1
|
Tasopoulou KM, Argiriou C, Tsaroucha AK, Georgiadis GS. Circulating miRNAs as biomarkers for diagnosis, surveillance and post-operative follow-up of abdominal aortic aneurysms. Ann Vasc Surg 2023:S0890-5096(23)00144-9. [PMID: 36921794 DOI: 10.1016/j.avsg.2023.02.029] [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: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/15/2023]
Abstract
OBJECTIVE To provide a summary of the current state of research in English medical literature on circulating miRNAs, as biomarkers for AAA. Additionally, for the most commonly mentioned circulating miRNAs in the literature, to attempt a documentation of the biological mechanisms underlying their role in AAA development. METHODS A literature search was undertaken in the MEDLINE database. Only reports that involved peripheral blood samples (whole blood, plasma, serum) were included. The following terms were used in combination: microrna, mirna, abdominal aortic aneurysm, human, circulating, plasma, serum, endovascular and EVAR. RESULTS A total of 25 reports, published from 2012 to 2022 were included with a total of 1259 patients with AAA, predominantly men (N= 1040, 90%). Six of these reports recruited healthy donors who underwent ultrasound screening for AAA as control samples. The majority of studies were undertaken in plasma samples and the most preferred microRNA profiling method was Real - Time quantitative polymerase chain reaction (qRT-PCR). The following nine miRNAs (out of a total of 76) were studied in more than two references: miR-145, miR-24, miR-33, miR-125, let-7, miR-15, miR-191, miR-29 and miR-133. CONCLUSION The nine miRNAs described in this study, are implicated in known pathogenetic mechanisms of AAA such as atherosclerosis, vascular smooth muscle cell phenotype switch and apoptosis, vascular inflammation, extracellular matrix degradation and lipid metabolism. Identifying disease-specific miRNAs, in combination with other clinical parameters, as indicators of AAA, is crucial for early diagnosis as well as follow-up of AAAs. For future research on miRNAs as AAA biomarkers, strict case and control group definitions, sample acquisition protocols, and miRNA expression profiling techniques are warranted.
Collapse
Affiliation(s)
- Kalliopi-Maria Tasopoulou
- Department of Vascular Surgery, Medical School, Democritus University of Thrace, University General Hospital of Evros, Alexandroupolis, Greece.
| | - Christos Argiriou
- Department of Vascular Surgery, Medical School, Democritus University of Thrace, University General Hospital of Evros, Alexandroupolis, Greece
| | - Alexandra K Tsaroucha
- Laboratory of Experimental Surgery and Surgical Research, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - George S Georgiadis
- Department of Vascular Surgery, Medical School, Democritus University of Thrace, University General Hospital of Evros, Alexandroupolis, Greece
| |
Collapse
|
2
|
Yang B, Zhang Z. Suppression of long intergenic non-protein coding RNA 1123 constrains lower extremity deep vein thrombosis via microRNA-125a-3p to target interleukin 1 receptor type 1. Bioengineered 2022; 13:13452-13461. [PMID: 35659191 PMCID: PMC9275874 DOI: 10.1080/21655979.2022.2076496] [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] [Indexed: 12/05/2022] Open
Abstract
Lower extremity deep vein thrombosis (LEDVT) is a disorder of venous return caused by abnormal blood clotting. LEDVT can obstruct the lumen and is the third most common vascular disease after cerebrovascular disease and coronary artery disease. LncRNAs are associated with thrombosis and potentially affect the pathogenesis of DVT. However, no studies have reported the effect of LINC01123 on LEDVT. The aim of this study was to investigate the effect of LINC01123 on LEDVT in rats via the miR-125a-3p/interleukin 1 receptor type 1 (IL1R1) axis. Lentiviral vectors that altering LINC01123, miR-125a-3p and IL1R1 expression were pre-injected into the tail vein of rats, and an LEDVT model was established 1 day later. Detection of LINC01123, miR-125a-3p and IL1R1 expression was performed. Inflammatory factors in femoral venous blood, the length and weight of the thrombus, the histomorphological changes were determined in the rat model. The targeting relation of miR-125a-3p with LINC01123 or IL1R1 was verified. The results presented that LEDVT rats expressed high LINC01123 and IL1R1 and low miR-125a-3p expression levels. After silencing LINC01123 or elevating miR-125a-3p, the rate of thrombosis, length and weight of thrombus, and levels of inflammatory factors were reduced. The targeting relation was presented between miR-125a-3p with LINC01123 or IL1R1. Elevating IL1R1 was available to turn around the action of silence of LINC01123 on LEDVT rats. All in all, suppression of LINC01123 restrains LEDVT via miR-125a-3p to target IL1R1.
Collapse
Affiliation(s)
- Baocai Yang
- Department of General surgery, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
- Department of Vascular Surgery, Yancheng First People’s Hospital of Jiangsu Province, Yancheng, Jiangsu Province, China
| | - ZiXiang Zhang
- Department of General surgery, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| |
Collapse
|
3
|
Song M, Li Y, Chen Z, Zhang J, Yang L, Zhang F, Song C, Miao M, Chang W, Shi H. The Long Non-Coding RNA FAM222A-AS1 Negatively Modulates MiR-Let-7f to Promote Colorectal Cancer Progression. Front Oncol 2022; 12:764621. [PMID: 35646686 PMCID: PMC9133450 DOI: 10.3389/fonc.2022.764621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 04/13/2022] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence indicates that lncRNAs are potential biomarkers and key regulators of tumor development and progression. The present study aimed to screen abnormal expression lncRNAs and investigate the mechanisms underlying the function in the progression of colorectal cancer (CRC). Potential CRC prognosis-associated dysregulated lncRNAs were screened and identified using bioinformatics analysis. Loss/gain-of-function experiments were performed to detect the biological roles of FAM222A-AS1 in CRC cell phenotypes in vitro and in vivo. The potential microRNAs that interact with FAM222A-AS1 were identified using online tools and were verified using qRT-PCR and luciferase reporter assay. The expression of FAM222A-AS1 is significantly upregulated in CRC tumor samples and cell lines. CRC patients with elevated FAM222A-AS1 expression in the tumor samples had unfavorable overall survival and disease-free survival. Silencing FAM222A-AS1 expression significantly inhibited CRC cell proliferation, migration, and invasion both in vitro and in vivo. Furthermore, FAM222A-AS1 was mainly distributed in the cytoplasm. It may directly bound to miR-let-7f and inhibit its expression and upregulate MYH9. In summary, FAM222A-AS1, as a novel oncogene in CRC, may promote the CRC progression by inhibiting miR-let-7f/MYH9 axis. The FAM222A-AS1/miR-let-7f/MYH9 signaling pathway may be a novel valuable target for inhibiting CRC.
Collapse
Affiliation(s)
- Mengmeng Song
- Department of Gastrointestinal Surgery/Clinical Nutrition, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Ye Li
- Department of Digestive Endoscopy, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhewen Chen
- Department of Nutrition, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jie Zhang
- Department of Endocrinology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, China
| | - Liuqing Yang
- Department of Gastrointestinal Surgery/Clinical Nutrition, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Fan Zhang
- Department of Environmental Health, Second Military Medical University, Shanghai, China
| | - Chunhua Song
- Department of Epidemiology and Statistics, Henan Key Laboratory of Tumor Epidemiology College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Mingyong Miao
- Department of Biochemistry, Second Military Medical University, Shanghai, China
- *Correspondence: Hanping Shi, ; Wenjun Chang, ; Mingyong Miao,
| | - Wenjun Chang
- Department of Environmental Health, Second Military Medical University, Shanghai, China
- *Correspondence: Hanping Shi, ; Wenjun Chang, ; Mingyong Miao,
| | - Hanping Shi
- Department of Gastrointestinal Surgery/Clinical Nutrition, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
- *Correspondence: Hanping Shi, ; Wenjun Chang, ; Mingyong Miao,
| |
Collapse
|
4
|
Li Y, Lv M, Lu M, Guan H. miR-124a Involves in the Regulation of Wnt/ β-Catenin and P53 Pathways to Inhibit Abdominal Aortic Aneurysm via Targeting BRD4. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9241959. [PMID: 35096137 PMCID: PMC8799344 DOI: 10.1155/2022/9241959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) belongs to a progressive, gradual aortic rupture, which can lead to death without surgical intervention. The key factors regulating the occurrence and progress of AAA are not clear. Increasing studies have indicated that microRNA (miRNA) plays an important role in cancer development. miR-124a serves as a tumor suppressor in several neoplasms, and its upregulation can greatly inhibit the life activities such as malignant growth and migration of tumor cells. AIM The objective of this study is to explore the association of miR-124a with AAA and to uncover the regulated mechanism of miR-124a on AAA progression. METHODS The specimens from the AAA patients were used for observing the miR-124a expression, and human aortic endothelial cells (hAoECs) were treated with AngII to establish the AAA cell models. The quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), CCK-8, transwell assay, flow cytometry assay, and western blot were conducted to unearth the regulation mechanism of miR-124a on AAA, and the dual-luciferase reporter assay was employed to investigate the downstream target of miR-124a. RESULTS miR-124a was significantly downregulated in the whole blood of the patients, and the decreased miR-124a was also observed in AAA cell models. Overexpressing miR-124a could effectively inhibit the proliferation and migration and promote the apoptosis of the AAA cells. The dual-luciferase reporter assay confirmed that BRD4 was a downstream target of miR-124a, and BRD4 upregulation could obviously reverse the effects of miR-124a on the phenotype of AAA cells. Moreover, it was found that miR-124a could regulate the activities of Wnt/β-catenin and P53 pathways via targeting the BRD4. CONCLUSION Our data suggested that miR-124a could regulate the activities of Wnt/β-catenin and P53 to suppress the AAA progression via targeting the BRD4.
Collapse
Affiliation(s)
- Yunhui Li
- Department of Vascular Surgery, Jinan People's Hospital Affiliated to Shandong First Medical University, China
| | - Meifeng Lv
- Pharmacy Department of Jinan Second Maternal and Child Health Hospital, China
| | - Mingshu Lu
- Department of Vascular Surgery, Jinan People's Hospital Affiliated to Shandong First Medical University, China
| | - Hongliang Guan
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, China
| |
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: 0] [Impact Index Per Article: 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
|
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
|
7
|
MicroRNAs and Long Noncoding RNAs as Novel Therapeutic Targets in Estrogen Receptor-Positive Breast and Ovarian Cancers. Int J Mol Sci 2021; 22:ijms22084072. [PMID: 33920789 PMCID: PMC8071157 DOI: 10.3390/ijms22084072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 01/27/2023] Open
Abstract
Aromatase inhibitors (AIs) such as anastrozole, letrozole, and exemestane have shown to prevent metastasis and angiogenesis in estrogen receptor (ER)-positive breast and ovarian tumors. They function primarily by reducing estrogen production in ER-positive post-menopausal breast and ovarian cancer patients. Unfortunately, current AI-based therapies often have detrimental side-effects, along with acquired resistance, with increased cancer recurrence. Thus, there is an urgent need to identify novel AIs with fewer side effects and improved therapeutic efficacies. In this regard, we and others have recently suggested noncoding RNAs (ncRNAs), specifically microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), as potential molecular targets for utilization in modulating cancer hallmarks and overcoming drug resistance in several cancers, including ER-positive breast and ovarian cancer. Herein, we describe the disruptive functions of several miRNAs and lncRNAs seen in dysregulated cancer metabolism, with a focus on the gene encoding for aromatase (CYP19A1 gene) and estrogen synthesis as a novel therapeutic approach for treating ER-positive breast and ovarian cancers. Furthermore, we discuss the oncogenic and tumor-suppressive roles of several miRNAs (oncogenic miRNAs: MIR125b, MIR155, MIR221/222, MIR128, MIR2052HG, and MIR224; tumor-suppressive miRNAs: Lethal-7f, MIR27B, MIR378, and MIR98) and an oncogenic lncRNA (MIR2052HG) in aromatase-dependent cancers via transcriptional regulation of the CYP19A1 gene. Additionally, we discuss the potential effects of dysregulated miRNAs and lncRNAs on the regulation of critical oncogenic molecules, such as signal transducer, and activator of transcription 3, β-catenin, and integrins. The overall goal of this review is to stimulate further research in this area and to facilitate the development of ncRNA-based approaches for more efficacious treatments of ER-positive breast and ovarian cancer patients, with a slight emphasis on associated treatment–delivery mechanisms.
Collapse
|
8
|
Micro-RNA miR-542-3p suppresses decidualization by targeting ILK pathways in human endometrial stromal cells. Sci Rep 2021; 11:7186. [PMID: 33785768 PMCID: PMC8009905 DOI: 10.1038/s41598-021-85295-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Decidualization of human endometrial stromal cells (HESCs) is a vital step for successful pregnancy. However, the process by which micro-RNAs (miRNAs) regulate decidualization remains elusive. Our current study was designed to identify the mechanism of miRNA miR-542-3p and its potential targets in regulating decidualization. The results showed that miR-542-3p was down-regulated in HESCs. Luciferase assay confirmed that integrin-linked kinase (ILK) is a direct target of miR-542-3p. Overexpression of miR-542-3p resulted in decreased ILK and downstream transforming growth factor β1 (TGF-β1) and SMAD family member 2 (SMAD2) expression. Additional expression of ILK attenuates the miR542-3p-induced down-regulation of TGF-β1 and SMAD2, changes properties such as suppression of proliferation and invasion, and induction of apoptosis, thereby affecting the differentiation of HESCs. Moreover, miR-542-3p overexpression caused down-regulation of the angiogenic factors vascular endothelial growth factor (VEGF), cyclooxygenase-2 (COX-2) and matrix metalloproteinase-9 (MMP-9), and the supernatant of HESCs overexpressing miR-542-3p inhibited the formation of tubular structures in human umbilical vein endothelial cells (HUVECs), suggesting that miR-542-3p inhibits angiogenesis of HUVECs. Furthermore, in our mouse model, following injection of miR-542-3p mimic into the endometrium of mice at pregnancy day 8 (D8), we found decreased miR-542-3p expression and loss of embryo implantation sites. In conclusion, miR-542-3p can affect the process of endometrial decidualization by down-regulating ILK. The present study adds further understanding of the process and regulation of decidualization.
Collapse
|
9
|
Sun B, Li F, Lai S, Zhang X, Wang H, Li Y, Wang W, Chen Y, Liu B, Zheng Y. Inhibition of CXCR2 alleviates the development of abdominal aortic aneurysm in Apo E-/- mice. Acta Cir Bras 2021; 36:e360105. [PMID: 33605308 PMCID: PMC7892195 DOI: 10.1590/acb360105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/19/2020] [Indexed: 01/08/2023] Open
Abstract
PURPOSE To investigate the relationship between atherosclerotic abdominal aortic aneurysm (AAA) and CXC chemokine receptor type 2 (CXCR2). METHODS Mouse AAA model was established by embedding angiotensin-II pump (1000 ng/kg/min) in ApoE-/- mice. Mice were received SB225002, a selective CXCR2 antagonist, for treatment. Blood pressure was recorded, and CXCR2+ macrophages were examined by flow cytometry analysis. Terminal-deoxynucleotidyl transferase mediated nick end labeling (TUNEL) staining was performed to detect cell apoptosis of abdominal aortic aneurysms. Macrophages were isolated from ApoE-/- mice and treated with Ang II and/or SB225002. Dihydroethidium staining was carried out to determine reactive oxygen species (ROS) activity. Enzyme-linked immunosorbent assay (ELISA) was performed to determine the production of IL-1β and TNF-α. The corresponding gene expressions were measured using real-time polymerase chain reaction (PCR), western blot, and immunohistochemistry staining. RESULTS We found that Ang II activated the expression of CXCR2 in monocytes during the formation of AAA. Inhibition of CXCR2 significantly reduced the size of AAA, attenuated inflammation and phenotypic changes in blood vessels. Ang II-induced macrophages exhibited elevated ROS activity, and elevated levels of 1β and TNF-α, which were then partly abolished by SB225002. CONCLUSIONS CXCR2 plays an important role in AAA, suggesting that inhibiting CXCR2 may be a new treatment for AAA.
Collapse
Affiliation(s)
- Bo Sun
- Chinese Academy of Medical Sciences, China; Weifang People’s Hospital, China
| | | | | | - Xu Zhang
- Weifang People’s Hospital, China
| | | | - Yuan Li
- Weifang People’s Hospital, China
| | - Wei Wang
- Weifang People’s Hospital, China
| | | | - Bao Liu
- Weifang People’s Hospital, China
| | | |
Collapse
|
10
|
Identification of Novel microRNA Profiles Dysregulated in Plasma and Tissue of Abdominal Aortic Aneurysm Patients. Int J Mol Sci 2020; 21:ijms21134600. [PMID: 32605321 PMCID: PMC7370113 DOI: 10.3390/ijms21134600] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022] Open
Abstract
microRNAs (miRNAs) are small RNAs that regulate different biological processes. Our objective was to identify miRNAs dysregulated in plasma and tissue of patients with abdominal aortic aneurysm (AAA) and explore new potential targets involved in AAA. Fifty-seven subjects were recruited for a plasma study (30 AAA patients, 16 healthy volunteers and 11 patients with atherosclerosis). The expression level of 179 miRNAs was screened in plasma from a subset of samples, and dysregulated miRNAs were validated in the entire study population. Dysregulated miRNAs were also quantified in aortic tissue of 21 AAA patients and 8 organ donors. Applying a gene set enrichment analysis, an interaction map of dysregulated miRNAs and their targets was built, and selected targets were quantified in tissue samples. miR-27b-3p and miR-221-3p were overexpressed in plasma of AAA patients compared with healthy controls, 1.6 times and 1.9 times, respectively. In AAA tissue, six miRNAs (miR-1, miR-27b-3p, miR-29b-3p, miR-133a-3p, miR-133b, and miR-195-5p) were underexpressed from 1.6 to 4.8 times and four miRNAs (miR-146a-5p, miR-21-5p, miR-144-3p, and miR-103a-3p) were overexpressed from 1.3 to 7.2 times. Thrombospondin-2, a target of miR-195-5p, was increased in AAA tissue and negatively correlated with the expression of miR-195-5p, suggesting their involvement in a common regulatory mechanism.
Collapse
|