/2022] Open
Abstract
Background Smoking is likely to facilitate airway inflammation and finally contributes to chronic obstructive pulmonary disease (COPD). This investigation was intended to elucidate miRNAs that were involved in smoking‐induced COPD. Methods Altogether 155 COPD patients and 77 healthy volunteers were recruited, and their serum levels of miR‐221‐3p and miR‐92a‐3p were determined. Besides, human bronchial epithelial cells (16HBECs) were purchased, and they were treated by varying concentrations of cigarette smoke extract (CSE). The 16HBECs were, additionally, transfected by miR‐221‐3p mimic, miR‐92a‐3p mimic, miR‐221‐3p inhibitor or miR‐92a‐3p inhibitor, and cytokines released by them, including TNF‐α, IL‐8, IL‐1β, and TGF‐β1, were monitored using enzyme linked immunosorbent assay (ELISA) kits. Results Chronic obstructive pulmonary disease patients possessed higher serum levels of miR‐221‐3p and miR‐92a‐3p than healthy volunteers (p < 0.05), and both miR‐221‐3p and miR‐92a‐3p were effective biomarkers in diagnosing stable COPD from acute exacerbation COPD. Moreover, viability of 16HBECs was undermined by CSE treatment (p < 0.05), and exposure to CSE facilitated 16HBECs’ release of TNF‐α, IL‐8, IL‐1β, and TGF‐β1 (p < 0.05). Furthermore, miR‐221‐3p/miR‐92a‐3p expression in 16HBECs was significantly suppressed after transfection of miR‐221‐3p/miR‐92a‐3p inhibitor (p < 0.05), which abated CSE‐triggered increase in cytokine production and decline in viability of 16HBECs (p < 0.05). Conclusion MiR‐221‐3p and miR‐92a‐3p were involved in CSE‐induced hyperinflammation of COPD, suggesting that they were favorable alternatives in diagnosing COPD patients with smoking history.
Collapse
Affiliation(s)
- Yahui Shen
- Department of Respiratory and Critical Care Medicine, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, China
| | - Huiyu Lu
- Department of Respiratory and Critical Care Medicine, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, China
| | - Guixian Song
- Department of Cardiology, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, China
| |
Collapse
|
33
|
Bao H, Li ZT, Xu LH, Su TY, Han Y, Bao M, Liu Z, Fan YJ, Lou Y, Chen Y, Jiang ZL, Gong XB, Qi YX. Platelet-Derived Extracellular Vesicles Increase Col8a1 Secretion and Vascular Stiffness in Intimal Injury. Front Cell Dev Biol 2021; 9:641763. [PMID: 33738288 PMCID: PMC7960786 DOI: 10.3389/fcell.2021.641763] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/09/2021] [Indexed: 12/31/2022] Open
Abstract
The arterial mechanical microenvironment, including stiffness, is a crucial pathophysiological feature of vascular remodeling, such as neointimal hyperplasia after carotid endarterectomy and balloon dilatation surgeries. In this study, we examined changes in neointimal stiffness in a Sprague-Dawley rat carotid artery intimal injury model and revealed that extracellular matrix (ECM) secretion and vascular stiffness were increased. Once the endothelial layer is damaged in vivo, activated platelets adhere to the intima and may secrete platelet-derived extracellular vesicles (pEVs) and communicate with vascular smooth muscle cells (VSMCs). In vitro, pEVs stimulated VSMCs to promote collagen secretion and cell adhesion. MRNA sequencing analysis of a carotid artery intimal injury model showed that ECM factors, including col8a1, col8a2, col12a1, and elastin, were upregulated. Subsequently, ingenuity pathway analysis (IPA) was used to examine the possible signaling pathways involved in the formation of ECM, of which the Akt pathway played a central role. In vitro, pEVs activated Akt signaling through the PIP3 pathway and induced the production of Col8a1. MicroRNA (miR) sequencing of pEVs released from activated platelets revealed that 14 of the top 30 miRs in pEVs targeted PTEN, which could promote the activation of the Akt pathway. Further research showed that the most abundant miR targeting PTEN was miR-92a-3p, which promoted Col8a1 expression. Interestingly, knockdown of Col8a1 expression in vivo abrogated the increase in carotid artery stiffness and simultaneously increased the degree of neointimal hyperplasia. Our results revealed that pEVs may deliver miR-92a-3p to VSMCs to induce the production and secretion of Col8a1 via the PTEN/PIP3/Akt pathway, subsequently increasing vascular stiffness. Therefore, pEVs and key molecules may be potential therapeutic targets for treating neointimal hyperplasia.
Collapse
Affiliation(s)
- Han Bao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zi-Tong Li
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Lei-Han Xu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Tong-Yue Su
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Yue Han
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Min Bao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ze Liu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yang-Jing Fan
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Lou
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Chen
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zong-Lai Jiang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Bo Gong
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| |
Collapse
|
34
|
Xu Y, Miao C, Cui J, Bian X. miR-92a-3p promotes ox-LDL induced-apoptosis in HUVECs via targeting SIRT6 and activating MAPK signaling pathway. ACTA ACUST UNITED AC 2021; 54:e9386. [PMID: 33470395 PMCID: PMC7812905 DOI: 10.1590/1414-431x20209386] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 10/12/2020] [Indexed: 12/19/2022]
Abstract
Atherosclerosis could be induced by multiple factors, including hypertension, hyperlipidemia, and smoking, and its pathogenesis has not been fully elucidated. MicroRNAs have been shown to possess great anti-atherosclerotic potential, but the precise function of miR-92a-3p in atherosclerosis and its potential molecular mechanism have not been well clarified. Flow cytometry assay and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazol-3-ium bromide (MTT) assay were performed to evaluate effects of oxidized low-density lipoprotein (ox-LDL) on proliferation and apoptosis of human umbilical vein endothelial cells (HUVECs), respectively. Malondialdehyde and superoxide dismutase levels in cell lysate were assessed with biochemical kits. The expression levels of miR-92a-3p and Sirtuin6 (SIRT6) in HUVECs exposed to ox-LDL were estimated by real-time quantitative polymerase chain reaction (RT-qPCR). In addition, the protein levels of SIRT6, c-Jun N-terminal kinase (JNK), phosphorylation JNK (p-JNK), p38 mitogen activated protein kinase (p38 MAPK), and phosphorylation p38 MAPK (p-p38 MAPK) were measured by western blot assays. The relationship between miR-92a-3p and SIRT6 was confirmed by dual-luciferase reporter assay. Ox-LDL induced apoptosis and oxidative stress in HUVECs in concentration- and time-dependent manners. Conversely, miR-92a-3p silencing inhibited apoptosis and SIRT6 expression in HUVECs. The overexpression of miR-92a-3p enhanced apoptosis and phosphorylation levels of JNK and p38 MAPK as well as inhibited proliferation in ox-LDL-induced HUVECs. In addition, SIRT6 was a target of miR-92a-3p. miR-92a-3p negatively regulated SIRT6 expression in ox-LDL-induced HUVECs to activate MAPK signaling pathway in vitro. In summary, miR-92a-3p promoted HUVECs apoptosis and suppressed proliferation in ox-LDL-induced HUVECs by targeting SIRT6 expression and activating MAPK signaling pathway.
Collapse
Affiliation(s)
- Yingchun Xu
- Department of Cardiology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, China
| | - Chunbo Miao
- Department of Internal Medicine, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, China
| | - Jinzhen Cui
- Department of Internal Medicine, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, China
| | - Xiaoli Bian
- Department of Cardiology, Yangzhou Jiangdu People's Hospital, Jiangdu District, Yangzhou, Jiangsu, China
| |
Collapse
|
35
|
Barbalata T, Moraru OE, Stancu CS, Devaux Y, Simionescu M, Sima AV, Niculescu LS. Increased miR-142 Levels in Plasma and Atherosclerotic Plaques from Peripheral Artery Disease Patients with Post-Surgery Cardiovascular Events. Int J Mol Sci 2020; 21:ijms21249600. [PMID: 33339419 PMCID: PMC7766790 DOI: 10.3390/ijms21249600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
There is an intensive effort to identify biomarkers to predict cardiovascular disease evolution. We aimed to determine the potential of microRNAs to predict the appearance of cardiovascular events (CVEs) in patients with peripheral artery disease (PAD) following femoral artery bypass surgery. Forty-seven PAD patients were enrolled and divided into two groups, without CVEs (n = 35) and with CVEs (n = 12), during 1 year follow-up. Intra-surgery atherosclerotic plaques from femoral arteries were collected and the levels of miR-142, miR-223, miR-155, and miR-92a of the primary transcripts of these microRNAs (pri-miRNAs), and gene expression of Drosha and Dicer were determined. Results showed that, in the plaques, miR-142, miR-223, and miR-155 expression levels were significantly increased in PAD patients with CVEs compared to those without CVEs. Positive correlations between these miRNAs and their pri-miRNAs levels and the Dicer/Drosha expression were observed. In the plasma of PAD patients with CVEs compared to those without CVEs, miR-223 and miR-142 were significantly increased. The multiple linear regression analyses revealed significant associations among several plasma lipids, oxidative and inflammatory parameters, and plasma miRNAs levels. Receiver operator characteristic (ROC) analysis disclosed that plasma miR-142 levels could be an independent predictor for CVEs in PAD patients. Functional bioinformatics analyses supported the role of these miRNAs in the regulation of biological processes associated with atherosclerosis. Taken together, these data suggest that plasma levels of miR-142, miR-223, miR-155, and miR-92a can significantly predict CVEs among PAD patients with good accuracy, and that plasma levels of miR-142 can be an independent biomarker to predict post-surgery CVEs development in PAD patients.
Collapse
Affiliation(s)
- Teodora Barbalata
- Lipidomics Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (T.B.); (C.S.S.); (M.S.); (A.V.S.)
| | - Oriana E. Moraru
- Emergency Clinical Hospital “Prof. Dr. Agrippa Ionescu”, 149 I.C. Brătianu Street, 077015 Baloteşti, Ilfov County, Romania;
| | - Camelia S. Stancu
- Lipidomics Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (T.B.); (C.S.S.); (M.S.); (A.V.S.)
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg;
| | - Maya Simionescu
- Lipidomics Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (T.B.); (C.S.S.); (M.S.); (A.V.S.)
| | - Anca V. Sima
- Lipidomics Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (T.B.); (C.S.S.); (M.S.); (A.V.S.)
| | - Loredan S. Niculescu
- Lipidomics Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (T.B.); (C.S.S.); (M.S.); (A.V.S.)
- Correspondence:
| |
Collapse
|
36
|
Arakawa Y, Itoh S, Fukazawa Y, Ishiguchi H, Kohmoto J, Hironishi M, Ito H, Kihira T. Association between oxidative stress and microRNA expression pattern of ALS patients in the high-incidence area of the Kii Peninsula. Brain Res 2020; 1746:147035. [PMID: 32739158 DOI: 10.1016/j.brainres.2020.147035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/30/2020] [Accepted: 07/26/2020] [Indexed: 11/29/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive degenerative disorder of the upper and lower motor neuron systems. The high incidence of ALS in the southern part of the Kii Peninsula of Japan (K-ALS) was reported in the 1960s, but it has gradually decreased to the worldwide average. Although causes of the high incidence of ALS in this area are unknown, our previous studies suggested that environmental factors, including essential mineral deficiency and increased metal-induced oxidative stress, play a role in its development. Recently, it has been reported that microRNAs (miRNA) contribute to the degeneration of nervous system such as ALS. The aim of this study is to explore specific miRNAs in K-ALS and evaluate relationships between oxidative stress. We comprehensively analyzed serum miRNAs and examined urinary 8-hydroxy-2'-deoxyguanosine (8-OHdG), serum Cu/Zn superoxide dismutase (SOD) and serum Nɛ-hexanoyl lysin (HEL) as oxidative stress markers in the patients with K-ALS, sporadic ALS (S-ALS), residents in this area (K-residents) and controls from another area. The expression levels of miR-92a-3p and miR-486-5p in the patients with K-ALS were significantly higher than those in controls. The HEL levels were significantly higher in the patients with K-ALS than in those with S-ALS and controls. The expression levels of miR-92a-3p and miR-486-5p were not correlated with the levels of HEL. A set of high levels of miR-92a-3p, miR-486-5p and serum HEL may be a useful biomarker for K-ALS in the Kii Peninsula. The findings should be further studied by a large number of subjects.
Collapse
Affiliation(s)
- Yuya Arakawa
- Ehime Prefectural University of Health Sciences, Japan; Kansai University of Health Sciences, Japan.
| | | | | | - Hiroshi Ishiguchi
- Wakayama Medical University, Japan; Shingu Municipal Medical Center, Japan
| | | | | | | | | |
Collapse
|
37
|
Kesidou D, da Costa Martins PA, de Windt LJ, Brittan M, Beqqali A, Baker AH. Extracellular Vesicle miRNAs in the Promotion of Cardiac Neovascularisation. Front Physiol 2020; 11:579892. [PMID: 33101061 PMCID: PMC7546892 DOI: 10.3389/fphys.2020.579892] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide claiming almost 17. 9 million deaths annually. A primary cause is atherosclerosis within the coronary arteries, which restricts blood flow to the heart muscle resulting in myocardial infarction (MI) and cardiac cell death. Despite substantial progress in the management of coronary heart disease (CHD), there is still a significant number of patients developing chronic heart failure post-MI. Recent research has been focused on promoting neovascularisation post-MI with the ultimate goal being to reduce the extent of injury and improve function in the failing myocardium. Cardiac cell transplantation studies in pre-clinical models have shown improvement in cardiac function; nonetheless, poor retention of the cells has indicated a paracrine mechanism for the observed improvement. Cell communication in a paracrine manner is controlled by various mechanisms, including extracellular vesicles (EVs). EVs have emerged as novel regulators of intercellular communication, by transferring molecules able to influence molecular pathways in the recipient cell. Several studies have demonstrated the ability of EVs to stimulate angiogenesis by transferring microRNA (miRNA, miR) molecules to endothelial cells (ECs). In this review, we describe the process of neovascularisation and current developments in modulating neovascularisation in the heart using miRNAs and EV-bound miRNAs. Furthermore, we critically evaluate methods used in cell culture, EV isolation and administration.
Collapse
Affiliation(s)
- Despoina Kesidou
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Paula A. da Costa Martins
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, Netherlands
- Faculty of Health, Medicine and Life Sciences, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Leon J. de Windt
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, Netherlands
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Abdelaziz Beqqali
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew Howard Baker
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Hosen MR, Goody PR, Zietzer A, Nickenig G, Jansen F. MicroRNAs As Master Regulators of Atherosclerosis: From Pathogenesis to Novel Therapeutic Options. Antioxid Redox Signal 2020; 33:621-644. [PMID: 32408755 DOI: 10.1089/ars.2020.8107] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Cardiovascular disease (CVD) remains the major cause of morbidity and mortality worldwide. Accumulating evidence indicates that atherosclerosis and its sequelae, coronary artery disease, contribute to the majority of cardiovascular deaths. Atherosclerosis is a chronic inflammatory disease of the arteries in which atherosclerotic plaques form within the vessel wall. Epidemiological studies have identified various risk factors for atherosclerosis, such as diabetes, hyperlipidemia, smoking, genetic predisposition, and sedentary lifestyle. Recent Advances: Through the advancement of genetic manipulation techniques and their use in cardiovascular biology, it was shown that small RNAs, especially microRNAs (miRNAs), are dynamic regulators of disease pathogenesis. They are considered to be central during the regulation of gene expression through numerous mechanisms and provide a means to develop biomarkers and therapeutic tools for the diagnosis and therapy of atherosclerosis. Circulating miRNAs encapsulated within membrane-surrounded vesicles, which originate from diverse subcellular compartments, are now emerging as novel regulators of intercellular communication. The miRNAs, in both freely circulating and vesicle-bound forms, represent a valuable tool for diagnosing and monitoring CVD, recently termed as "liquid biopsy." Critical Issues: However, despite the recent advancements in miRNA-based diagnostics and therapeutics, understanding how miRNAs can regulate atherosclerosis is still crucial to achieving an effective intervention and reducing the disease burden. Future Directions: We provide a landscape of the current developmental progression of RNA therapeutics as a holistic approach for treating CVD in different animal models and clinical trials. Future interrogations are warranted for the development of miRNA-based therapeutics to overcome challenges for the treatment of the disease.
Collapse
Affiliation(s)
- Mohammed Rabiul Hosen
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - Philip Roger Goody
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - Andreas Zietzer
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - Felix Jansen
- Department of Internal Medicine II, Molecular Cardiology, Heart Center Bonn, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| |
Collapse
|
39
|
Yu G, Liu P, Shi Y, Li S, Liu Y, Fan Z, Zhu W. Stimulation of endothelial progenitor cells by microRNA-31a-5p to induce endothelialization in an aneurysm neck after coil embolization by modulating the Axin1-mediated β-catenin/vascular endothelial growth factor pathway. J Neurosurg 2020; 133:918-926. [PMID: 31398705 DOI: 10.3171/2019.5.jns182901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 05/01/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Emerging evidence shows that frequent recurrence of intracranial aneurysms (IAs) after endovascular coiling is attributable to the lack of endothelialization across the aneurysm neck. Recently, much attention has been given to the role of microRNAs (miRs) in vascular disease, although their contributory role to IA is poorly understood. METHODS Adult male Sprague-Dawley rats were subjected to microsurgery to create a coiled embolization aneurysm model, and were injected with miR-31a-5p agomir or a negative control agomir via the tail vein at a dose of 10 mg/kg per week for 4 weeks after IA induction. H & E staining, scanning electron microscopy, and flow cytometry were performed to evaluate the effects of miR-31a-5p agomir on endothelialization and the number of circulating endothelial progenitor cells (EPCs). The effects of miR-31a-5p on the viability and functioning of EPCs were also determined using Cell Counting Kit-8, wound-healing assay, and tube formation assays. RESULTS The authors tested the ability of miR-31a-5p to promote EPC-induced endothelialization in a model of coiled embolization aneurysm. miR-31a-5p agomir improved endothelialization and elevated the number of circulating EPCs in the peripheral blood compared to a negative control agomir-treated group. In addition, the number of vWF- and KDR-positive cells in the aneurysm neck was increased in the miR-31a-5p agomir-treated group. Furthermore, upregulation of miR-31a-5p promoted EPC proliferation, migration, and tube formation and enhanced the expression of the proangiogenic factor vascular endothelial growth factor in vitro. Mechanistically, miR-31a-5p directly targeted the 3' untranslated region (3'UTR) of Axin1 messenger RNA and repressed its expression. Besides, miR-31a-5p exerted its effect on EPCs by regulating the Axin1-mediated Wnt/β-catenin pathway. CONCLUSIONS Collectively, these results indicate that miR-31a-5p is an important regulator of EPC mobilization and endothelialization and may have a positive effect on aneurysm repair.
Collapse
|
40
|
Li S, Yuan L, Su L, Lian Z, Liu C, Zhang F, Cui Y, Wu M, Chen H. Decreased miR‑92a‑3p expression potentially mediates the pro‑angiogenic effects of oxidative stress‑activated endothelial cell‑derived exosomes by targeting tissue factor. Int J Mol Med 2020; 46:1886-1898. [PMID: 32901851 PMCID: PMC7521555 DOI: 10.3892/ijmm.2020.4713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is an essential pathological feature of vulnerable atherosclerotic plaque. Exosome‑derived microRNAs (miRNAs or miRs) have been proven to be important regulators of angiogenesis. However, the role of exosomes, which are secreted by endothelial cells (ECs) under conditions of oxidative stress, in angiogenesis remain unclear. The present study aimed to investigate the effects and mechanism of oxidative stress‑activated endothelial‑derived exosomes in angiogenesis. Exosomes were isolated from H2O2‑stimulated human umbilical vein ECs (HUVECs; termed Exo-H2O2) by differential centrifugation and characterized by transmission electron microscopy, nanoparticle tracking analysis and western blot analysis. Exo-H2O2 enhanced HUVEC proliferation, migration and tube formation, as determined by EdU incorporation assay, scratch wound migration assay and tube formation assay, respectively. miR‑92a‑3p was identified as the predominantly downregulated miRNA in the Exo-H2O2‑treated HUVECs by small RNA sequencing, and the expression of primary miR‑92a (pri‑miR‑92a‑1) was also decreased, as shown by RT‑qPCR. Similarly, the inhibition of miR‑92a‑3p promoted angiogenesis in vitro and in vivo. miR‑92a‑3p overexpression blocked the pro‑angiogenic effects of Exo-H2O2 on target ECs. Tissue factor (TF), a molecule involved in angiogenesis, was increased in HUVECs in which miR‑92a‑3p expression was downregulated, as shown by mRNA sequencing. TF was also predicted as a target of miR‑92a‑3p by using the RNAhybrid program. The overexpression or suppression of miR‑92a‑3p modified TF expression at both the mRNA and protein level, as measured by RT‑qPCR and western blot analysis, respectively. Luciferase reporter assays suggested that miR‑92a‑3p inhibited TF expression by binding to the 3' untranslated region of TF. On the whole, the findings of the present study demonstrate that exosomes released from oxidative stress‑activated ECs stimulate angiogenesis by inhibiting miR‑92a‑3p expression in recipient ECs, and TF may be involved in the regulatory effects of miR‑92a‑3p on angiogenesis.
Collapse
Affiliation(s)
- Sufang Li
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Lan Yuan
- Medical and Healthy Analytical Center, Peking University, Beijing 100191, P.R. China
| | - Lina Su
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Zheng Lian
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Chuanfen Liu
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Feng Zhang
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Yuxia Cui
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Manyan Wu
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| | - Hong Chen
- Department of Cardiology, Peking University People's Hospital, Beijing 100191, P.R. China
| |
Collapse
|
41
|
Abplanalp WT, Fischer A, John D, Zeiher AM, Gosgnach W, Darville H, Montgomery R, Pestano L, Allée G, Paty I, Fougerousse F, Dimmeler S. Efficiency and Target Derepression of Anti-miR-92a: Results of a First in Human Study. Nucleic Acid Ther 2020; 30:335-345. [PMID: 32707001 DOI: 10.1089/nat.2020.0871] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
MicroRNA (miRNA) inhibition is a promising therapeutic strategy in several disease indications. MRG-110 is a locked nucleic acid-based antisense oligonucleotide that targets miR-92a-3p and experimentally was shown to have documented therapeutic effects on cardiovascular disease and wound healing. To gain first insights into the activity of anti-miR-92a in humans, we investigated miR-92a-3p expression in several blood compartments and assessed the effect of MRG-110 on target derepression. Healthy adults were randomly assigned (5:2) to receive a single intravenous dose of MRG-110 or placebo in one of seven sequential ascending intravenous dose cohorts ranging from 0.01 to 1.5 mg/kg body weight. MiR-92a-3p whole blood levels were time and dose dependently decreased with half-maximal inhibition of 0.27 and 0.31 mg/kg at 24 and 72 h after dosing, respectively. In the high-dose groups, >95% inhibition was detected at 24-72 h postinfusion and significant inhibition was observed for 2 weeks. Similar inhibitory effects were detected in isolated CD31+ cells, and miR-92a-3p expression was also inhibited in extracellular vesicles in the high-dose group. Target derepression was measured in whole blood and showed that ITGA5 and CD93 were increased at a dose of 1.5 mg/kg. Single-cell RNA sequencing of peripheral blood cells revealed a cell type-specific derepression of miR-92a targets. Together this study demonstrates that systemic infusion of anti-miR-92a efficiently inhibits miR-92a in the peripheral blood compartment and derepresses miR-92a targets in humans.
Collapse
Affiliation(s)
- Wesley Tyler Abplanalp
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt, Germany
| | - Ariane Fischer
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - David John
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Andreas M Zeiher
- German Center for Cardiovascular Research (DZHK), Frankfurt, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| | - Willy Gosgnach
- CentEX Biotechnology, Institut de Recherche SERVIER, Paris, France
| | - Helene Darville
- CentEX Biotechnology, Institut de Recherche SERVIER, Paris, France
| | | | | | - Guillaume Allée
- Center for Therapeutic Innovation Cardiovascular and Metabolic Disease, Institut de Recherches Internationales SERVIER, Paris, France
| | - Isabelle Paty
- Center for Therapeutic Innovation Cardiovascular and Metabolic Disease, Institut de Recherches Internationales SERVIER, Paris, France
| | - Francoise Fougerousse
- Center for Therapeutic Innovation Cardiovascular and Metabolic Disease, Institut de Recherches Internationales SERVIER, Paris, France
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| |
Collapse
|
42
|
Neointimal hyperplasia in the inferior vena cava of adenine-induced chronic kidney disease rats with aortocaval fistulas. Clin Exp Nephrol 2020; 24:1007-1014. [PMID: 32666345 DOI: 10.1007/s10157-020-01927-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 07/04/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND The failure of autologous arteriovenous fistulas (AVFs) occurs primarily due to stenosis in the anastomotic site, which is mainly related to the development of neointimal hyperplasia (NIH). Therefore, we conducted a study to establish a novel approach to create aortocaval fistulas (ACFs) in adenine-induced (AD) chronic kidney disease (CKD) rats to study the NIH in the inferior vena cava. METHODS Ten adult female rats received a 0.75% adenine-rich diet for 4 weeks to induce CKD and underwent ACF surgery. Ten healthy rats served as controls. A 5-10-mm segment of a vein immediately adjacent to that the portion of the vein used for creating the fistula was surgically removed at the time of creating the fistula, and reconstruction of the failed fistula from the same patient was used as controls. ACF was assessed using duplex scans and histopathological analyses. RESULTS At the end of the experiment, AD rats showed higher serum creatinine and urea nitrogen than those of vehicle-treated rats. Remarkable histological changes in kidney tissues demonstrated successful CKD models. Sections of the ACF in AD rats and veins removed at the time of the reconstruction of the failed fistula of the patient demonstrated that the eccentric neointima formation is irregularly thickened, with several small vessels within a more cellular region of the neointima. Immunohistochemistry demonstrated the presence of myofibroblasts, contractile smooth muscle cells and macrophages within the neointima. CONCLUSIONS Our rat models with ACFs showed typical features of NIH in the formation of fistula stenosis, which can resemble clinical findings in uremic patients.
Collapse
|
43
|
Sonnenschein K, Fiedler J, Pfanne A, Just A, Mitzka S, Geffers R, Pich A, Bauersachs J, Thum T. Therapeutic modulation of RNA-binding protein Rbm38 facilitates re-endothelialization after arterial injury. Cardiovasc Res 2020; 115:1804-1810. [PMID: 30843048 PMCID: PMC6755352 DOI: 10.1093/cvr/cvz063] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/13/2018] [Accepted: 03/01/2019] [Indexed: 12/12/2022] Open
Abstract
Aims Delayed re-endothelialization after balloon angioplasty in patients with coronary or peripheral artery disease impairs vascular healing and leads to neointimal proliferation. In the present study, we examined the effect of RNA-binding motif protein 38 (Rbm38) during re-endothelialization in a murine model of experimental vascular injury. Methods and results Left common carotid arteries of C57BL/6 mice were electrically denudated and endothelial regeneration was evaluated. Profiling of RNA-binding proteins revealed dysregulated expression of Rbm38 in the denudated and regenerated areas. We next tested the importance of Rbm38 in human umbilical vein endothelial cells (HUVECS) and analysed its effects on cellular proliferation, migration and apoptosis. Rbm38 silencing in vitro demonstrated important beneficial functional effects on migratory capacity and proliferation of endothelial cells. In vivo, local silencing of Rbm38 also improved re-endothelialization of denuded carotid arteries. Luciferase reporter assay identified miR-98 and let-7f to regulate Rbm38 and the positive proliferative properties of Rbm38 silencing in vitro and in vivo were mimicked by therapeutic overexpression of these miRNAs. Conclusion The present data identified Rbm38 as an important factor of the regulation of various endothelial cell functions. Local inhibition of Rbm38 as well as overexpression of the upstream regulators miR-98 and let-7f improved endothelial regeneration in vivo and thus may be a novel therapeutic entry point to avoid endothelial damage after balloon angioplasty.
Collapse
Affiliation(s)
- Kristina Sonnenschein
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany.,Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Angelika Pfanne
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Annette Just
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Saskia Mitzka
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany
| | - Robert Geffers
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Pich
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,Excellence Cluster REBIRTH, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany.,Excellence Cluster REBIRTH, Hannover Medical School, Hannover, Germany.,National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
44
|
Nader J, Metzinger L, Maitrias P, Caus T, Metzinger-Le Meuth V. Aortic valve calcification in the era of non-coding RNAs: The revolution to come in aortic stenosis management? Noncoding RNA Res 2020; 5:41-47. [PMID: 32195449 PMCID: PMC7075756 DOI: 10.1016/j.ncrna.2020.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/04/2020] [Accepted: 02/24/2020] [Indexed: 01/08/2023] Open
Abstract
Aortic valve stenosis remains the most frequent structural heart disease, especially in the elderly. During the last decade, we noticed an important consideration and a huge number of publications related to the medical and surgical treatment of this disease. However, the molecular aspect of this degenerative issue has also been more widely studied recently. As evidenced in oncologic but also cardiac research fields, the emergence of microRNAs in the molecular screening and follow-up makes them potential biomarkers in the future, for the diagnosis, follow-up and treatment of aortic stenosis. Herein, we present a review on the implication of microRNAs in the aortic valve disease management. After listing and describing the main miRNAs of interest in the field, we provide an outline to develop miRNAs as innovative biomarkers and innovative therapeutic strategies, and describe a groundbreaking pre-clinical study using inhibitors of miR-34a in a pre-clinical model of aortic valve stenosis.
Collapse
Affiliation(s)
- Joseph Nader
- Department of Cardiac Surgery, Amiens University Hospital, Amiens, France
| | - Laurent Metzinger
- HEMATIM EA4666, C.U.R.S, Université de Picardie Jules Verne, 80025, AMIENS Cedex 1, France
| | - Pierre Maitrias
- Department of Vascular Surgery, Polyclinique Saint Côme, Compiègne, France
| | - Thierry Caus
- Department of Cardiac Surgery, Amiens University Hospital, Amiens, France
| | - Valérie Metzinger-Le Meuth
- HEMATIM EA4666, C.U.R.S, Université de Picardie Jules Verne, 80025, AMIENS Cedex 1, France.,INSERM U1148, Laboratory for Vascular Translational Science (LVTS), UFR SMBH, Université Paris 13-Sorbonne Paris Cité, 93017, BOBIGNY CEDEX, France
| |
Collapse
|
45
|
Circulating exosomes from patients with peripheral artery disease influence vascular cell migration and contain distinct microRNA cargo. JVS Vasc Sci 2020; 1:28-41. [PMID: 32550603 PMCID: PMC7299234 DOI: 10.1016/j.jvssci.2020.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Objective Peripheral artery disease (PAD) is a chronic condition characterized by inflammation. Emerging literature suggests that circulating exosomes and their microRNA (miRNA) contents may influence atherosclerosis and vascular remodeling. We hypothesize that circulating exosomes in patients with PAD directly modulate vascular cell phenotype and contain proinflammatory miRNAs. Methods Exosomes (particle size, 30-150 nm) were isolated from plasma of healthy individuals (n = 6), patients with mild PAD (mPAD; median Rutherford class, 2.5; n = 6), and patients with severe PAD (sPAD; median Rutherford class, 4; n = 5). Exosome identity, size, and concentration were determined by Western blot and nanoparticle tracking analysis. Human vascular smooth muscle cell (VSMC) and endothelial cell (EC) migration was assessed by a standard wound closure assay after exposure to exosome preparations. Monocyte-derived macrophages isolated from healthy volunteers were exposed to exosome preparations, and targeted gene expression was analyzed using quantitative polymerase chain reaction. Exosome miRNA cargos were isolated, and a panel of defined, vascular-active miRNAs was assessed by quantitative polymerase chain reaction. Results There was no difference in overall exosome particle concentration or size between the three groups (one-way analysis of variance [ANOVA], P > .05). Compared with exosomes from healthy individuals, exosomes from mPAD and sPAD patients increased VSMC migration (1.0 ± 0.09-fold vs 1.5 ± 0.09-fold vs 2.0 ± 0.12-fold wound closure; ANOVA, P < .0001) and inhibited EC migration (1.8 ± 0.07-fold vs 1.5 ± 0.04-fold vs 1.3 ± 0.02-fold wound closure; ANOVA, P < .01) in a stepwise fashion. Exosomes also induced changes in monocyte-derived macrophage gene expression that did not appear PAD specific. Hierarchical analysis of exosome miRNA revealed distinct clustering of vascular-active miRNAs between the three groups. Several miRNAs that promote inflammatory pathways in vascular cells were expressed at higher levels in exosomes from sPAD patients. Conclusions Circulating exosomes from individuals with PAD exert in vitro functional effects on VSMCs and ECs that may promote adverse vessel remodeling. Exosomes from healthy individuals, mPAD patients, and sPAD patients contain distinct signatures of immune-regulatory miRNA. Together these data suggest that the proinflammatory cargo of circulating exosomes correlates with atherosclerosis severity in PAD patients and could influence vascular injury and repair. Exosomes and their cargo have been implicated in several vascular remodeling processes including atherosclerosis, angiogenesis, and neointimal hyperplasia. In this study, we demonstrate that circulating exosomes from individuals with peripheral artery disease exert in vitro effects on vascular cells that may adversely affect vessel remodeling. Moreover, these exosomes contain elevated levels of vascular-active microRNA. Our results suggest that exosomes may serve as both biomarkers and effectors of vascular disease in patients with peripheral artery disease and motivate further investigation into the role of exosomes and their contents in aberrant remodeling in vascular diseases.
Collapse
|
46
|
Xing X, Li Z, Yang X, Li M, Liu C, Pang Y, Zhang L, Li X, Liu G, Xiao Y. Adipose-derived mesenchymal stem cells-derived exosome-mediated microRNA-342-5p protects endothelial cells against atherosclerosis. Aging (Albany NY) 2020; 12:3880-3898. [PMID: 32096479 PMCID: PMC7066923 DOI: 10.18632/aging.102857] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
Abstract
Exosomes are reported to mediate several disease-related microRNAs (miRNAs) to affect the progression of diseases, including atherosclerosis. Here, we aimed to screen the atherosclerosis-associated miRNAs and preliminarily investigate the potential regulatory mechanism of atherosclerosis. First, the lesion model for human umbilical vein endothelial cells (HUVECs) was favorably constructed. Later, through RNA-sequencing and bioinformatics analyses, miR-342-5p was identified in lesion model for HUVECs. MiR-342-5p overexpression or knockdown evidently promoted or inhibited the apoptosis of HUVECs impaired by H2O2. Mechanistically, PPP1R12B was found to have great potential as a target of miR-342-5p in HUVECs impaired by H2O2, supported by RNA-sequencing and a series of bioinformatics analyses. Meanwhile, the effect of miR-342-5p on PPP1R12B expression in HUVECs’ lesion model was explored, revealing that miR-342-5p had an inhibitory role in PPP1R12B expression. Additionally, adipose-derived mesenchymal stem cells (ADSCs) in spindle-like shape and their derived exosomes with 30 to 150 nm diameter were characterized. Furthermore, results showed miR-342-5p was evidently decreased in the presence of ADSCs-derived exosomes. These findings indicated ADSCs-derived exosomes restrained the expression of miR-324-5p in lesion model. Collectively, this work demonstrates an atherosclerosis-associated miR-342-5p and reveals a preliminary possible mechanism in which miR-342-5p mediated by ADSCs-derived exosomes protects endothelial cells against atherosclerosis.
Collapse
Affiliation(s)
- Xiaohui Xing
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250000, Shandong Province, P.R. China.,Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Zhongchen Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China.,Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, Shandong Province, P.R. China
| | - Xin Yang
- Department of Otolaryngology, General Hospital of Central Theater Command of PLA, Wuhan 430070, Hubei, China
| | - Mengyou Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Chao Liu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Yuejiu Pang
- Department of Senile Neurology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250000, Shandong Province, P.R. China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Xueyuan Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Guangcun Liu
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250000, Shandong Province, P.R. China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng 250000, Shandong Province, P.R. China
| |
Collapse
|
47
|
Reconstruction of a lncRNA-Associated ceRNA Network in Endothelial Cells under Circumferential Stress. Cardiol Res Pract 2020; 2020:1481937. [PMID: 32148949 PMCID: PMC7042510 DOI: 10.1155/2020/1481937] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background Numerous studies have highlighted that long noncoding RNA (lncRNA) can indirectly regulate the expression of mRNAs by binding to microRNA (miRNA). LncRNA-associated ceRNA networks play a vital role in the initiation and progression of several pathological mechanisms. However, the lncRNA-miRNA-mRNA ceRNA network in endothelial cells under cyclic stretch is seldom studied. Methods The miRNA, mRNA, and lncRNA expression profiles of 6 human umbilical vein endothelial cells (HUVECs) under circumferential stress were obtained by next-generation sequencing (NGS). We identified the differential expression of miRNAs, mRNAs, and lncRNAs using the R software package GDCRNATools. Cytoscape was adopted to construct a lncRNA-miRNA-mRNA ceRNA network. In addition, through GO and KEGG pathway annotations, we analyzed gene functions and their related pathways. We also adopted ELISA and TUNEL to investigate the effect of si-NEAT1 on endothelial inflammation and apoptosis. Results We recognized a total of 32978 lncRNAs, 1046 miRNAs, and 31958 mRNAs in 6 samples; among them, 155 different expressed lncRNAs, 74 different expressed miRNAs, and 960 different mRNAs were adopted. Based on the established theory, the ceRNA network was composed of 13 lncRNAs, 44 miRNAs, and 115 mRNAs. We constructed and visualized a lncRNA-miRNA-mRNA network, and the top 20 nodes are identified after calculating their degrees. The nodes with most degrees in three kinds of RNAs are hsa-miR-4739, NEAT1, and MAP3K2. Functional analysis showed that different biological processes enriched in biological regulation, response to stimulus and cell communication. Pathway analysis was mainly enriched in longevity regulating, cell cycle, mTOR, and FoxO signaling pathway. Circumferential stress can significantly downregulate NEAT1, and after transducing si-NEAT1 for 24 h, inflammatory cytokine IL-6 and MCP-1 were significantly increased; furthermore, fewer TUNEL-positive cells were found in the si-NEAT1 treated group. Conclusions The establishing of a ceRNA network can help further understand the mechanism of vein graft failure. Our data demonstrated that NEAT1 may be a core factor among the mechanical stress factors and that cyclic stress can significantly reduce expression of NEAT1, give rise to inflammation in the early stage of endothelial dysfunction, and promote EC apoptosis, which may play an essential role in vein graft failure.
Collapse
|
48
|
Dai R, Liu Y, Zhou Y, Xiong X, Zhou W, Li W, Zhou W, Chen M. Potential of circulating pro-angiogenic microRNA expressions as biomarkers for rapid angiographic stenotic progression and restenosis risks in coronary artery disease patients underwent percutaneous coronary intervention. J Clin Lab Anal 2020; 34:e23013. [PMID: 31495986 PMCID: PMC6977144 DOI: 10.1002/jcla.23013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This study aimed to investigate the correlation of pro-angiogenic microRNA (miRNA) expressions with rapid angiographic stenotic progression (RASP) and restenosis risks in coronary artery disease (CAD) patients underwent percutaneous coronary intervention (PCI) with drug-eluting stents (DES). METHODS A total of 286 CAD patients underwent PCI with DES were consecutively recruited in this study. Plasma samples were collected before PCI operation, and 14 pro-angiogenic miRNAs were measured by real-time quantitative reverse transcription-polymerase chain reaction. Rapid angiographic stenotic progression at nontarget lesions and restenosis at stented lesions were evaluated by quantitative coronary angiography at 12 months after PCI operation. RESULTS The occurrence rates of RASP and restenosis were 39.5% and 22.4%, respectively. Let-7f, miR-19a, miR-19b-1, miR-92a, miR-126, miR-210, and miR-296 were decreased in RASP patients than non-RASP patients, among which let-7f, miR-19a, miR-126, miR-210, and miR-296 independently correlated with lower RASP occurrence by multivariate analysis, followed by receiver-operating characteristic (ROC) curve exhibited that these five miRNAs showed great value in predicting RASP risk with area under curve (AUC) 0.879 (95% CI: 0.841-0.917). Besides, let-7f, miR-19a, miR-92a, miR-126, miR-130a, and miR-210 were reduced in restenosis patients than non-restenosis patients, among them miR-19a, miR-126, miR-210, and miR-378 independently correlated with lower restenosis occurrence by multivariate analysis, followed by ROC curve disclosed that these four miRNAs had good value in predicting restenosis risk with AUC 0.776 (95% CI: 0.722-0.831). CONCLUSIONS Circulating let-7f, miR-19a, miR-126, miR-210, and miR-296 independently correlate with reduced RASP risk, while miR-19a, miR-126, miR-210, and miR-378 independently correlate with decreased restenosis risk in CAD patients underwent PCI with DES.
Collapse
Affiliation(s)
- Rui Dai
- Department of Cardiology, Tongji Medical CollegeThe Central Hospital of Wuhan, Huazhong University of Science & TechnologyWuhanChina
| | - Yijue Liu
- Emergency Department, Tongji Medical CollegeThe Central Hospital of Wuhan, Huazhong University of Science & TechnologyWuhanChina
| | - Yi Zhou
- Department of Cardiology, Tongji Medical CollegeThe Central Hospital of Wuhan, Huazhong University of Science & TechnologyWuhanChina
| | - Xiaoju Xiong
- Department of Cardiology, Tongji Medical CollegeThe Central Hospital of Wuhan, Huazhong University of Science & TechnologyWuhanChina
| | - Wei Zhou
- Department of Cardiology, Tongji Medical CollegeThe Central Hospital of Wuhan, Huazhong University of Science & TechnologyWuhanChina
| | - Weijuan Li
- Department of Cardiology, Tongji Medical CollegeThe Central Hospital of Wuhan, Huazhong University of Science & TechnologyWuhanChina
| | - Wenping Zhou
- Department of Cardiology, Tongji Medical CollegeThe Central Hospital of Wuhan, Huazhong University of Science & TechnologyWuhanChina
| | - Manhua Chen
- Department of Cardiology, Tongji Medical CollegeThe Central Hospital of Wuhan, Huazhong University of Science & TechnologyWuhanChina
| |
Collapse
|
49
|
Solly EL, Dimasi CG, Bursill CA, Psaltis PJ, Tan JTM. MicroRNAs as Therapeutic Targets and Clinical Biomarkers in Atherosclerosis. J Clin Med 2019; 8:E2199. [PMID: 31847094 PMCID: PMC6947565 DOI: 10.3390/jcm8122199] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/11/2019] [Indexed: 12/21/2022] Open
Abstract
Atherosclerotic cardiovascular disease remains the leading cause of morbidity and mortality worldwide. Atherosclerosis develops over several decades and is mediated by a complex interplay of cellular mechanisms that drive a chronic inflammatory milieu and cell-to-cell interactions between endothelial cells, smooth muscle cells and macrophages that promote plaque development and progression. While there has been significant therapeutic advancement, there remains a gap where novel therapeutic approaches can complement current therapies to provide a holistic approach for treating atherosclerosis to orchestrate the regulation of complex signalling networks across multiple cell types and different stages of disease progression. MicroRNAs (miRNAs) are emerging as important post-transcriptional regulators of a suite of molecular signalling pathways and pathophysiological cellular effects. Furthermore, circulating miRNAs have emerged as a new class of disease biomarkers to better inform clinical diagnosis and provide new avenues for personalised therapies. This review focusses on recent insights into the potential role of miRNAs both as therapeutic targets in the regulation of the most influential processes that govern atherosclerosis and as clinical biomarkers that may be reflective of disease severity, highlighting the potential theranostic (therapeutic and diagnostic) properties of miRNAs in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Emma L. Solly
- Vascular Research Centre, Heart and Vascular Health Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide SA 5000, Australia; (E.L.S.); (C.G.D.); (C.A.B.); (P.J.P.)
- Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| | - Catherine G. Dimasi
- Vascular Research Centre, Heart and Vascular Health Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide SA 5000, Australia; (E.L.S.); (C.G.D.); (C.A.B.); (P.J.P.)
| | - Christina A. Bursill
- Vascular Research Centre, Heart and Vascular Health Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide SA 5000, Australia; (E.L.S.); (C.G.D.); (C.A.B.); (P.J.P.)
- Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| | - Peter J. Psaltis
- Vascular Research Centre, Heart and Vascular Health Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide SA 5000, Australia; (E.L.S.); (C.G.D.); (C.A.B.); (P.J.P.)
- Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| | - Joanne T. M. Tan
- Vascular Research Centre, Heart and Vascular Health Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide SA 5000, Australia; (E.L.S.); (C.G.D.); (C.A.B.); (P.J.P.)
- Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| |
Collapse
|
50
|
Liu Y, Li Q, Hosen MR, Zietzer A, Flender A, Levermann P, Schmitz T, Frühwald D, Goody P, Nickenig G, Werner N, Jansen F. Atherosclerotic Conditions Promote the Packaging of Functional MicroRNA-92a-3p Into Endothelial Microvesicles. Circ Res 2019; 124:575-587. [PMID: 30582459 DOI: 10.1161/circresaha.118.314010] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Microvesicle-incorporated microRNAs (miRs) are biomarkers and effectors of cardiovascular disease. Whether microvesicle-miR expression is regulated in coronary artery disease (CAD) or not is unknown. OBJECTIVE Here, we explore the expression of circulating microvesicle-miRs in patients with CAD and investigate the role of microvesicle-miR in endothelial cells. METHODS AND RESULTS Circulating microvesicles were isolated from patients' plasma by using ultracentrifugation. Electron microscopy was used to determine the size of the microvesicles. A Taqman miR array revealed certain microvesicle-miRs are significantly regulated in patients with stable CAD compared with patients with ACS. To validate the miR array results, 180 patients with angiographically excluded CAD (n=41), stable CAD (n=77), and acute coronary syndrome (n=62) were prospectively studied. Nine miRs involved in regulation of vascular performance-miR-126-3p, miR-222-3p, miR-let-7d-5p, miR-21-5p, miR-26a-5p, miR-92a-3p, miR-139-5p, miR-30b-5p, and miR-199a-5p-were quantified in circulating microvesicles by real-time polymerase chain reaction (PCR). Among these, miR-92a-3p was significantly increased in patients with CAD compared with non-CAD patients. Microvesicle-sorting experiments showed endothelial cells (ECs) were the major cell source for microvesicles containing miR-92a-3p. In vitro oxLDL (oxidized low-density lipoprotein) and IL-6 (interleukin-6) stimulation increased miR-92a-3p expression in parent ECs and upregulated the expression level of endothelial microvesicle (EMV)-incorporated miR-92a-3p. Labeling of miR-92a-3p and EMVs demonstrated that functional miR-92a-3p was transported into recipient ECs, which accelerated cell migration and proliferation. Knockdown of miR-92a-3p in EMVs abrogated EMV-mediated effects on EC migration, proliferation, and blocked vascular network formation in a matrigel plug. Polymerase chain reaction-based gene profiling showed that the expression of THBS1 (thrombospondin 1) protein-a target of miR-92a-3p and an inhibitor of angiogenesis-was significantly reduced in ECs by EMVs. Knockdown of miR-92a-3p in EMVs abrogated EMV-mediated inhibition of the THBS1 gene and protein expression. CONCLUSIONS Atherosclerotic conditions promote the packaging of endothelial miR-92a-3p into EMVs. EMV-mediated transfer of functional miR-92a-3p regulates angiogenesis in recipient ECs by a THBS1-dependent mechanism.
Collapse
Affiliation(s)
- Yangyang Liu
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Qian Li
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.).,Department of Cardiology, Second Hospital of Jilin University, Changchun, China (Q.L.)
| | - Mohammed Rabiul Hosen
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Andreas Zietzer
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Anna Flender
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Paula Levermann
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Theresa Schmitz
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Daniel Frühwald
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Philip Goody
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Georg Nickenig
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Nikos Werner
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| | - Felix Jansen
- From the Department of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany (Y.L., Q.L., M.R.H., A.Z., A.F., P.L., T.S., D.F., P.G., G.N., N.W., F.J.)
| |
Collapse
|