1
|
Ye Z, Zhu S, Li G, Lu J, Huang S, Du J, Shao Y, Ji Z, Li P. Early matrix softening contributes to vascular smooth muscle cell phenotype switching and aortic dissection through down-regulation of microRNA-143/145. J Mol Cell Cardiol 2024; 192:1-12. [PMID: 38718921 DOI: 10.1016/j.yjmcc.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 05/04/2024] [Accepted: 05/04/2024] [Indexed: 05/14/2024]
Abstract
Thoracic aortic dissection (TAD) is characterized by extracellular matrix (ECM) dysregulation. Aberrations in the ECM stiffness can lead to changes in cellular functions. However, the mechanism by which ECM softening regulates vascular smooth muscle cell (VSMCs) phenotype switching remains unclear. To understand this mechanism, we cultured VSMCs in a soft extracellular matrix and discovered that the expression of microRNA (miR)-143/145, mediated by activation of the AKT signalling pathway, decreased significantly. Furthermore, overexpression of miR-143/145 reduced BAPN-induced aortic softening, switching the VSMC synthetic phenotype and the incidence of TAD in mice. Additionally, high-throughput sequencing of immunoprecipitated RNA indicated that the TEA domain transcription factor 1 (TEAD1) is a common target gene of miR-143/145, which was subsequently verified using a luciferase reporter assay. TEAD1 is upregulated in soft ECM hydrogels in vitro, whereas the switch to a synthetic phenotype in VSMCs decreases after TEAD1 knockdown. Finally, we verified that miR-143/145 levels are associated with disease severity and prognosis in patients with thoracic aortic dissection. ECM softening, as a result of promoting the VSMCs switch to a synthetic phenotype by downregulating miR-143/145, is an early trigger of TAD and provides a therapeutic target for this fatal disease. miR-143/145 plays a role in the early detection of aortic dissection and its severity and prognosis, which can offer information for future risk stratification of patients with dissection.
Collapse
Affiliation(s)
- Zhaofei Ye
- Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Shuolin Zhu
- Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Guoqi Li
- Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Jie Lu
- Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Shan Huang
- Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Jie Du
- Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Yihui Shao
- Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, China.
| | - Zhili Ji
- Beijing Chaoyang Hospital of Capital Medical University, China.
| | - Ping Li
- Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, China.
| |
Collapse
|
2
|
MicroRNA-34b-5p binds enhancer of zeste 2 to inhibit milk fat globule-EGF factor 8 expression, affecting liver fibrosis. J Physiol Biochem 2022; 78:885-895. [PMID: 36138295 DOI: 10.1007/s13105-022-00914-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/18/2022] [Indexed: 10/14/2022]
Abstract
Activated hepatic stellate cells (HSCs) are considered the major drivers in the process of hepatic fibrosis. This study intends to explore the mechanism underlying microRNA (miR)-34b-5p effects over liver fibrosis through the enhancer of zeste 2 (EZH2)/milk fat globule-EGF factor 8 (MFGE8) axis in HSCs. A liver fibrosis model was generated by carbon tetrachloride (CCl4) in C57BL/6 J mice and subjected to histological examinations and detection of HSC activation and miR-34b-5p/EZH2/MFGE8 expression. Primary HSCs were treated with transforming growth factor (TGF)-β and tested for proliferation, activation, and expression of fibrosis-related factors. A dual luciferase reporter assay was performed for confirming the targeted relationship between miR-34b-5p and EZH2. Chromatin immunoprecipitation was used to measure EZH2 enrichment in the MFGE8 promoter region. We found that miR-34b-5p was lowly expressed in the CCl4-induced mouse model. Overexpression of miR-34b-5p suppressed both TGF-β-induced HSC proliferation and the expression of fibrosis-related factors and HSC activation markers. A dual luciferase assay showed a binding relationship between miR-34b-5p and EZH2. Overexpression of miR-34b-5p reduced TGF-β-induced HSC activation by inhibiting EZH2 to promote MFGE8 expression. Overexpression of miR-34b-5p inhibited liver fibrosis in vivo through the EZH2/MFGE8 axis. Conclusively, overexpressing miR-34b-5p reduced TGF-β-induced HSC activation by inhibiting EZH2 and thereby promoting MFGE8 expression, and inhibited liver fibrosis in vivo through the EZH2/MFGE8 axis.
Collapse
|
3
|
Gao Y, Gao Y, Niu Z, Liu J, Feng H, Sun J, Wang L, Pan L. CCCTC-binding factor-mediated microRNA-340-5p suppression aggravates myocardial injury in rats with severe acute pancreatitis through activation of the HMGB1/TLR4 axis. Immunopharmacol Immunotoxicol 2022; 44:306-315. [PMID: 35238277 DOI: 10.1080/08923973.2022.2043898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 02/13/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Severe acute pancreatitis (SAP) is a life-threatening disorder associated with multisystem organ failure. This study aimed to investigate the function of high mobility group box 1 (HMGB1) in SAP-induced myocardial injury. METHODS A rat model with SAP was induced. The pathological changes in rat pancreatic and cardiac tissues were examined by HE staining. Cardiomyocyte apoptosis in rat cardiac tissues, and the serum levels of myocardial injury markers and pro-inflammatory cytokines were examined. Rat primary cardiomyocytes were treated with H2O2 for in vitro experiments. The regulatory molecules of HMGB1 were predicted by bioinformatics analysis. Altered expression of HMGB1, microRNA (miR)-340-5p and CCCTC-binding factor (CTCF) was introduced in rats or cells to investigate their roles in myocardial injury. RESULTS CTCF and HMGB1 were highly expressed but miR-340-5p was poorly expressed in cardiac tissues of rats with SAP. HMGB1 silencing reduced toll-like receptor 4 (TLR4) expression to promote proliferation and reduce apoptosis of H2O2-treated cardiomyocytes. miR-340-5p targeted HMGB1 mRNA, while CTCF suppressed miR-340-5p transcription. CTCF upregulation or miR-340-5p downregulation blocked the effects of HMGB1 silencing on cardiomyocytes. In vivo, CTCF silencing alleviated injury in rat pancreatic and cardiac tissues and reduced the expression of creatine kinase-MB (CK-MB), lactic dehydrogenase, interleukin (IL)-1β, IL-6 and tumor necrosis factor-α (TNF-α) in rat serum. But further overexpression of HMGB1 or inhibition of miR-340-5p aggravated the symptoms in rats. CONCLUSION This study demonstrated that CTCF reduces transcription of miR-340-5p to promote HMGB1 expression, which activates TLR4 expression and promotes myocardial injury in rats with SAP.
Collapse
Affiliation(s)
- Yazhou Gao
- Department of Emergency Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yanxia Gao
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Zequn Niu
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jie Liu
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Hui Feng
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jiangli Sun
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Liming Wang
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Longfei Pan
- Department of Emergency Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
4
|
Hasemaki N, Andreou NP, Legaki E, Katsargyris A, Gazouli M, Klonaris C. Association of miRNA-145 Single Nucleotide Polymorphisms in Abdominal Aortic Aneurysms. In Vivo 2022; 36:1120-1125. [PMID: 35478113 PMCID: PMC9087113 DOI: 10.21873/invivo.12810] [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: 01/17/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM MicroRNAs (miRNAs) are non-coding RNA molecules that exert post-transcriptional gene expression regulation in response to cellular or environmental changes. Genetic variation affects their synthesis and cellular actions, and single nucleotide polymorphisms (SNPs) are one example of genetic variants studied in relation to various diseases. Literature indicates that the differentially expressed miRNA-145 in patients' serum is an essential biomarker for abdominal aortic aneurysm (AAA). However, the correlation between specific miR-145 genetic polymorphisms with AAA susceptibility is inadequately studied. MATERIALS AND METHODS Eighty-seven AAA patients and 122 healthy controls were recruited. Peripheral blood samples were genotyped for miRNA-145 SNPs; rs55945735, rs73798217 and rs353291. RESULTS The GG genotype of the rs55945735 polymorphism (p=0.047) and the AG genotype of the rs353291 polymorphism (p=0.036) were overrepresented in AAA patients compared to healthy individuals, revealing an association with susceptibility to AAA development. CONCLUSION SNPs rs55945735 and rs353291 are associated with AAA susceptibility.
Collapse
Affiliation(s)
- Natasha Hasemaki
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Nikolaos-Panagiotis Andreou
- Department of Basic Medical Science, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Legaki
- Department of Basic Medical Science, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Katsargyris
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Department of Vascular and Endovascular Surgery, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Maria Gazouli
- Department of Basic Medical Science, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece;
| | - Christos Klonaris
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
5
|
Si K, Lu D, Tian J. Integrated analysis and the identification of a circRNA-miRNA-mRNA network in the progression of abdominal aortic aneurysm. PeerJ 2022; 9:e12682. [PMID: 35036156 PMCID: PMC8711282 DOI: 10.7717/peerj.12682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/02/2021] [Indexed: 11/20/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a disease commonly seen in the elderly. The aneurysm diameter increases yearly, and the larger the AAA the higher the risk of rupture, increasing the risk of death. However, there are no current effective interventions in the early stages of AAA. Methods Four gene expression profiling datasets, including 23 normal artery (NOR) tissue samples and 97 AAA tissue samples, were integrated in order to explore potential molecular biological targets for early intervention. After preprocessing, differentially expressed genes (DEGs) between AAA and NOR were identified using LIMMA package. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis were conducted using the DAVID database. The protein-protein interaction network was constructed and hub genes were identified using the STRING database and plugins in Cytoscape. A circular RNA (circRNA) profile of four NOR tissues versus four AAA tissues was then reanalyzed. A circRNA-miRNA-mRNA interaction network was constructed after predictions were made using the Targetscan and Circinteractome databases. Results A total of 440 DEGs (263 up-regulated and 177 down-regulated) were identified in the AAA group, compared with the NOR group. The majority were associated with the extracellular matrix, tumor necrosis factor-α, and transforming growth factor-β. Ten hub gene-encoded proteins (namely IL6, RPS27A, JUN, UBC, UBA52, FOS, IL1B, MMP9, SPP1 and CCL2) coupled with a higher degree of connectivity hub were identified after protein‐protein interaction network analysis. Our results, in combination with the results of previous studies revealed that miR-635, miR-527, miR-520h, miR-938 and miR-518a-5p may be affected by circ_0005073 and impact the expression of hub genes such as CCL2, SPP1 and UBA52. The miR-1206 may also be affected by circ_0090069 and impact RPS27A expression. Conclusions This circRNA-miRNA-mRNA network may perform critical roles in AAA and may be a novel target for early intervention.
Collapse
Affiliation(s)
- Ke Si
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Da Lu
- Department of Vascular Surgery, Shanghai General Hospital, Shanghai, People's Republic of China
| | - Jianbo Tian
- Institute of Information Engineering, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
6
|
Zhao K, Wu T, Yang C, Pan H, Xu T, Zhang J, Guo X, Tu J, Zhang D, Kong X, Zhou B, Sun W. Low-intensity pulsed ultrasound prevents angiotensin II-induced aortic smooth muscle cell phenotypic switch via hampering miR-17-5p and enhancing PPAR-γ. Eur J Pharmacol 2021; 911:174509. [PMID: 34547245 DOI: 10.1016/j.ejphar.2021.174509] [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: 07/07/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 10/20/2022]
Abstract
Vascular events can trigger a pathological phenotypic switch in vascular smooth muscle cells (VSMCs), decreasing and disrupting the plasticity and diversity of vascular networks. The development of novel therapeutic approaches is necessary to prevent these changes. We aimed to investigate the effects and associated mechanisms of low-intensity pulsed ultrasound (LIPUS) irradiation on the angiotensin II (AngII)-induced phenotypic switch in VSMCs. In vivo, AngII was infused subcutaneously for 4 weeks to stimulate vascular remodeling in mice, and LIPUS irradiation was applied for 20 min every 2 days for 4 weeks. In vitro, cultured rat aortic VSMCs (RAVSMCs) were pretreated once with LIPUS irradiation for 20 min before 48-h AngII stimulation. Our results showed that LIPUS irradiation prevents AngII-induced vascular remodeling of the whole wall artery without discriminating between adventitia and media in vivo and RAVSMC phenotypic switching in vitro. LIPUS irradiation downregulated miR-17-5p expression and upregulated peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. The PPAR-γ activator rosiglitazone could mimic the favorable effects of LIPUS irradiation on AngII-treated RAVSMCs. In contrast, GW9662 could impede the LIPUS-mediated downregulation of RAVSMC proliferation and inflammation under AngII stimulation conditions in vivo and in vitro. Also, the miR-17-5p agomir has the same effects as GW9662 in vitro. Besides, the inhibitory effects of GW9662 against the anti-remodeling effects of LIPUS irradiation in AngII-induced RAVSMCs could be blocked by pretreatment with the miR-17-5p antagomir. Overall, LIPUS irradiation prevents AngII-induced RAVSMCs phenotypic switching through hampering miR-17-5p and enhancing PPAR-γ, suggesting a new approach for the treatment of vascular disorders.
Collapse
MESH Headings
- Animals
- MicroRNAs/genetics
- MicroRNAs/metabolism
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Angiotensin II/pharmacology
- Male
- Rats
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/radiation effects
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/radiation effects
- Phenotype
- Ultrasonic Waves
- Aorta/drug effects
- Aorta/metabolism
- Aorta/cytology
- Vascular Remodeling/drug effects
- Vascular Remodeling/radiation effects
- Cells, Cultured
- Mice
- Rats, Sprague-Dawley
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tingting Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chuanxi Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China; Department of Cardiology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200082, China
| | - Haotian Pan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tianhua Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jing Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics, Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics, Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Dong Zhang
- Key Laboratory of Modern Acoustics, Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Bin Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China; Department of Genetics, Pediatrics and Medicine Cardiology, Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
7
|
Ding YC, Zhang XJ, Zhang JX, Zhai ZY, Zhang MX, Jiang BH. Progression and Regression of Abdominal Aortic Aneurysms in Mice. Curr Med Sci 2021; 41:901-908. [PMID: 34643880 DOI: 10.1007/s11596-021-2425-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 05/06/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a significant medical problem with a high mortality rate. Nevertheless, the underlying mechanism for the progression and regression of AAA is unknown. METHODS Experimental model of AAA was first created by porcine pancreatic elastase incubation around the infrarenal aorta of C57BL/6 mice. Then, AAA progression and regression were evaluated based on the diameter and volume of AAA. The aortas were harvested for hematoxylin-eosin staining (HE), orcein staining, sirius red staining, immunofluorescence analysis and perls' prussian blue staining at the indicated time point. Finally, β-aminopropionitrile monofumarate (BAPN) was used to explore the underlying mechanism of the regression of AAA. RESULTS When we extended the observation period to 100 days, we not only observed an increase in the AAA diameter and volume in the early stage, but also a decrease in the late stage. Consistent with AAA diameter and volume, the aortic thickness showed the same tendency based on HE staining. The elastin and collagen content first degraded and then regenerated, which corresponds to the early deterioration and late regression of AAA. Then, endogenous up-regulation of lysyl oxidase (LOX) was detected, accompanying the regression of AAA, as detected by an immunofluorescent assay. BAPN and LOX inhibitor considerably inhibited the regression of AAA, paralleling the degradation of elastin lamella and collagen. CONCLUSION Taken together, we tentatively conclude that endogenous re-generation of LOX played an influential role in the regression of AAA. Therefore, regulatory factors on the generation of LOX exhibit promising therapeutic potential against AAA.
Collapse
Affiliation(s)
- Yu-Chao Ding
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,The College of Basic Medical Science, China Medical University, Shenyang, 110122, China
| | - Xian-Jing Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ji-Xiu Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zi-Yi Zhai
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,The College of Basic Medical Science, China Medical University, Shenyang, 110122, China
| | - Mei-Xia Zhang
- The College of Basic Medical Science, China Medical University, Shenyang, 110122, China.
| | - Bao-Hong Jiang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
8
|
Wågsäter D, Ravn H, Wanhainen A, Isaksson H, Björck M. Circulating microRNA in patients with popliteal and multiple artery aneurysms. JVS Vasc Sci 2021; 2:129-135. [PMID: 34617063 PMCID: PMC8489194 DOI: 10.1016/j.jvssci.2021.04.003] [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: 12/17/2020] [Accepted: 04/13/2021] [Indexed: 11/23/2022] Open
Abstract
Background Patients with popliteal artery aneurysm (PA) often have multiple aneurysms, such as bilateral disease or a concomitant abdominal aortic aneurysm (AAA). microRNAs (miRs) are regulators of biological processes and have been investigated as biomarkers for AAA. The aim of this study was to explore if the presence of multiple aneurysms and/or location correlated with miR levels in blood. Methods Using quantitative polymerase chain reaction, 23 miRs were analyzed in plasma from 183 patients with PA. Results Fifteen of the miRs were associated with the number and/or location of aneurysms (1.3- to 2.1-fold changes). Levels of miR-93 (1.4-fold) and miR-215 (1.6- to 1.9-fold) were changed in all compared groups. MiR-24 and miR-23a were altered in those with AAA (1.4- and 1.5-fold, respectively) or bilateral PA (1.5- and 1.4-fold, respectively), compared with in those without. MiR-145 were significantly altered (1.7-fold) in those with isolated PA and AAA, whereas miR-326 were altered in those with bilateral (2.3-fold) and isolated PA (1.9-fold). Conclusions Different miRs seem to be important or to be markers for different subgroups of patients with PA. The identified miRs target vascular smooth muscle cell proliferation and vascular inflammation. Further studies are needed to increase the understanding of the pathogenesis of aneurysmal disease. Patients with popliteal artery aneurysm often have multiple aneurysms, such as bilateral disease or concomitant abdominal aortic aneurysms, but the molecular pathogenesis of the disease is not fully understood. MicroRNAs are important regulators of gene expression and biological processes and have recently been investigated as possible biomarkers for abdominal aortic aneurysm. This study identified 11 microRNAs that were altered in subgroups of patients with popliteal artery aneurysm, which could be important regulators to study in interventional studies.
Collapse
Affiliation(s)
- Dick Wågsäter
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Correspondence: Dick Wågsäter, PhD, Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Hans Ravn
- Department of Surgical Sciences, Section of Vascular Surgery, Uppsala University, Uppsala, Sweden
- Department of Vascular Surgery, Hospital Lillebaelt, University of Southern Denmark, Kolding, Denmark
| | - Anders Wanhainen
- Department of Surgical Sciences, Section of Vascular Surgery, Uppsala University, Uppsala, Sweden
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Helena Isaksson
- School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Martin Björck
- Department of Surgical Sciences, Section of Vascular Surgery, Uppsala University, Uppsala, Sweden
| |
Collapse
|
9
|
Pan L, Niu Z, Gao Y, Wang L, Liu Z, Liu J, Sun J, Pei H. Silencing of CREB Inhibits HDAC2/TLR4/NF-κB Cascade to Relieve Severe Acute Pancreatitis-Induced Myocardial Injury. Inflammation 2021; 44:1565-1580. [PMID: 33725236 DOI: 10.1007/s10753-021-01441-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 02/06/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
The purpose of the present study is to investigate the role of CREB in cardiomyocytes proliferation in regulation of HDAC2-dependent TLR4/NF-κB pathway in severe acute pancreatitis (SAP)-induced myocardial injury. The SAP rat model was developed by injecting sodium touracholate into SD rats and then infected with lentivirus vectors expressing sh-CREB in the presence/absence of LPS. The pathological alterations of rat pancreatic and cardiac tissues were observed by HE staining. TUNEL assay was used to study apoptosis of cardiomyocytes. Next, the loss- and gain-function assay was conducted in LPS-induced myocardial injury cardiomyocytes to define the roles of CREB, HDAC2, and TLR4 in cardiomyocyte proliferation, apoptosis, inflammation, and myocardial injury in vitro. ChIP assay was used to study the enrichment of CREB bound to HDAC2 promoter. RT-qPCR and Western blot analysis were used to detect the expressions of related mRNA and proteins in the NF-κB pathway, respectively. CREB was found to be overexpressed in both SAP tissues and cells. CREB directly bound to the promoter of HDAC2 and activated its expression. Overexpressed CREB or HDAC2 inhibited proliferation and promoted apoptosis of cardiomyocytes. Suppression of CREB inhibited the HDAC2/TLR4/NF-κB cascade to promote proliferation and inhibit apoptosis of cardiomyocytes. The in vitro results were validated in vivo experiments. Coherently, suppression of CREB can inhibit HDAC2/TLR4/NF-κB cascade to promote cardiomyocyte proliferation, thus ameliorating SAP-induced myocardial injury.
Collapse
Affiliation(s)
- Longfei Pan
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, 710004, Xi'an, Shaanxi Province, People's Republic of China.
| | - Zequn Niu
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, 710004, Xi'an, Shaanxi Province, People's Republic of China
| | - Yanxia Gao
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, 710004, Xi'an, Shaanxi Province, People's Republic of China
| | - Liming Wang
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, 710004, Xi'an, Shaanxi Province, People's Republic of China
| | - Zhong Liu
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, 710004, Xi'an, Shaanxi Province, People's Republic of China
| | - Jie Liu
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, 710004, Xi'an, Shaanxi Province, People's Republic of China
| | - Jiangli Sun
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, 710004, Xi'an, Shaanxi Province, People's Republic of China
| | - Honghong Pei
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xincheng District, 710004, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
10
|
Schellinger IN, Dannert AR, Mattern K, Raaz U, Tsao PS. Unresolved Issues in RNA Therapeutics in Vascular Diseases With a Focus on Aneurysm Disease. Front Cardiovasc Med 2021; 8:571076. [PMID: 33937351 PMCID: PMC8081859 DOI: 10.3389/fcvm.2021.571076] [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: 06/09/2020] [Accepted: 02/23/2021] [Indexed: 12/20/2022] Open
Abstract
New technologies have greatly shaped the scientific and medical landscape within the last years. The unprecedented expansion of data and information on RNA biology has led to the discovery of new RNA classes with unique functions and unexpected modifications. Today, the biggest challenge is to transfer the large number of findings in basic RNA biology into corresponding clinical RNA-based therapeutics. Lately, this research begins to yield positive outcomes. RNA drugs advance to the final phases of clinical trials or even receive FDA approval. Furthermore, the introduction of the RNA-guided gene-editing technology CRISPR and advances in the delivery of messenger RNAs have triggered a major progression in the field of RNA-therapeutics. Especially short interfering RNAs and antisense oligonucleotides are promising examples for novel categories of therapeutics. However, several issues need to be addressed including intracellular delivery, toxicity, and immune responses before utilizing RNAs in a clinical setting. In this review, we provide an overview on opportunities and challenges for clinical translation of RNA-based therapeutics, with an emphasis on advances in novel delivery technologies and abdominal aortic aneurysm disease where non-coding RNAs have been shown to play a crucial regulatory role.
Collapse
Affiliation(s)
- Isabel N Schellinger
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany.,Department for Endocrinology, Nephrology and Rheumatology, University Medical Center Leipzig, University of Leipzig, Leipzig, Germany.,Department for Angiology, University Medical Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Angelika R Dannert
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany
| | - Karin Mattern
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany
| | - Uwe Raaz
- Department of Cardiology and Pneumology, Heart Center at the University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK) e.V., Partner Site Göttingen, Göttingen, Germany
| | - Philip S Tsao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Veteran Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| |
Collapse
|
11
|
MicroRNA-194 acts as a suppressor during abdominal aortic aneurysm via inhibition of KDM3A-mediated BNIP3. Life Sci 2021; 277:119309. [PMID: 33662431 DOI: 10.1016/j.lfs.2021.119309] [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: 12/14/2020] [Revised: 02/19/2021] [Accepted: 02/26/2021] [Indexed: 11/21/2022]
Abstract
AIMS Abdominal aortic aneurysm (AAA) is a serious disorder with a high disability rates and mortality rates. Accumulating evidence has identified the vital functions of microRNAs (miRNAs) in the treatment of AAA. Hence, this study is aimed at exploring the modulatory role of miR-194 in the development of AAA. MAIN METHODS After the establishment of mouse AAA models, the expression of miR-194 was determined by quantitative reverse transcription polymerase chain reaction (RT-qPCR), while lysine demethylase 3A (KDM3A) was determined by Western blot analysis in vascular smooth muscle cells (VSMCs) from the abdominal aorta. Cell apoptosis, levels of inflammatory factors as well as expressions of matrix metallopeptidase 2 (MMP2) and matrix metallopeptidase 9 (MMP9) were measured after altering the expression of miR-194 and KDM3A in VSMCs. Moreover, the interactions among miR-194, KDM3A, and BCL2 interacting protein 3 (BNIP3) were investigated by chromatin immunoprecipitation (ChIP) assay and dual-luciferase reporter gene assay. KEY FINDINGS miR-194 was poorly expressed while the expression of KDM3A was up-regulated in mice with AAA. miR-194 inhibited the expression of KDM3A while BNIP3 was positively mediated by KDM3A. More importantly, the number of macrophages was significantly reduced whereas the rate of apoptosis in VSMCs was enhanced. miR-194 reduced the inflammatory response and oxidative stress by repressing KDM3A-mediated BNIP3 expression. SIGNIFICANCES miR-194 played a suppressive role in the progression of AAA by inhibiting the expression of BNIP3 via KDM3A, representing a promising target for AAA management.
Collapse
|
12
|
Tang Y, Fan W, Zou B, Yan W, Hou Y, Kwabena Agyare O, Jiang Z, Qu S. TGF-β signaling and microRNA cross-talk regulates abdominal aortic aneurysm progression. Clin Chim Acta 2020; 515:90-95. [PMID: 33388307 DOI: 10.1016/j.cca.2020.12.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
Abdominal aortic aneurysms (AAA) are permanent and irreversible local dilatations of the abdominal aortic wall. Recent data indicate that the transforming growth factor-beta (TGF-β) signaling pathway exerts a protective effect on the development of AAA. Some dysregulated microRNAs (miRNA) also appear involved in the expansion of AAA and miRNA-based therapeutics have been shown to effectively inhibit this process. New evidence has revealed that TGF-β signaling and miRNA interaction may of physiologic and pathophysiologic significance including the progression of AAA. As such, miRNA that regulate TGF-β signaling may hold promise as potential therapeutic targets. This review explores potential crosstalk between TGF-β signaling and miRNA in AAA in order improve our understanding of this pathology and explore development of potential therapeutic targets.
Collapse
Affiliation(s)
- Ying Tang
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Wenjing Fan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Emergency Department, The Second Affiliated Hospital, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Bu Zou
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Wei Yan
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Yangfeng Hou
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China; Clinic Department, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Oware Kwabena Agyare
- International College, Hengyang Medical School, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Zhisheng Jiang
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China
| | - Shunlin Qu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
13
|
Du Z, Wu T, Liu L, Luo B, Wei C. Extracellular vesicles-derived miR-150-5p secreted by adipose-derived mesenchymal stem cells inhibits CXCL1 expression to attenuate hepatic fibrosis. J Cell Mol Med 2020; 25:701-715. [PMID: 33342075 PMCID: PMC7812282 DOI: 10.1111/jcmm.16119] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/30/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022] Open
Abstract
Hepatic fibrosis (HF) is involved in aggravated wound‐healing response as chronic liver injury. Extracellular vesicles (EVs) carrying microRNA (miR) have been reported as therapeutic targets for liver diseases. In this study, we set out to explore whether adipose‐derived mesenchymal stem cells (ADMSCs)‐derived EVs containing miR‐150‐5p affect the progression of HF. Carbon tetrachloride (CCl4) was firstly used to induce HF mouse models in C57BL/6J mice, and activation of hepatic stellate cells (HSCs) was achieved using transforming growth factor β (TGF‐β). EVs were then isolated from ADMSCs and co‐cultured with HSCs. The relationship between miR‐150‐5p and CXCL1 was identified using dual luciferase gene reporter assay. Following loss‐ and gain‐function experimentation, HSC proliferation was examined by MTT assay, and levels of fibrosis‐, HSC activation‐ and apoptosis‐related genes were determined in vitro. Additionally, pathological scores, collagen volume fraction (CVF) as well as levels of inflammation‐ and hepatic injury‐associated genes were determined in in vivo. Down‐regulated miR‐150‐5p and elevated CXCL1 expression levels were detected in HF tissues. ADMSCs‐derived EVs transferred miR‐150‐5p to HSCs. CXCL1 was further verified as the downstream target gene of miR‐150‐5p. Moreover, ADMSCs‐EVs containing miR‐150‐5p markedly inhibited HSC proliferation and activation in vitro. Meanwhile, in vivo experiments also concurred with the aforementioned results as demonstrated by inhibited CVF, reduced inflammatory factor levels and hepatic injury‐associated indicators. Both experiments results were could be reversed by CXCL1 over‐expression. Collectively, our findings indicate that ADMSCs‐derived EVs containing miR‐150‐5p attenuate HF by inhibiting the CXCL1 expression.
Collapse
Affiliation(s)
- Zhiyong Du
- Department of Hepatobiliary and Pancreatic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Guangzhou, China.,The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Tianchong Wu
- Department of Hepatobiliary and Pancreatic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Guangzhou, China.,The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Linsen Liu
- Department of Hepatobiliary and Pancreatic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Guangzhou, China.,The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Biwei Luo
- Department of Hepatobiliary and Pancreatic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Guangzhou, China.,The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Cuifeng Wei
- Department of Endocrinology, Jingmen First People's Hospital, Jingmen, China
| |
Collapse
|
14
|
Lin H, You B, Lin X, Wang X, Zhou D, Chen Z, Chen Y, Wang R. Silencing of long non-coding RNA Sox2ot inhibits oxidative stress and inflammation of vascular smooth muscle cells in abdominal aortic aneurysm via microRNA-145-mediated Egr1 inhibition. Aging (Albany NY) 2020; 12:12684-12702. [PMID: 32629426 PMCID: PMC7377859 DOI: 10.18632/aging.103077] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/29/2020] [Indexed: 12/15/2022]
Abstract
Long non-coding RNAs (lncRNAs) have been largely reported to contribute to the development and progression of abdominal aortic aneurysm (AAA), a common vascular degenerative disease. The present study was set out with the aim to investigate the possible role of lncRNA Sox2ot in the development of AAA. In this study, we found that lncRNA Sox2ot and early growth response factor-1 (Egr1) were highly expressed, while microRNA (miR)-145 was poorly expressed in Ang II-induced AAA mice and oxidative stress-provoked vascular smooth muscle cell (VSMC) model. Egr1 was a potential target gene of miR-145, and lncRNA Sox2ot could competitively bind to miR-145 to upregulate Egr1 expression. Overexpression of miR-145-5p was found to attenuate oxidative stress and inflammation by inhibiting Egr1 both in vivo and in vitro, which was counteracted by lncRNA Sox2ot. Taken together, the present study provides evidence that downregulation of lncRNA Sox2ot suppressed the expression of Egr1 through regulating miR-145, thus inhibiting the development of AAA, highlighting a theoretical basis for AAA treatment.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/cytology
- Aorta, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/metabolism
- Apolipoproteins E/genetics
- Cells, Cultured
- Down-Regulation
- Early Growth Response Protein 1/antagonists & inhibitors
- Early Growth Response Protein 1/metabolism
- Gene Silencing
- Inflammation
- Mice
- Mice, Knockout
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Oxidative Stress/genetics
- Oxidative Stress/physiology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
Collapse
Affiliation(s)
- Huyu Lin
- Department of Cardiovascular Surgery, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, P.R. China
| | - Bin You
- Department of Cardiovascular Surgery, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, P.R. China
| | - Xiandong Lin
- Department of Cardiovascular Surgery, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, P.R. China
| | - Xiaohu Wang
- Department of Cardiology, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, P.R. China
| | - Dongsheng Zhou
- Department of Imaging, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, P.R. China
| | - Zhiqun Chen
- Department of Cardiovascular Surgery, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, P.R. China
| | - Yuanxiang Chen
- Department of Cardiovascular Surgery, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, P.R. China
| | - Ren Wang
- Department of Cardiovascular Surgery, Provincial Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou 350001, P.R. China
| |
Collapse
|
15
|
Yang P, Cai Z, Wu K, Hu Y, Liu L, Liao M. Identification of key microRNAs and genes associated with abdominal aortic aneurysm based on the gene expression profile. Exp Physiol 2019; 105:160-173. [PMID: 31553078 DOI: 10.1113/ep087705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022]
Abstract
NEW FINDINGS What is the central question of this study? The aim was to identify abdominal aortic aneurysm (AAA)-associated microRNAs and their target genes in AAA using microarray analysis. What is the main finding and its importance? The main finding was that miR-145 and miR-30c-2* were found to be downregulated microRNAs in AAA, which could exert suppressive effects on AAA progression, and that miR-145 might target RAC2, whereas miR-30c-2* might target PIK3CD, IL1B and RAC2. The findings obtained from the study provide an enhanced understanding of microRNA as a therapeutic target to limit AAA. ABSTRACT The aim of the study was to identify abdominal aortic aneurysm (AAA)-associated microRNAs (miRNAs) and genes potentially contributing to AAA. Differential analysis was performed to screen out differentially expressed genes (DEGs) and miRNAs in expression datasets of AAA-related miRNAs [GSE51226 (mouse)] and genes [GSE51227 (mouse) and GSE7084 (human)]. Then, gene ontology (GO) enrichment analysis of DEGs was compared with aneurysm-related GO to screen out DEGs related to the disease. The target genes of differential miRNAs were predicted and used to construct a miRNA-DEG regulatory network, followed by Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of target genes. Moreover, the protein-protein interaction network of target genes of miRNAs in the core position (hub-miRNA) with AAA-related genes was constructed to screen out hub genes. Finally, the target relationship between hub-miRNAs and their target genes was verified. There were 20 upregulated miRNAs and 20 downregulated miRNAs in AAA screened from the GSE51226 dataset (mouse). In addition, there were 1154 upregulated genes and 821 downregulated genes in the GSE51227 dataset (mouse), of which 246 DEGs were enriched in aneurysm-related GO entries in AAA. miR-145 and miR-30c-2* were the key miRNAs of AAA, both of which were downregulated in AAA and influenced pathways so as to affect AAA by regulating their respective target genes. The disease-related gene ACTA2 was downregulated, whereas DEGs including PIK3CD, IL1B, RAC2 and SELL were upregulated in AAA. Finally, it was proved that miR-145 targeted RAC2 and SELL, whereas miR-30c-2* targeted PIK3CD, IL1B and RAC2. Taken together, miR-145 and miR-30c-2*, downregulated in AAA, could potentially affect AAA, and miR-145 might target RAC2, whereas miR-30c-2* might target PIK3CD, IL1B and RAC2.
Collapse
Affiliation(s)
- Pu Yang
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| | - Zhou Cai
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| | - Kai Wu
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| | - Yu Hu
- Center for Experimental Medical Research, Third Xiangya Hospital, Central South University, Changsha, 410013, P.R. China
| | - Ling Liu
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| | - Mingmei Liao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, P.R. China
| |
Collapse
|
16
|
Miyake T, Miyake T, Kurashiki T, Morishita R. Molecular Pharmacological Approaches for Treating Abdominal Aortic Aneurysm. Ann Vasc Dis 2019; 12:137-146. [PMID: 31275464 PMCID: PMC6600097 DOI: 10.3400/avd.ra.18-00076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 02/07/2019] [Indexed: 12/12/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is considered to be a potent life-threatening disorder in elderly individuals. Although many patients with a small AAA are detected during routine abdominal screening, there is no effective therapeutic option to prevent the progression or regression of AAA in the clinical setting. Recent advances in molecular biology have led to the identification of several important molecules, including microRNA and transcription factor, in the process of AAA formation. Regulation of these factors using nucleic acid drugs is expected to be a novel therapeutic option for AAA. Nucleic acid drugs can bind to target factors, mRNA, microRNA, and transcription factors in a sequence-specific fashion, resulting in a loss of function of the target molecule at the transcriptional or posttranscriptional level. Of note, inhibition of a transcription factor using a decoy strategy effectively suppresses experimental AAA formation, by regulating the expression of several genes associated with the disease progression. This review focuses on recent advances in molecular therapy of using nucleic acid drugs to treat AAA.
Collapse
Affiliation(s)
- Takashi Miyake
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tetsuo Miyake
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tomohiro Kurashiki
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
17
|
Smooth muscle-specific LKB1 deletion exaggerates angiotensin II-induced abdominal aortic aneurysm in mice. J Mol Cell Cardiol 2019; 130:131-139. [DOI: 10.1016/j.yjmcc.2019.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/12/2019] [Accepted: 03/27/2019] [Indexed: 11/19/2022]
|
18
|
Abstract
Abdominal aortic aneurysm (AAA) is a local dilatation of the abdominal aortic vessel wall and is among the most challenging cardiovascular diseases as without urgent surgical intervention, ruptured AAA has a mortality rate of >80%. Most patients present acutely after aneurysm rupture or dissection from a previously asymptomatic condition and are managed by either surgery or endovascular repair. Patients usually are old and have other concurrent diseases and conditions, such as diabetes mellitus, obesity, and hypercholesterolemia making surgical intervention more difficult. Collectively, these issues have driven the search for alternative methods of diagnosing, monitoring, and treating AAA using therapeutics and less invasive approaches. Noncoding RNAs-short noncoding RNAs (microRNAs) and long-noncoding RNAs-are emerging as new fundamental regulators of gene expression. Researchers and clinicians are aiming at targeting these microRNAs and long noncoding RNAs and exploit their potential as clinical biomarkers and new therapeutic targets for AAAs. While the role of miRNAs in AAA is established, studies on long-noncoding RNAs are only beginning to emerge, suggesting their important yet unexplored role in vascular physiology and disease. Here, we review the role of noncoding RNAs and their target genes focusing on their role in AAA. We also discuss the animal models used for mechanistic understanding of AAA. Furthermore, we discuss the potential role of microRNAs and long noncoding RNAs as clinical biomarkers and therapeutics.
Collapse
Affiliation(s)
- Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering,
Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Reinier A. Boon
- Institute for Cardiovascular Regeneration, Center of
Molecular Medicine, Goethe University, Frankfurt, Germany
- Department of Physiology, Amsterdam Cardiovascular
Sciences, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The
Netherlands
- German Center of Cardiovascular Research DZHK, Frankfurt,
Germany
| | - Lars Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm,
Sweden
- Department of Vascular and Endovascular Surgery, Technical
University Munich, Munich, Germany
- German Center for Cardiovascular Research DZHK, Munich,
Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Center of
Molecular Medicine, Goethe University, Frankfurt, Germany
- German Center of Cardiovascular Research DZHK, Frankfurt,
Germany
- Corresponding authors: Hanjoong Jo, PhD, John and Jan Portman
Professor, Wallace H. Coulter Department of Biomedical Engineering, Emory
University and Georgia Institute of Technology, 1760 Haygood Drive, Atlanta, GA
30322, , Stefanie Dimmeler, PhD, Institute for
Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University
Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt, Germany,
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering,
Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Division of Cardiology, Emory University, Atlanta, GA,
USA
- Corresponding authors: Hanjoong Jo, PhD, John and Jan Portman
Professor, Wallace H. Coulter Department of Biomedical Engineering, Emory
University and Georgia Institute of Technology, 1760 Haygood Drive, Atlanta, GA
30322, , Stefanie Dimmeler, PhD, Institute for
Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University
Frankfurt, Theodor Stern Kai 7, 60590, Frankfurt, Germany,
| |
Collapse
|
19
|
Peng J, He X, Zhang L, Liu P. MicroRNA‑26a protects vascular smooth muscle cells against H2O2‑induced injury through activation of the PTEN/AKT/mTOR pathway. Int J Mol Med 2018; 42:1367-1378. [PMID: 29956734 PMCID: PMC6089772 DOI: 10.3892/ijmm.2018.3746] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/20/2018] [Indexed: 01/12/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a common disease, which is characterized by the apoptosis of vascular smooth muscle cells (VSMCs). In previous years, microRNAs (miRNAs) have been associated with AAA and functionally implicated in the pathogenesis of this disease. However, the role of miRNAs in the apoptosis of VSMCs remains to be fully elucidated. The present study aimed to elucidate the role and mechanism of miRNAs in protecting against hydrogen peroxide (H2O2)-induced apoptosis in VSMCs. The expression of miRNAs in peripheral blood from patients diagnosed with AAA was analyzed using a microarray and reverse transcription polymerase chain reaction. A VSMC injury model induced by H2O2 was used to determine the potential role of miR-26a against cell injury. Cell viability, cell apoptosis and reactive oxygen species (ROS) generation were determined by a CCK8 assay, flow cytometry and a 2′,7′-DCF diacetate assay, respectively. It was observed that miRNA (miR)-26a (miR-26a-1-5p) was significantly downregulated in peripheral blood samples from patients with AAA. It was revealed that H2O2 treatment dose-dependently inhibited cell viability, enhanced apoptosis and induced the production of ROS, which indicated the success of the model establishment. It was also observed that miR-26a was downregulated in the VSMCs following H2O2 stimulation. The upregulation of miR-26a attenuated H2O2-induced cell injury, as evidenced by the enhancement of cell viability, and inhibition of the activity of caspase-3, apoptosis and ROS production. In addition, phosphatase and tensin homolog (PTEN), a well-known regulator of the AKT/mammalian target of rapamycin (mTOR) pathway, was found to be a direct target of miR-26a in the VSMCs and this was validated using a luciferase reporter assay. Overexpression of PTEN by pcDNA-PTEN plasmids markedly eliminated the protective effects of the overexpression of miR-26a on H2O2-induced cell injury. Finally, it was found that miR-26a mediated its anti-apoptotic action by reactivation of the AKT/mTOR pathway, as demonstrated by the upregulation of phosphorylated (p-)AKT and p-mTOR, and the Akt inhibitor API-2 reversing the protective effects on VSMCs mediated by miR-26a. These results indicated that miR-26a protected VSMCs against H2O2-induced injury through activation of the PTEN/AKT/mTOR pathway, and miR-26a may be considered as a potential prognostic biomarker and therapeutic target in the treatment of AAA.
Collapse
Affiliation(s)
- Junlu Peng
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xinqi He
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Lei Zhang
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Peng Liu
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
20
|
Spin JM, Li DY, Maegdefessel L, Tsao PS. Non-coding RNAs in aneurysmal aortopathy. Vascul Pharmacol 2018; 114:110-121. [PMID: 29909014 DOI: 10.1016/j.vph.2018.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/21/2018] [Accepted: 06/09/2018] [Indexed: 02/07/2023]
Abstract
Aortic aneurysms represent a major public health burden, and currently have no medical treatment options. The pathophysiology behind these aneurysms is complex and variable, depending on location and underlying cause, and generally involves progressive dysfunction of all elements of the aortic wall. Changes in smooth muscle behavior, endothelial signaling, extracellular matrix remodeling, and to a variable extent inflammatory signaling and cells, all contribute to the dilation of the aorta, ultimately resulting in high mortality and morbidity events including dissection and rupture. A large number of researchers have identified non-coding RNAs as crucial regulators of aortic aneurysm development, both in humans and in animal models. While most work to-date has focused on microRNAs, intriguing information has also begun to emerge regarding the role of long-non-coding RNAs. This review summarizes the currently available data regarding the involvement of non-coding RNAs in aneurysmal aortopathies. Going forward, these represent key potential therapeutic targets that might be leveraged in the future to slow or prevent aortic aneurysm formation, progression and rupture.
Collapse
Affiliation(s)
- Joshua M Spin
- Cardiovascular Medicine and Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, USA
| | - Daniel Y Li
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Lars Maegdefessel
- Vascular Biology Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technical University of Munich, Munich, Germany; Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Philip S Tsao
- Cardiovascular Medicine and Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, USA.
| |
Collapse
|
21
|
Alajbegovic A, Holmberg J, Albinsson S. Molecular Regulation of Arterial Aneurysms: Role of Actin Dynamics and microRNAs in Vascular Smooth Muscle. Front Physiol 2017; 8:569. [PMID: 28848449 PMCID: PMC5554360 DOI: 10.3389/fphys.2017.00569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/21/2017] [Indexed: 12/21/2022] Open
Abstract
Aortic aneurysms are defined as an irreversible increase in arterial diameter by more than 50% relative to the normal vessel diameter. The incidence of aneurysm rupture is about 10 in 100,000 persons per year and ruptured arterial aneurysms inevitably results in serious complications, which are fatal in about 40% of cases. There is also a hereditary component of the disease and dilation of the ascending thoracic aorta is often associated with congenital heart disease such as bicuspid aortic valves (BAV). Furthermore, specific mutations that have been linked to aneurysm affect polymerization of actin filaments. Polymerization of actin is important to maintain a contractile phenotype of smooth muscle cells enabling these cells to resist mechanical stress on the vascular wall caused by the blood pressure according to the law of Laplace. Interestingly, polymerization of actin also promotes smooth muscle specific gene expression via the transcriptional co-activator MRTF, which is translocated to the nucleus when released from monomeric actin. In addition to genes encoding for proteins involved in the contractile machinery, recent studies have revealed that several non-coding microRNAs (miRNAs) are regulated by this mechanism. The importance of these miRNAs for aneurysm development is only beginning to be understood. This review will summarize our current understanding about the influence of smooth muscle miRNAs and actin polymerization for the development of arterial aneurysms.
Collapse
Affiliation(s)
- Azra Alajbegovic
- Department of Experimental Medical Science, Lund UniversityLund, Sweden
| | - Johan Holmberg
- Department of Experimental Medical Science, Lund UniversityLund, Sweden
| | | |
Collapse
|