1
|
Wiyono AV, Ardinal AP, Raharjo PP. Unraveling the significance of innate inflammation in vascular disease. Int Rev Immunol 2025:1-16. [PMID: 40255209 DOI: 10.1080/08830185.2025.2489346] [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: 04/15/2024] [Revised: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/22/2025]
Abstract
Atheroma formation is initiated by the activation of endothelial and smooth muscle cells, as well as immune cells, including neutrophils, lymphocytes, monocytes, macrophages, and dendritic cells. Monocytes, macrophages, and neutrophils are the innate immune cells that provide a rapid initial line of defence against vascular disease. These cells have a short lifespan and cannot retain memories, making them potential therapeutic targets for the inflammatory process associated with atherosclerosis. In addition, macrophages comprise the majority of vessel wall infiltrates and are, therefore, implicated in all stages of atherosclerosis progression. Neutrophils are the most common type of leukocyte found in circulation, and their high levels of matrix-degrading protease explain their significance in fibrous cap destabilization. However, the activation of immune cells becomes more complex by various microenvironmental stimuli and cytokines, which ultimately transform immune cells into their pro-inflammatory state. Different types of macrophage subsets with distinct functions in inflammation, such as M1 macrophages, cause an increase in pro-inflammatory cytokines and produce reactive oxygen species and nitric oxide, further worsening the disease. This review aims to shed light on immune-mediated inflammation in cardiovascular disease by focusing on the role of macrophage subsets in vascular inflammation and plaque stability, as well as the interaction between neutrophils and monocyte-macrophages.
Collapse
Affiliation(s)
- Alice Valeria Wiyono
- Faculty of Life Sciences & Medicine, King's College London, London, UK
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Rumah Sakit Umum Pusat Hasan Sadikin, Bandung, Indonesia
| | | | - Pradana Pratomo Raharjo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Rumah Sakit Umum Pusat Hasan Sadikin, Bandung, Indonesia
| |
Collapse
|
2
|
Euler G, Parahuleva M. Monocytic microRNAs-Novel targets in atherosclerosis therapy. Br J Pharmacol 2025; 182:206-219. [PMID: 38575391 DOI: 10.1111/bph.16367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/02/2024] [Accepted: 02/16/2024] [Indexed: 04/06/2024] Open
Abstract
Atherosclerosis is a chronic proinflammatory disease of the vascular wall resulting in narrowing of arteries due to plaque formation, thereby causing reduced blood supply that is the leading cause for diverse end-organ damage with high mortality rates. Monocytes/macrophages, activated by elevated circulating lipoproteins, are significantly involved in the formation and development of atherosclerotic plaques. The imbalance between proinflammatory and anti-inflammatory macrophages, arising from dysregulated macrophage polarization, appears to be a driving force in this process. Proatherosclerotic processes acting on monocytes/macrophages include accumulation of cholesterol in macrophages leading to foam cell formation, as well as dysfunctional efferocytosis, all of which contribute to the formation of unstable plaques. In recent years, microRNAs (miRs) were identified as factors that could modulate monocyte/macrophage function and may therefore interfere with the atherosclerotic process. In this review, we present effects of monocyte/macrophage-derived miRs on atherosclerotic processes in order to reveal new treatment options using miRmimics or antagomiRs. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Affiliation(s)
- Gerhild Euler
- Institute of Physiology, Justus Liebig University, Giessen, Germany
| | - Mariana Parahuleva
- Internal Medicine/Cardiology and Angiology, University Hospital of Giessen and Marburg, Marburg, Germany
| |
Collapse
|
3
|
Nazir A, Uwishema O, Shariff S, Franco WXG, El Masri N, Ayele ND, Munyangaju I, Alzain FE, Wojtara M. A Thorough Navigation of miRNA's Blueprint in Crafting Cardiovascular Fate. Health Sci Rep 2024; 7:e70136. [PMID: 39502130 PMCID: PMC11535861 DOI: 10.1002/hsr2.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Cardiovascular diseases contribute significantly to global morbidity and mortality. MicroRNAs are crucial in the development and progression of these diseases by regulating gene expression in various cells and tissues. Their roles in conditions like atherosclerosis, heart failure, myocardial infarction, and arrhythmias have been widely researched. Materials and Methods The present study provides an overview of existing evidence regarding miRNAs' role in cardiovascular disease pathogenesis. Furthermore, the study examines current state-of-the-art technologies used in the study of miRNAs in cardiovascular disease. As a final point, we examine how miRNAs may serve as disease biomarkers, therapeutic targets, and prognostic indicators. Results In cardiology, microRNAs, small noncoding RNA molecules, are crucial to the posttranscriptional regulation of genes. Their role in regulating cardiac cell differentiation and maturation is critical during the development of the heart. They maintain the cardiac function of an adult heart by contributing to its electrical and contractile activity. By binding to messenger RNA molecules, they inhibit protein translation or degrade mRNA. Several cardiovascular diseases are associated with dysregulation of miRNAs, including arrhythmias, hypertension, atherosclerosis, and heart failure. miRNAs can be used as biomarkers to diagnose and predict diseases as well as therapeutic targets. A variety of state-of-the-art technologies have aided researchers in discovering, profiling, and analyzing miRNAs, including microarray analysis, next-generation sequencing, and others. Conclusion Developing new diagnostics and therapeutic approaches is becoming more feasible as researchers refine their understanding of miRNA function. Ultimately, this will reduce the burden of cardiovascular disease around the world.
Collapse
Affiliation(s)
- Abubakar Nazir
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineKing Edward Medical UniversityPakistan
| | - Olivier Uwishema
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
| | - Sanobar Shariff
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineYerevan State Medical UniversityYerevanArmenia
| | - William Xochitun Gopar Franco
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineUniversity of GuadalajaraGuadalajaraMexico
| | - Noha El Masri
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Faculty of MedicineBeirut Arab UniversityLebanon
| | - Nitsuh Dejene Ayele
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of Internal Medicine, Faculty of MedicineWolkite UniversityWolkiteEthiopia
| | - Isabelle Munyangaju
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Barcelona Institute for Global Health—Hospital ClínicUniversitat de Barcelona
| | - Fatima Esam Alzain
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineCollege of Medicine and General Surgery—Sudan University of Science and Technology
| | - Magda Wojtara
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
4
|
Ardinal AP, Wiyono AV, Estiko RI. Unveiling the therapeutic potential of miR-146a: Targeting innate inflammation in atherosclerosis. J Cell Mol Med 2024; 28:e70121. [PMID: 39392102 PMCID: PMC11467738 DOI: 10.1111/jcmm.70121] [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: 05/20/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Atherosclerosis is the foremost vascular disease, precipitating debilitating complications. Although therapeutic strategies have historically focused on reducing cholesterol deposition, recent insights emphasize the pivotal role of inflammation. Innate inflammation significantly contributes to plaque instability and rupture, underscoring the need for intervention across all disease stages. Numerous studies have highlighted the therapeutic potential of targeting innate immune pathways in atherosclerosis, revealing significant advancements in understanding the molecular mechanisms underlying inflammatory processes within arterial lesions. Notably, research has demonstrated that the modulation of microRNA-146a (miR-146a) expression impacts innate inflammation, effectively halts atherosclerosis progression, and enhances plaque stability by targeting interleukin-1 receptor-associated kinase (IRAK) and activating TNF receptor-associated factor 6 (TRAF6), a signalling pathway involving toll-like receptors (TLRs). Understanding the intricate mechanisms involved is crucial. This study provides a comprehensive analysis of the evidence and underlying mechanisms through which miR-146a exerts its effects. Integrating these findings into clinical practice may herald a transformative era in managing atherosclerotic cardiovascular disease.
Collapse
|
5
|
Dutta B, Mahanty M, Kesavalu L, Rahaman SO. Mechanisms underlying TRPV4-mediated regulation of miR-146a expression. Front Immunol 2024; 15:1437540. [PMID: 39403372 PMCID: PMC11471512 DOI: 10.3389/fimmu.2024.1437540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Persistent inflammation is a major contributor in the development of various inflammatory diseases like atherosclerosis. Our study investigates how transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel, interacts with microRNA-146a (miR-146a), within the context of inflammation and atherosclerosis. Micro-RNAs play a critical role in controlling gene expression, and miR-146a is notable for its anti-inflammatory actions. TRPV4 is activated by diverse soluble and mechanical stimuli, and often associated with inflammatory responses in various diseases. Here, we find that TRPV4 negatively regulates miR-146a expression in macrophages, especially following stimulation by lipopolysaccharides or alterations in matrix stiffness. We show that in atherosclerosis, a condition characterized by matrix stiffening, TRPV4 decreases miR-146a expression in aortic tissue macrophages. We find that TRPV4's impact on miR-146a is independent of activation of NFκB, Stat1, P38, and AKT, but is rather mediated through a mechanism involving histone deacetylation instead of DNA methylation at the miR-146a promoter site. Furthermore, we show that N-terminal residues 1 to 130 in TRPV4 is essential in suppression of miR-146a expression in LPS-stimulated macrophages. Altogether, this study identifies a regulatory mechanism of miR-146a expression by TRPV4 which may open new potential therapeutic strategies for managing inflammatory diseases.
Collapse
Affiliation(s)
- Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| | - Manisha Mahanty
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| | - Lakshmyya Kesavalu
- Department of Periodontology and Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, United States
| | - Shaik O. Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| |
Collapse
|
6
|
Raju S, Turner ME, Cao C, Abdul-Samad M, Punwasi N, Blaser MC, Cahalane RM, Botts SR, Prajapati K, Patel S, Wu R, Gustafson D, Galant NJ, Fiddes L, Chemaly M, Hedin U, Matic L, Seidman M, Subasri V, Singh SA, Aikawa E, Fish JE, Howe KL. Multiomics unveils extracellular vesicle-driven mechanisms of endothelial communication in human carotid atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.599781. [PMID: 38979218 PMCID: PMC11230219 DOI: 10.1101/2024.06.21.599781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Background: Carotid atherosclerosis is orchestrated by cell-cell communication that drives progression along a clinical continuum (asymptomatic to symptomatic). Extracellular vesicles (EVs) are cell-derived nanoparticles representing a new paradigm in cellular communication. Little is known about their biological cargo, cellular origin/destination, and functional roles in human atherosclerotic plaque. Methods: EVs were enriched via size exclusion chromatography from human carotid endarterectomy samples dissected into paired plaque and marginal zones (symptomatic n=16, asymptomatic n=13). EV cargos were assessed via whole transcriptome miRNA sequencing and mass spectrometry-based proteomics. EV multi-omics were integrated with bulk and single cell RNA-sequencing (scRNA-seq) datasets to predict EV cellular origin and ligand-receptor interactions, and multi-modal biological network integration of EV-cargo was completed. EV functional impact was assessed with endothelial angiogenesis assays. Results: Carotid plaques contained more EVs than adjacent marginal zones, with differential enrichment for EV-miRNAs and EV-proteins in key atherogenic pathways. EV cellular origin analysis suggested that tissue EV signatures originated from endothelial cells (EC), smooth muscle cells (SMC), and immune cells. Integrated tissue vesiculomics and scRNA-seq indicated complex EV-vascular cell communication that changed with disease progression and plaque vulnerability (i.e., symptomatic disease). Plaques from symptomatic patients, but not asymptomatic patients, were characterized by increased involvement of endothelial pathways and more complex ligand-receptor interactions, relative to their marginal zones. Plaque-EVs were predicted to mediate communication with ECs. Pathway enrichment analysis delineated an endothelial signature with roles in angiogenesis and neovascularization - well-known indices of plaque instability. This was validated functionally, wherein human carotid symptomatic plaque EVs induced sprouting angiogenesis in comparison to their matched marginal zones. Conclusion: Our findings indicate that EVs may drive dynamic changes in plaques through EV- vascular cell communication and effector functions that typify vulnerability to rupture, precipitating symptomatic disease. The discovery of endothelial-directed angiogenic processes mediated by EVs creates new therapeutic avenues for atherosclerosis.
Collapse
|
7
|
Dutta B, Mahanty M, Kesavalu L, Rahaman SO. Mechanisms underlying TRPV4-mediated regulation of miR-146a expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587984. [PMID: 38617263 PMCID: PMC11014524 DOI: 10.1101/2024.04.03.587984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Persistent inflammation is a major contributor in the development of various inflammatory diseases like atherosclerosis. Our study investigates how transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel, interacts with microRNA-146a (miR-146a), within the context of inflammation and atherosclerosis. Micro-RNAs play a critical role in controlling gene expression, and miR-146a is notable for its anti-inflammatory actions. TRPV4 is activated by diverse soluble and mechanical stimuli, and often associated with inflammatory responses in various diseases. Here, we find that TRPV4 negatively regulates miR-146a expression in macrophages, especially following stimulation by lipopolysaccharides or alterations in matrix stiffness. We show that in atherosclerosis, a condition characterized by matrix stiffening, TRPV4 decreases miR-146a expression in aortic tissue macrophages. We find that TRPV4's impact on miR-146a is independent of activation of NFκB, Stat1, P38, and AKT, but is rather mediated through a mechanism involving histone deacetylation instead of DNA methylation at the miR-146a promoter site. Furthermore, we show that N-terminal residues 1 to 130 in TRPV4 is essential in suppression of miR-146a expression in LPS-stimulated macrophages. Altogether, this study identifies a regulatory mechanism of miR-146a expression by TRPV4 which may open new potential therapeutic strategies for managing inflammatory diseases.
Collapse
|
8
|
Shao BZ, Liu MZ, Zhu DN, Yan H, Ke P, Wei W, Han T, Liu C. Depletion of β-arrestin-1 in macrophages enhances atherosclerosis in ApoE -/- mice. Int Immunopharmacol 2023; 125:111085. [PMID: 37866313 DOI: 10.1016/j.intimp.2023.111085] [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: 07/12/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 10/24/2023]
Abstract
Autophagy in atherosclerotic plaque macrophage contributes to the alleviation of atherosclerosis through the promotion of lipid metabolism. β-arrestins are multifunctional proteins participating various kinds of cellular signaling pathways. Here we aimed to determine the role of β-arrestin-1, an important member of β-arrestin family, in atherosclerosis, and whether autophagy was involved in this process. ApoE-/-β-arrestin-1fl/flLysM-Cre mice were created through bone marrow transplantation for the atherosclerosis model with conditional myeloid knocking out β-arrestin-1. Bone marrow-derived macrophages (BMDMs) were used for the in vitro studies. Oil red O staining was used to detect the lesional area. F4/80, Masson trichrome and picro-Sirius red staining were applied for the determination of plaque stability. Real-time PCR was used for the detection of levels of lipid metabolism-related receptors. Electron microscopy and tandem fluorescent mRFP-GFP-LC3 plasmid was applied to test autophagy level. We found that β-arrestin-1 was highly increased in expression in plaque macrophage on the occurrence of atherosclerosis. Conditional myeloid knocking out β-arrestin-1 largely promotes plaque formation and vulnerability. In murine macrophage with lipid loading, knocking down β-arrestin-1 enhanced foam cell formation and levels of plasma and cellular cholesterol, while overexpressing β-arrestin-1 led to the opposite effects. The alleviative effects induced by macrophage β-arrestin-1 in atherosclerosis were involved in autophagy, based on the reduction of autophagy level with the knocking down of macrophage β-arrestin-1 and administration of autophagy inhibitors which largely attenuated the decreasing effect on foam cell formation. Our results demonstrated for the first time that macrophage β-arrestin-1 protected against atherosclerosis through the induction of autophagy.
Collapse
Affiliation(s)
- Bo-Zong Shao
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Meng-Zhen Liu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Dan-Ni Zhu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Hui Yan
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Ping Ke
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Wei Wei
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Ting Han
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Chong Liu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China.
| |
Collapse
|
9
|
Michalak-Stoma A, Walczak K, Adamczyk M, Kowal M, Krasowska D. Selected miRNA and Psoriasis-Cardiovascular Disease (CVD)-Overweight/Obesity Network-A Pilot Study. Int J Mol Sci 2023; 24:13916. [PMID: 37762217 PMCID: PMC10530775 DOI: 10.3390/ijms241813916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Psoriasis is nowadays recognized as a multifactorial systemic disease with complex and not fully understood pathogenesis. In psoriatic patients, the increased cardiovascular disease (CVD) risk and frequent comorbidities like obesity are observed. The aim of this study was to investigate differences in miRNA (miR-22-3p, miR-133a-3p, miR-146a-5p, miR-369-3p, and Let-7b-5p) involved in CVD risk among psoriatic patients with overweight/obesity and with normal weight. The study comprised 28 male psoriatic patients and 16 male healthy controls. miRNA isolated from peripheral blood mononuclear cells was reverse-transcribed and RT-qPCR was performed. We have found decreased levels of miR-22, miR-133a, miR-146a, and miR-369 among the psoriatic patients. There was a statistically significant difference in miR-22 and miR-146a levels between psoriatic patients with overweight/obesity and with normal weight. There were positive correlations between miR-22 and miR-146a levels and psoriatic arthritis (PsA) in psoriatic patients with normal weight and between the miR-133a level and PsA in the overweight/obese patients. The decreased levels of selected miRNA are consistent with the levels observed in CVD indicating their impact on the CVD risk in psoriatic patients. miR-22 and miR-146 may be recognized as one of the contributing factors in the obesity-CVD-psoriasis network.
Collapse
Affiliation(s)
- Anna Michalak-Stoma
- Chair and Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, ul. Staszica 16, 20-081 Lublin, Poland; (K.W.); (M.A.); (M.K.); (D.K.)
| | | | | | | | | |
Collapse
|
10
|
Zapata-Martínez L, Águila S, de los Reyes-García AM, Carrillo-Tornel S, Lozano ML, González-Conejero R, Martínez C. Inflammatory microRNAs in cardiovascular pathology: another brick in the wall. Front Immunol 2023; 14:1196104. [PMID: 37275892 PMCID: PMC10233054 DOI: 10.3389/fimmu.2023.1196104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
The regulatory role of microRNAs (miRNAs) is mainly mediated by their effect on protein expression and is recognized in a multitude of pathophysiological processes. In recent decades, accumulating evidence has interest in these factors as modulatory elements of cardiovascular pathophysiology. Furthermore, additional biological processes have been identified as new components of cardiovascular disease etiology. In particular, inflammation is now considered an important cardiovascular risk factor. Thus, in the present review, we will focus on the role of a subset of miRNAs called inflamma-miRs that may regulate inflammatory status in the development of cardiovascular pathology. According to published data, the most representative candidates that play functional roles in thromboinflammation are miR-21, miR-33, miR-34a, miR-146a, miR-155, and miR-223. We will describe the functions of these miRNAs in several cardiovascular pathologies in depth, with specific emphasis on the molecular mechanisms related to atherogenesis. We will also discuss the latest findings on the role of miRNAs as regulators of neutrophil extracellular traps and their impact on cardiovascular diseases. Overall, the data suggest that the use of miRNAs as therapeutic tools or biomarkers may improve the diagnosis or prognosis of adverse cardiovascular events in inflammatory diseases. Thus, targeting or increasing the levels of adequate inflamma-miRs at different stages of disease could help mitigate or avoid the development of cardiovascular morbidities.
Collapse
|
11
|
Zhang Y, Shan L, Li D, Tang Y, Qian W, Dai J, Du M, Sun X, Zhu Y, Wang Q, Zhou L. Identification of key biomarkers associated with immune cells infiltration for myocardial injury in dermatomyositis by integrated bioinformatics analysis. Arthritis Res Ther 2023; 25:69. [PMID: 37118825 PMCID: PMC10142164 DOI: 10.1186/s13075-023-03052-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Dermatomyositis (DM) is an acquired autoimmune disease that can cause damage to various organs, including the heart muscle. However, the mechanisms underlying myocardial injury in DM are not yet fully understood. METHODS In this study, we utilized publicly available datasets from the Gene Expression Omnibus (GEO) database to identify hub-genes that are enriched in the immune system process in DM and myocarditis. Weighted gene co-expression network analysis (WGCNA), differentially expressed genes (DEGs) analysis, protein-protein interaction (PPI), and gene ontology (GO) analysis were employed to identify these hub-genes. We then used the CIBERSORT method to analyze immune cell infiltration in skeletal muscle specimens of DM and myocardium specimens of myocarditis respectively. Correlation analysis was performed to investigate the relationship between key genes and infiltrating immune cells. Finally, we predicted regulatory miRNAs of hub-genes through miRNet and validated their expression in online datasets and clinical samples. RESULTS Using integrated bioinformatics analysis, we identified 10 and 5 hub-genes that were enriched in the immune system process in the database of DM and myocarditis respectively. The subsequent intersections between hub-genes were IFIT3, OAS3, ISG15, and RSAD2. We found M2 macrophages increased in DM and myocarditis compared to the healthy control, associating with the expression of IFIT3, OAS3, ISG15, and RSAD2 in DM and myocarditis positively. Gene function enrichment analysis (GSEA) showed that IFIT3, OAS3, ISG15, and RSAD2 were mainly enriched in type I interferon (IFN) signaling pathway, cellular response to type I interferon, and response to type I interferon. Finally, we verified that the expression of miR-146a-5p was significantly higher in the DM with myocardial injury than those without myocardial injury (p = 0.0009). CONCLUSION Our findings suggest that IFIT3, OAS3, ISG15, and RSAD2 may play crucial roles in the underlying mechanism of myocardial injury in DM. Serum miR-146a-5p could be a potential biomarker for myocardial injury in DM.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Linwei Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongyu Li
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yinghong Tang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Qian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayi Dai
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengdi Du
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoxuan Sun
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yinsu Zhu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Wang
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Lei Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
Ren Y, Zhang H. Emerging role of exosomes in vascular diseases. Front Cardiovasc Med 2023; 10:1090909. [PMID: 36937921 PMCID: PMC10017462 DOI: 10.3389/fcvm.2023.1090909] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/11/2023] [Indexed: 03/06/2023] Open
Abstract
Exosomes are biological small spherical lipid bilayer vesicles secreted by most cells in the body. Their contents include nucleic acids, proteins, and lipids. Exosomes can transfer material molecules between cells and consequently have a variety of biological functions, participating in disease development while exhibiting potential value as biomarkers and therapeutics. Growing evidence suggests that exosomes are vital mediators of vascular remodeling. Endothelial cells (ECs), vascular smooth muscle cells (VSMCs), inflammatory cells, and adventitial fibroblasts (AFs) can communicate through exosomes; such communication is associated with inflammatory responses, cell migration and proliferation, and cell metabolism, leading to changes in vascular function and structure. Essential hypertension (EH), atherosclerosis (AS), and pulmonary arterial hypertension (PAH) are the most common vascular diseases and are associated with significant vascular remodeling. This paper reviews the latest research progress on the involvement of exosomes in vascular remodeling through intercellular information exchange and provides new ideas for understanding related diseases.
Collapse
Affiliation(s)
- Yi Ren
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Honggang Zhang
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Tie J, Takanari H, Ota K, Okuda T. Role of miR-143 and miR-146 in Risk Evaluation of Coronary Artery Diseases in Autopsied Samples. Genes (Basel) 2023; 14:471. [PMID: 36833398 PMCID: PMC9956587 DOI: 10.3390/genes14020471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/05/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Coronary artery disease (CAD) is a common and fatal cardiovascular disease. Among known CAD risk factors, miRNA polymorphisms, such as Has-miR-143 (rs41291957 C>G) and Has-miR-146a (rs2910164 G>A), have emerged as important genetic markers of CAD. Despite many genetic association studies in multiple populations, no study assessing the association between CAD risk and SNPs of miR-143 and miR-146 was documented in the Japanese people. Therefore, using the TaqMan SNP assay, we investigated two SNP genotypes in 151 subjects with forensic autopsy-proven CAD. After pathological observation, we used ImageJ software to assess the degree of coronary artery atresia. Moreover, the genotypes and miRNA content of the two groups of samples with atresia <10% and >10% were analyzed. The results showed that the CC genotype of rs2910164 was more frequent in patients with CAD than in controls, which was associated with the risk of CAD in the study population. However, Has-miR-143 rs41291957 genotype did not show a clear correlation with the risk of CAD.
Collapse
Affiliation(s)
- Jian Tie
- Department of Legal Medicine, Nihon University School of Medicine, Tokyo 1738610, Japan
| | - Hiroki Takanari
- Department of Interdisciplinary Research for Medicine and Photonics, Institute of Post-LED, Tokushima 7700814, Japan
| | - Koya Ota
- Department of Legal Medicine, Nihon University School of Medicine, Tokyo 1738610, Japan
| | - Takahisa Okuda
- Department of Legal Medicine, Nihon University School of Medicine, Tokyo 1738610, Japan
| |
Collapse
|
14
|
de Los Reyes-García AM, Zapata-Martínez L, Águila S, Lozano ML, Martínez C, González-Conejero R. microRNAs as biomarkers of risk of major adverse cardiovascular events in atrial fibrillation. Front Cardiovasc Med 2023; 10:1135127. [PMID: 36895835 PMCID: PMC9988920 DOI: 10.3389/fcvm.2023.1135127] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Atrial fibrillation is a complex and multifactorial disease. Although prophylactic anticoagulation has great benefits in avoiding comorbidities, adverse cardiovascular events still occur and thus in recent decades, many resources have been invested in the identification of useful markers in the prevention of the risk of MACE in these patients. As such, microRNAs, that are small non-coding RNAs whose function is to regulate gene expression post-transcriptionally, have a relevant role in the development of MACE. miRNAs, have been investigated for many years as potential non-invasive biomarkers of several diseases. Different studies have shown their utility in the diagnosis and prognosis of cardiovascular diseases. In particular, some studies have associated the presence of certain miRNAs in plasma with the development of MACE in AF. Despite these results, there are still many efforts to be done to allow the clinical use of miRNAs. The lack of standardization concerning the methodology in purifying and detecting miRNAs, still provides contradictory results. miRNAs also have a functional impact in MACE in AF through the dysregulation of immunothrombosis. Indeed, miRNAs may be a link between MACE and inflammation, through the regulation of neutrophil extracellular traps that are a key element in the establishment and evolution of thrombotic events. The use of miRNAs as therapy against thromboinflammatory processes should also be a future approach to avoid the occurrence of MACE in atrial fibrillation.
Collapse
Affiliation(s)
- Ascensión M de Los Reyes-García
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - Laura Zapata-Martínez
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - Sonia Águila
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - María L Lozano
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - Constantino Martínez
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| | - Rocío González-Conejero
- Servicio de Hematología y Oncología Médica, Hospital General Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia IMIB Pascual Parrilla, Murcia, Spain
| |
Collapse
|
15
|
Han B, Wang J, Hu F, Liu Y, Sun Y, Meng K, Lu P, Tang H. Functional mechanism of EGR3 in cerebral ischemia/reperfusion injury in rats by modulating transcription of pri-miR-146a/146b to miR-146 and suppressing SORT1 expression. Brain Res 2022; 1797:148096. [PMID: 36150456 DOI: 10.1016/j.brainres.2022.148096] [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: 06/01/2022] [Revised: 09/01/2022] [Accepted: 09/16/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE EGR3 is implicated in angiogenesis in rats with cerebral ischemia/reperfusion injury (CIRI). This research aimed to explore the effect and in vivo and ex vivo mechanisms of EGR3 in CIRI. METHODS CIRI rat models were established via middle cerebral artery occlusion. Cell models were established via oxygen-glucose deprivation/reoxygenation (OGD/R). Brain injury was assessed by neurological scoring, HE, and TTC staining. Inflammatory factors and oxidative stress markers were measured using corresponding kits. Mitochondrial membrane potential and mitochondrial respiration were examined by flow cytometry and respirometry. EGR3-miR-146 network was predicted on TransmiR v2.0 database. Target genes of miR-146 were screened on Starbase, Targetscan, and miRDB databases. miR-146 expression was determined by RT-qPCR. Levels of EGR3 and SORT1 were determined by Western blot. Binding relationships among EGR3, miR-146, and SORT1 were validated by dual-luciferase assay. EGR3, miR-146, and SORT1 levels were altered by injection or cell transfection to observe their functions. RESULTS EGR3 was poorly-expressed in CIRI rats and OGD/R-induced neurons. EGR3 overexpression reduced inflammatory factor levels and attenuated oxidative stress and mitochondrial injury in CIRI rats and OGD/R-induced neurons. EGR3 bound to miR-146b promoter region. EGR3 promoted pri-miR-146a/146b processing and stimulated miR-146 transcription. miR-146 overexpression ameliorated oxidative stress and mitochondrial injury and miR-146 downregulation abolished the effect of EGR3 overexpression in vitro. miR-146 targeted SORT1. SORT1 overexpression invalidated the protective function of miR-146 overexpression on oxidative stress and mitochondrial injury in vitro. CONCLUSION EGR3 protected against CIRI by mitigating oxidative stress and mitochondrial injury via the miR-146/SORT1 axis.
Collapse
Affiliation(s)
- Bin Han
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi Province, China
| | - Jing Wang
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi Province, China
| | - Fengyun Hu
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi Province, China
| | - Yi Liu
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi Province, China
| | - Yaxuan Sun
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi Province, China
| | - Kun Meng
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi Province, China
| | - Pengyu Lu
- Department of Neurology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi Province, China
| | - Haifeng Tang
- Department of Emergency, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China.
| |
Collapse
|
16
|
Scavenger receptor-targeted plaque delivery of microRNA-coated nanoparticles for alleviating atherosclerosis. Proc Natl Acad Sci U S A 2022; 119:e2201443119. [PMID: 36122215 PMCID: PMC9522431 DOI: 10.1073/pnas.2201443119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis treatments by gene regulation are garnering attention, yet delivery of gene cargoes to atherosclerotic plaques remains inefficient. Here, we demonstrate that assembly of therapeutic oligonucleotides into a three-dimensional spherical nucleic acid nanostructure improves their systemic delivery to the plaque and the treatment of atherosclerosis. This noncationic nanoparticle contains a shell of microRNA-146a oligonucleotides, which regulate the NF-κB pathway, for achieving transfection-free cellular entry. Upon an intravenous injection into apolipoprotein E knockout mice fed with a high-cholesterol diet, this nanoparticle naturally targets class A scavenger receptor on plaque macrophages and endothelial cells, contributing to elevated delivery to the plaques (∼1.2% of the injected dose). Repeated injections of the nanoparticle modulate genes related to immune response and vascular inflammation, leading to reduced and stabilized plaques but without inducing severe toxicity. Our nanoparticle offers a safe and effective treatment of atherosclerosis and reveals the promise of nucleic acid nanotechnology for cardiovascular disease.
Collapse
|
17
|
Yu Y, Guo D, Zhao L. MiR-199 Aggravates Doxorubicin-Induced Cardiotoxicity by Targeting TAF9b. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:4364779. [PMID: 35873641 PMCID: PMC9307339 DOI: 10.1155/2022/4364779] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/18/2022]
Abstract
The clinical application of doxorubicin (DOX) is limited because of its cardiotoxicity. However, the pathogenic mechanism of DOX and the role of miRNA in DOX-induced cardiotoxicity remain to be further studied. This study aimed to investigate the role of miR-199 in DOX-mediated cardiotoxicity. A mouse model of myocardial cell injury induced by DOX was established. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expression changes of miR-199 and TATA-binding protein associated factor 9B (TAF9b) in DOX-induced cardiac injury. Cell apoptosis was detected by TUNEL staining and flow cytometry. The expression levels of apoptosis-related proteins, namely, Bax and Bcl-2, were detected by qPCR. The expression of Beclin-1 and LC3b was detected by western blotting. The binding effect of miR-199 with TAF9b was verified by dual-luciferase reporter gene assay. In this study, overexpression of miR-199 could promote cardiotoxicity. Inhibition of miR-199 could alleviate DOX-mediated myocardial injury. Further studies showed that miR-199 targeted TAF9b. Moreover, miR-199 promoted apoptosis of myocardial cells and aggravated autophagy. Furthermore, we demonstrated that TAF9B knockdown reversed the myocardial protective effect of miR-199 inhibitors. Therefore, miR-199 promoted DOX-mediated cardiotoxicity by targeting TAF9b, thereby aggravating apoptosis and regulating autophagy.
Collapse
Affiliation(s)
- Yangsheng Yu
- Department of Cardiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Degang Guo
- Emergency Department, Third People's Hospital of Liaocheng City, Liaocheng 252000, China
| | - Lin Zhao
- Department of Cardiology, Sunshine Union Hospital of Weifang, Weifang 261000, Shandong, China
| |
Collapse
|
18
|
Yang D, Haemmig S, Chen J, McCoy M, Cheng HS, Zhou H, Pérez-Cremades D, Cheng X, Sun X, Haneo-Mejia J, Vellarikkal SK, Gupta RM, Barrera V, Feinberg MW. Endothelial cell-specific deletion of a microRNA accelerates atherosclerosis. Atherosclerosis 2022; 350:9-18. [PMID: 35462240 PMCID: PMC10165557 DOI: 10.1016/j.atherosclerosis.2022.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/29/2022] [Accepted: 04/07/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Chronic vascular endothelial inflammation predisposes to atherosclerosis; however, the cell-autonomous roles for endothelial-expressing microRNAs (miRNAs) are poorly understood in this process. MiR-181b is expressed in several cellular constituents relevant to lesion formation. The aim of this study is to examine the role of genetic deficiency of the miR-181b locus in endothelial cells during atherogenesis. METHODS AND RESULTS Using a proprotein convertase subtilisin/kexin type 9 (PCSK9)-induced atherosclerosis mouse model, we demonstrated that endothelial cell (EC)-specific deletion of miR-181a2b2 significantly promoted atherosclerotic lesion formation, cell adhesion molecule expression, and the influx of lesional macrophages in the vessel wall. Yet, endothelium deletion of miR-181a2b2 did not affect body weight, lipid metabolism, anti-inflammatory Ly6Clow or the pro-inflammatory Ly6Cinterm and Ly6Chigh fractions in circulating peripheral blood mononuclear cells (PBMCs), and pro-inflammatory or anti-inflammatory mediators in both bone marrow (BM) and PBMCs. Mechanistically, bulk RNA-seq and gene set enrichment analysis of ECs enriched from the aortic arch intima, as well as single cell RNA-seq from atherosclerotic lesions, revealed that endothelial miR-181a2b2 serves as a critical regulatory hub in controlling endothelial inflammation, cell adhesion, cell cycle, and immune response during atherosclerosis. CONCLUSIONS Our study establishes that deficiency of a miRNA specifically in the vascular endothelium is sufficient to profoundly impact atherogenesis. Endothelial miR-181a2b2 deficiency regulates multiple key pathways related to endothelial inflammation, cell adhesion, cell cycle, and immune response involved in the development of atherosclerosis.
Collapse
Affiliation(s)
- Dafeng Yang
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Stefan Haemmig
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jingshu Chen
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael McCoy
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Henry S Cheng
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Haoyang Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Daniel Pérez-Cremades
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiao Cheng
- Department of Biochemistry, University of Nebraska - Lincoln, Lincoln, NE, 68588, USA
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska - Lincoln, Lincoln, NE, 68588, USA
| | - Jorge Haneo-Mejia
- Department of Pathology and Laboratory Medicine and Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Shamsudheen K Vellarikkal
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajat M Gupta
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor Barrera
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Howe KL, Cybulsky M, Fish JE. The Endothelium as a Hub for Cellular Communication in Atherogenesis: Is There Directionality to the Message? Front Cardiovasc Med 2022; 9:888390. [PMID: 35498030 PMCID: PMC9051343 DOI: 10.3389/fcvm.2022.888390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells line every blood vessel and thereby serve as an interface between the blood and the vessel wall. They have critical functions for maintaining homeostasis and orchestrating vascular pathogenesis. Atherosclerosis is a chronic disease where cholesterol and inflammatory cells accumulate in the artery wall below the endothelial layer and ultimately form plaques that can either progress to occlude the lumen or rupture with thromboembolic consequences - common outcomes being myocardial infarction and stroke. Cellular communication lies at the core of this process. In this review, we discuss traditional (e.g., cytokines, chemokines, nitric oxide) and novel (e.g., extracellular vesicles) modes of endothelial communication with other endothelial cells as well as circulating and vessel wall cells, including monocytes, macrophages, neutrophils, vascular smooth muscle cells and other immune cells, in the context of atherosclerosis. More recently, the growing appreciation of endothelial cell plasticity during atherogenesis suggests that communication strategies are not static. Here, emerging data on transcriptomics in cells during the development of atherosclerosis are considered in the context of how this might inform altered cell-cell communication. Given the unique position of the endothelium as a boundary layer that is activated in regions overlying vascular inflammation and atherosclerotic plaque, there is a potential to exploit the unique features of this group of cells to deliver therapeutics that target the cellular crosstalk at the core of atherosclerotic disease. Data are discussed supporting this concept, as well as inherent pitfalls. Finally, we briefly review the literature for other regions of the body (e.g., gut epithelium) where cells similarly exist as a boundary layer but provide discrete messages to each compartment to govern homeostasis and disease. In this light, the potential for endothelial cells to communicate in a directional manner is explored, along with the implications of this concept - from fundamental experimental design to biomarker potential and therapeutic targets.
Collapse
Affiliation(s)
- Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Myron Cybulsky
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Zhou J, Liu L, Wu P, Zhao L, Wu Y. Fusobacterium nucleatum Accelerates Atherosclerosis via Macrophage-Driven Aberrant Proinflammatory Response and Lipid Metabolism. Front Microbiol 2022; 13:798685. [PMID: 35359716 PMCID: PMC8963492 DOI: 10.3389/fmicb.2022.798685] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/03/2022] [Indexed: 12/18/2022] Open
Abstract
Periodontitis, an oral chronic inflammatory disease, is reported to show an association with atherosclerotic vascular disease. Fusobacterium nucleatum is an oral commensal bacterium that is abundantly implicated in various forms of periodontal diseases; however, its role in the pathogenesis of atherosclerosis is unclear. This study aimed to elucidate the underlying pathogenic mechanisms of atherosclerosis induced by F. nucleatum to provide new insight on the prevention and treatment of atherosclerosis. We used an animal model, that is, ApoE–/– mice were infected with F. nucleatum by oral gavage, and in vitro co-culture models to assess the pathogenicity of F. nucleatum. The results indicate that F. nucleatum ATCC 25586 invaded aortic tissues and substantially increased the progression of atherosclerotic lesions. In addition, F. nucleatum changed plaque composition into a less-stable phenotype, characterized with increased subcutaneous macrophage infiltration, M1 polarization, lipid deposition, cell apoptosis, and reduced extracellular matrix and collagen content. The serum levels of pro-atherosclerotic factors, such as interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, monocyte chemoattractant protein-1 (MCP-1), c-reactive protein, and oxidized low-density lipoprotein (ox-LDL) and microRNAs (miR-146a, miR-155, and miR-23b) were considerably increased after F. nucleatum stimulation, whereas HDL-c level was reduced. F. nucleatum induced in vitro macrophage apoptosis in a time- and dose-dependent manner. F. nucleatum facilitated ox-LDL–induced cholesterol phagocytosis and accumulation by regulating the expression of lipid metabolism-related genes (AR-A1, ACAT1, ABCA1, and ABCG1). F. nucleatum further worsened the atherosclerotic plaque microenvironment by considerably increasing the levels of IL-6; IL-1β; TNF-α; MCP-1; and MMP-2, 8, and 9 and by suppressing fibronectin (FN) 1 levels during foam cell formation. This study shows that F. nucleatum ATCC 25586 is implicated in atherosclerosis by causing aberrant activation and lipid metabolism in macrophage.
Collapse
Affiliation(s)
- Jieyu Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiyao Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
miRNA-29a inhibits atherosclerotic plaque formation by mediating macrophage autophagy via PI3K/AKT/mTOR pathway. Aging (Albany NY) 2022; 14:2418-2431. [PMID: 35288486 PMCID: PMC8954956 DOI: 10.18632/aging.203951] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 02/28/2022] [Indexed: 11/25/2022]
Abstract
Background: miR-29a plays a vital role in AS, but the relationship between the miR-29a-targeted PI3K signaling pathway and AS remains unclear. Therefore, this study was carried out. Methods: Gene expression profiles from the GEO database containing AS samples were analyzed. ApoE−/− mice and RAW264.7 cells were treated with miR-29a negative control (NC), miR-29a mimic and miR-29a inhibitor to establish the AS model. Then MOVAT staining, TEM, Western blotting, and immunofluorescence staining were adopted for testing target proteins. Results: DEGs were identified from GSE137578, GSE132651, GSE113969, GSE43292, and GSE97210 datasets. It was found that there were targeted binding sites between miR-29a and PIK3CA. Besides, GO and KEGG analysis demonstrated that autophagy was an enriched pathway in AS. Later, PPI network was depicted, and hub genes were then determined. The results revealed that miR-29a suppressed the areas of plaques and lesional macrophages, but had no impact on VSMCs. TEM results showed the organelles pyknosis of lesional macrophages damaged morphological changes. Furthermore, miR-29a amplified the M2-like macrophages but suppressed the polarization of M1-like macrophages in atherosclerotic plaques. According to mouse and RAW 264.7 cell experiments, miR-29a significantly inhibited the protein expressions of PI3K, p-PI3K, p-AKT, and p-mTOR, which were consistent with the increased expressions of autophagy-related proteins, Beclin 1 and LC3II. However, the miR-29a suppression exhibited the contrary results. Conclusion: MiR-29a elevation induces the increase of autophagy by down-regulating the PI3K/AKT/mTOR pathway in the progression of AS, indicating that miR-29a is a novel therapeutic strategy for AS.
Collapse
|
22
|
Biochemical Characterization and Molecular Determination of Estrogen Receptor-α (ESR1 PvuII-rs2234693 T>C) and MiRNA-146a (rs2910164 C>G) Polymorphic Gene Variations and Their Association with the Risk of Polycystic Ovary Syndrome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19053114. [PMID: 35270805 PMCID: PMC8910123 DOI: 10.3390/ijerph19053114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is regarded as one of the most frequently encountered endocrine disorders and affects millions of young women worldwide, resulting in an array of complex metabolic alterations and reproductive failure. PCOS is a risk factor for diabetes mellitus, obstructive sleep apnea, obesity and depression in patients. Estrogen receptors (ESRs) are significant candidates in endocrine function and ovarian response in women. Moreover, microRNAs and long non-coding RNAs are emerging as principal mediators of gene expression and epigenetic pathways in various disease states. This study has characterized the clinical parameters in PCOS patients with comprehensive biochemical profiling compared to healthy controls and further examined the influence of allelic variations for estrogen receptor-α (ESR1 PvuII-rs2234693 T>C) and miRNA-146a (rs2910164 C>G) gene polymorphism on the risk of and susceptibility to PCOS. In this case-control study, we have used amplification refractory mutation specific (ARMS)-PCR to detect and determine the presence of these polymorphic variants in the study subjects. Our results demonstrated that most of the biochemical markers, which were analyzed in the study, show statistically significant alterations in PCOS patients, including fasting glucose, free insulin, HOMA-IR, LDL, HDL, cholesterol and hormones such as FSH, LH, testosterone and progesterone, which correlate with the established biochemical alterations in the disorder. Further, it is reported that for estrogen receptor-α (ESR1 PvuII-rs2234693 T>C), the frequency of the T allele (fT) was significantly higher among patients (0.64 vs. 0.44) compared to controls, while the frequency of the C allele (fC) was lower in patients (0.36 vs. 0.56) compared to controls. However, it was found that there was no association of an increased risk of PCOS with the ESR1 PvuII-rs2234693 C>T gene polymorphism. On the contrary, the study found strong association of miRNA-146a (rs2910164 C>G) gene polymorphism with an enhanced risk of PCOS. The frequency of the C allele (fC) was significantly higher among patients (0.52 vs. 0.36) compared to controls. The frequency of the G allele (fG) was found to be lower in patients (0.48 vs. 0.64) compared to controls. The codominant, dominant and recessive models display a statistically significant association of polymorphic variations with PCOS. Moreover, the G allele was associated strongly with PCOS susceptibility with an OR = 1.92 (95%) CI = (1.300−2.859), RR = 1.38 (1.130−1.691) p-value < 0.001.
Collapse
|
23
|
Gunter S, Michel FS, Fourie SS, Singh M, le Roux R, Manilall A, Mokotedi LP, Millen AME. The effect of TNF-α inhibitor treatment on microRNAs and endothelial function in collagen induced arthritis. PLoS One 2022; 17:e0264558. [PMID: 35213638 PMCID: PMC8880872 DOI: 10.1371/journal.pone.0264558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 02/11/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic inflammation causes dysregulated expression of microRNAs. Aberrant microRNA expression is associated with endothelial dysfunction. In this study we determined whether TNF-α inhibition impacted the expression of miRNA-146a-5p and miRNA-155-5p, and whether changes in the expression of these miRNAs were related to inflammation-induced changes in endothelial function in collagen-induced arthritis (CIA). Sixty-four Sprague-Dawley rats were divided into control (n = 24), CIA (n = 24) and CIA+etanercept (n = 16) groups. CIA and CIA+etanercept groups were immunized with bovine type-II collagen, emulsified in incomplete Freund’s adjuvant. Upon signs of arthritis, the CIA+etanercept group received 10mg/kg of etanercept intraperitoneally, every three days. After six weeks of treatment, mesenteric artery vascular reactivity was assessed using wire-myography. Serum concentrations of TNF-α, C-reactive protein, interleukin-6, vascular adhesion molecule-1 (VCAM-1) and pentraxin-3 (PTX-3) were measured by ELISA. Relative expression of circulating miRNA-146a-5p and miRNA-155-5p were determined using RT-qPCR. Compared to controls, circulating miRNA-155-5p, VCAM-1 and PTX-3 concentrations were increased, and vessel relaxation was impaired in the CIA (all p<0.05), but not in the CIA+etanercept (all p<0.05) groups. The CIA group had greater miRNA-146a-5p expression compared to the CIA+etanercept group (p = 0.005). Independent of blood pressure, miRNA-146a-5p expression was associated with increased PTX-3 concentrations (p = 0.03), while miRNA-155-5p expression was associated with impaired vessel relaxation (p = 0.01). In conclusion, blocking circulating TNF-α impacted systemic inflammation-induced increased expression of miRNA-146a-5p and miRNA-155-5p, which were associated with endothelial inflammation and impaired endothelial dependent vasorelaxation, respectively.
Collapse
Affiliation(s)
- Sulè Gunter
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- * E-mail:
| | - Frederic S. Michel
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Serena S. Fourie
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mikayra Singh
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Regina le Roux
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ashmeetha Manilall
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lebogang P. Mokotedi
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Aletta M. E. Millen
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
24
|
miR-146a contributes to atherosclerotic plaque stability by regulating the expression of TRAF6 and IRAK-1. Mol Biol Rep 2022; 49:4205-4216. [PMID: 35195809 DOI: 10.1007/s11033-022-07253-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/09/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease. The vulnerable plaque of atherosclerotic can lead to the development of many diseases including acute coronary syndrome and coronary heart disease. It is well known that miR-146a is the key brake miRNA of the inflammatory signal transduction pathway. However, the effect of miR-146a on the stability of atherosclerotic plaque remains to be elucidated. METHODS AND RESULTS We constructed animal models of atherosclerosis and foam cell models, and overexpressed and knocked-down miR-146a in models. After staining with Hematoxylin-Eosin (HE), Oil Red O, immunocytochemistry (IHC) and Sirius Red, we used the proportion of (Lipids area + Macrophage area) and (SMCs area + collagen area) to evaluate atherosclerotic plaque stability. TUNEL and flow cytometry were performed to detect the apoptosis level of macrophages. Levels of inflammatory factors were detected via ELISA assay. The results showed that miR-146a, IRAK1 and TRAF6 were abnormally expressed in plaques of atherosclerotic animals. Overexpression of miR-146a contributed to the stability of plaques that inhibited plaque formation, macrophage apoptosis and levels of pro-inflammatory factors. The Dual-luciferase reporter gene assay, IF and FISH were used to verify the regulatory mechanism of miR-146a on IRAK1 and TRAF6. We found that IRAK1 and TRAF6 promoted lipid uptake, apoptosis, and release of pro-inflammatory factors of RAW264.7 macrophages, whereas miR-146a restored RAW264.7 macrophages phenotype by inhibiting IRAK1 and TRAF6 expression. CONCLUSIONS We display for the first time that miR-146a inhibits the formation of foam cells, RAW264.7 macrophage apoptosis and pro-inflammatory reaction through negative regulation of IRAK1 and TRAF6 expression, thereby enhancing the stability of atherosclerotic plaques.
Collapse
|
25
|
Di Pietro P, Carrizzo A, Sommella E, Oliveti M, Iacoviello L, Di Castelnuovo A, Acernese F, Damato A, De Lucia M, Merciai F, Iesu P, Venturini E, Izzo R, Trimarco V, Ciccarelli M, Giugliano G, Carnevale R, Cammisotto V, Migliarino S, Virtuoso N, Strianese A, Izzo V, Campiglia P, Ciaglia E, Levkau B, Puca AA, Vecchione C. Targeting the ASMase/S1P pathway protects from sortilin-evoked vascular damage in hypertension. J Clin Invest 2022; 132:146343. [PMID: 35104805 PMCID: PMC8803332 DOI: 10.1172/jci146343] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 11/24/2021] [Indexed: 12/24/2022] Open
Abstract
Sortilin has been positively correlated with vascular disorders in humans. No study has yet evaluated the possible direct effect of sortilin on vascular function. We used pharmacological and genetic approaches coupled with study of murine and human samples to unravel the mechanisms recruited by sortilin in the vascular system. Sortilin induced endothelial dysfunction of mesenteric arteries through NADPH oxidase 2 (NOX2) isoform activation, dysfunction that was prevented by knockdown of acid sphingomyelinase (ASMase) or sphingosine kinase 1. In vivo, recombinant sortilin administration induced arterial hypertension in WT mice. In contrast, genetic deletion of sphingosine-1-phosphate receptor 3 (S1P3) and gp91phox/NOX2 resulted in preservation of endothelial function and blood pressure homeostasis after 14 days of systemic sortilin administration. Translating these research findings into the clinical setting, we detected elevated sortilin levels in hypertensive patients with endothelial dysfunction. Furthermore, in a population-based cohort of 270 subjects, we showed increased plasma ASMase activity and increased plasma levels of sortilin, S1P, and soluble NOX2-derived peptide (sNOX2-dp) in hypertensive subjects, and the increase was more pronounced in hypertensive subjects with uncontrolled blood pressure. Our studies reveal what we believe is a previously unrecognized role of sortilin in the impairment of vascular function and in blood pressure homeostasis and suggest the potential of sortilin and its mediators as biomarkers for the prediction of vascular dysfunction and high blood pressure.
Collapse
Affiliation(s)
- Paola Di Pietro
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy.,Department of Vascular Physiopathology, IRCCS Neuromed, Pozzilli, Italy
| | - Eduardo Sommella
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano, Italy
| | - Marco Oliveti
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Licia Iacoviello
- Department of Medicine and Surgery, Research Center in Epidemiology and Preventive Medicine (EPIMED), University of Insubria, Varese, Italy.,Department of Epidemiology and Prevention, IRCCS Neuromed, Pozzilli, Italy
| | | | - Fausto Acernese
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano, Italy
| | - Antonio Damato
- Department of Vascular Physiopathology, IRCCS Neuromed, Pozzilli, Italy
| | | | - Fabrizio Merciai
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano, Italy.,PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, Italy
| | - Paola Iesu
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | | | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Giuseppe Giugliano
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Roberto Carnevale
- Mediterranea Cardiocentro, Naples, Italy.,Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Vittoria Cammisotto
- Department of General Surgery and Surgical Speciality Paride Stefanini, Sapienza University of Rome, Rome, Italy
| | - Serena Migliarino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Nicola Virtuoso
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Andrea Strianese
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Viviana Izzo
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Pietro Campiglia
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano, Italy.,European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Bodo Levkau
- Institute for Molecular Medicine III, Heinrich-Heine-University, Medical Faculty, Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Annibale A Puca
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy.,Ageing Unit, IRCCS MultiMedica, Milan, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy.,Department of Vascular Physiopathology, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
26
|
McLaughlin S, Smyth D, Alarcon EI, Suuronen EJ. Characterization of the Monocyte Response to Biomaterial Therapy for Cardiac Repair. Methods Mol Biol 2022; 2485:279-298. [PMID: 35618913 DOI: 10.1007/978-1-0716-2261-2_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Biomaterials are scaffolds designed to mimic the extracellular matrix and stimulate tissue repair. Biomaterial therapies have shown promise for improving wound healing in cardiac tissue after ischemic injury. An unintentional consequence of biomaterial delivery may be the stimulation of inflammation through recruitment of circulating monocytes into the tissue. Monocytes are a type of leukocyte (white blood cell) that play a critical role in pathogen recognition, phagocytosis of foreign material, and presentation of antigens to initiate an adaptive immune response. An increase in the pro-inflammatory subset of monocytes, marked by Ly6C antigen expression, in response to biomaterials can lead to rapid material degradation, ineffective treatment, and worsening of tissue injury. Flow cytometry is a leading method for screening the recruitment of monocytes to the heart in response to biomaterial injection. Here, we describe the isolation of leukocytes from the heart, blood, and spleen of mice treated with a biomaterial post-myocardial infarction and describe a flow cytometry protocol used to quantify the levels of major leukocyte subtypes, including Ly6C+ inflammatory monocytes.
Collapse
Affiliation(s)
- Sarah McLaughlin
- BioEngineering and Therapeutic Solutions (BEaTS), Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - David Smyth
- Cardiac Function Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Emilio I Alarcon
- BioEngineering and Therapeutic Solutions (BEaTS), Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Erik J Suuronen
- BioEngineering and Therapeutic Solutions (BEaTS), Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
27
|
Abstract
Regulatory RNAs like microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) control vascular and immune cells' phenotype and thus play a crucial role in atherosclerosis. Moreover, the mutual interactions between miRNAs and lncRNAs link both types of regulatory RNAs in a functional network that affects lesion formation. In this review, we deduce novel concepts of atherosclerosis from the analysis of the current data on regulatory RNAs' role in endothelial cells (ECs) and macrophages. In contrast to arterial ECs, which adopt a stable phenotype by adaptation to high shear stress, macrophages are highly plastic and quickly change their activation status. At predilection sites of atherosclerosis, such as arterial bifurcations, ECs are exposed to disturbed laminar flow, which generates a dysadaptive stress response mediated by miRNAs. Whereas the highly abundant miR-126-5p promotes regenerative proliferation of dysadapted ECs, miR-103-3p stimulates inflammatory activation and impairs endothelial regeneration by aberrant proliferation and micronuclei formation. In macrophages, miRNAs are essential in regulating energy and lipid metabolism, which affects inflammatory activation and foam cell formation.Moreover, lipopolysaccharide-induced miR-155 and miR-146 shape inflammatory macrophage activation through their oppositional effects on NF-kB. Most lncRNAs are not conserved between species, except a small group of very long lncRNAs, such as MALAT1, which blocks numerous miRNAs by providing non-functional binding sites. In summary, regulatory RNAs' roles are highly context-dependent, and therapeutic approaches that target specific functional interactions of miRNAs appear promising against cardiovascular diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany.
| | - Saffiyeh Saboor Maleki
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Maliheh Nazari-Jahantigh
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
28
|
Barutta F, Corbetta B, Bellini S, Guarrera S, Matullo G, Scandella M, Schalkwijk C, Stehouwer CD, Chaturvedi N, Soedamah-Muthu SS, Durazzo M, Gruden G. MicroRNA 146a is associated with diabetic complications in type 1 diabetic patients from the EURODIAB PCS. J Transl Med 2021; 19:475. [PMID: 34823560 PMCID: PMC8614036 DOI: 10.1186/s12967-021-03142-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/11/2021] [Indexed: 12/11/2022] Open
Abstract
Background MicroRNA-146a-5p (miR-146a-5p) is a key regulator of inflammatory processes. Expression of miR-146a-5p is altered in target organs of diabetic complications and deficiency of miR-146a-5p has been implicated in their pathogenesis. We investigated if serum miR-146a-5p levels were independently associated with micro/macrovascular complications of type 1 diabetes (DM1). Methods A nested case–control study from the EURODIAB PCS of 447 DM1 patients was performed. Cases (n = 294) had one or more complications of diabetes, whereas controls (n = 153) did not have any complication. Total RNA was isolated from all subjects and miR-146a-5p levels measured by qPCR. Both the endogenous controls U6 snRNA and the spike (Cel-miR-39) were used to normalize the results. Logistic regression analysis was carried out to investigate the association of miR-146a-5p with diabetes complications. Results MiR-146a-5p levels were significantly lower in cases [1.15 (0.32–3.34)] compared to controls [1.74 (0.44–6.74) P = 0.039]. Logistic regression analysis showed that levels of miR-146a-5p in the upper quartile were inversely associated with reduced odds ratio (OR) of all complications (OR 0.34 [95% CI 0.14–0.76]) and particularly with cardiovascular diseases (CVD) (OR 0.31 [95% CI 0.11–0.84]) and diabetic retinopathy (OR 0.40 [95% CI 0.16–0.99]), independently of age, sex, diabetes duration, A1c, hypertension, AER, eGFR, NT-proBNP, and TNF-α. Conclusions In this large cohort of DM1 patients, we reported an inverse and independent association of miR-146a-5p with diabetes chronic complications and in particular with CVD and retinopathy, suggesting that miR-146a-5p may be a novel candidate biomarker of DM1 complications. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03142-4.
Collapse
Affiliation(s)
- Federica Barutta
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy.
| | - Beatrice Corbetta
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| | - Stefania Bellini
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| | - Simonetta Guarrera
- Italian Institute for Genomic Medicine, IIGM, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Giuseppe Matullo
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy.,Medical Genetics Unit, AOU Città Della Salute E Della Scienza, Turin, Italy
| | - Michela Scandella
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| | - Casper Schalkwijk
- Department of Internal Medicine and Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Coen D Stehouwer
- Department of Internal Medicine and Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Nish Chaturvedi
- Institute of Cardiovascular Science, University College London, London, UK
| | - Sabita S Soedamah-Muthu
- Center of Research On Psychology in Somatic Diseases (CORPS), Department of Medical and Clinical Psychology, Tilburg University, Tilburg, The Netherlands.,Institute for Food, Nutrition and Health, University of Reading, Reading, UK
| | - Marilena Durazzo
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| | - Gabriella Gruden
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| |
Collapse
|
29
|
Liu H, Wang H, Ma J, Qiao Z, Zhang L, Ge G. MicroRNA-146a-3p/HDAC1/KLF5/IKBα signal axis modulates plaque formation of atherosclerosis mice. Life Sci 2021; 284:119615. [PMID: 34004248 DOI: 10.1016/j.lfs.2021.119615] [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: 09/06/2020] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Atherosclerosis (AS) is a multifocal, smoldering immune inflammatory disease of medium and large arteries driven by lipids. The aim of this study is to discuss the mechanism of microRNA-146a-3p (miR-146a-3p)/histone deacetylase 1 (HDAC1)/Krüppel-like factor 5 (KLF5)/inhibitors of kappa B α (IKBα) signal axis in plaque formation of AS mice. METHODS ApoE-/- mice were fed with high-fat feed for 12 weeks to establish AS mice model. The expression of miR-146a-3p, KLF5, HDAC1 and IKBα in aortic wall tissues of AS mice was tested. The targeting relationship between miR-146a-3p and HDAC1 was verified. AS mice were injected with miR-146a-3p antagomir or HDAC1 overexpression to verify the impacts of miR-146a-3p and HDAC1 on blood lipids and inflammatory factors in serum, aortic wall apoptotic cells, antioxidant stress capacity and the plaque area in AS mice. VECs proliferation and apoptosis were also measured in vitro. RESULTS miR-146a-3p and KLF5 were increased while HDAC1 and IKBα were reduced in aortic wall tissues of AS mice. miR-146a-3p directly targeted to HDAC1. Depletion of miR-146a-3p or restoration of HDAC1 was correlated to lower plasma lipid level, reduced inflammatory factors in serum, attenuated aortic wall apoptosis, increased antioxidant stress capacity and improved the stability of pathological plaque of AS mice. miR-146a-3p down-regulation or HDAC1 up-regulation promoted VECs proliferation and inhibited apoptosis. CONCLUSION Functional studies show that depleted miR-146a-3p advances HDAC1 and IKBα expression as well as inhibits KLF5 expression to facilitate the stability of pathological plaques in AS mice.
Collapse
Affiliation(s)
- Huajin Liu
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China.
| | - Hongwei Wang
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| | - Jiangwei Ma
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| | - Zengyong Qiao
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| | - Li Zhang
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| | - Guanghao Ge
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| |
Collapse
|
30
|
Liu F, Wang S, Luo Z. Associations of the miRNA-146a rs2910164 and the miRNA-499a rs3746444 Polymorphisms With Plasma Lipid Levels: A Meta-Analysis. Front Genet 2021; 12:746686. [PMID: 34646311 PMCID: PMC8503190 DOI: 10.3389/fgene.2021.746686] [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: 07/24/2021] [Accepted: 08/24/2021] [Indexed: 12/02/2022] Open
Abstract
Background: The studies of miRNAs are vibrant and remain at the forefront in the cardiovascular system. Emerging studies indicate that the genetic polymorphisms of the miRNA gene may affect lipid metabolism; this study aims to clarify the specific correlations between the rs2910164 and rs3746444 polymorphisms and lipid levels. Methods and Results: A comprehensive search of literature was performed from December 31, 2020, to May 31, 2021, by searching of the PubMed and the Cochrane databases. The standardized mean difference (SMD) and 95% confidence interval (CI) were used to evaluate the differences in lipid levels between the genotypes. rs2910164, a functional polymorphism in the miRNA-146a gene, was associated with increased triglycerides (TG) (SMD = 0.35, 95% CI = 0.15-0.54, p < 0.001), total cholesterol (TC) (SMD = 0.43, 95% CI = 0.16-0.70, p < 0.001), and low-density lipoprotein cholesterol (LDL-C) (SMD = 0.37, 95% CI = 0.11-0.63, p = 0.01) as well as decreased high-density lipoprotein cholesterol (HDL-C) (SMD = -0.27, 95% CI = -0.47-0.07, p = 0.01) levels. rs3746444, a functional polymorphism in the miRNA-499a gene, was only correlated with decreased TG (SMD = -0.09, 95% CI = -0.17-0.01, P = 0.03) levels. Conclusions: The miRNA-146a rs2910164 polymorphism is significantly associated with atherogenic dyslipidemia.
Collapse
Affiliation(s)
- Fuqiang Liu
- Department of Cardiology, First People's Hospital of Chengdu, Chengdu, China
| | - Shengping Wang
- Department of Cardiology, First People's Hospital of Chengdu, Chengdu, China
| | - Zhi Luo
- Department of Internal Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
31
|
Tonyan ZN, Nasykhova YA, Danilova MM, Glotov AS. Genetics of macrovascular complications in type 2 diabetes. World J Diabetes 2021; 12:1200-1219. [PMID: 34512887 PMCID: PMC8394234 DOI: 10.4239/wjd.v12.i8.1200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/04/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder that currently affects more than 400 million worldwide and is projected to cause 552 million cases by the year 2030. Long-term vascular complications, such as coronary artery disease, myocardial infarction, stroke, are the leading causes of morbidity and mortality among diabetic patients. The recent advances in genome-wide technologies have given a powerful impetus to the study of risk markers for multifactorial diseases. To date, the role of genetic and epigenetic factors in modulating susceptibility to T2DM and its vascular complications is being successfully studied that provides the accumulation of genomic knowledge. In the future, this will provide an opportunity to reveal the pathogenetic pathways in the development of the disease and allow to predict the macrovascular complications in T2DM patients. This review is focused on the evidence of the role of genetic variants and epigenetic changes in the development of macrovascular pathology in diabetic patients.
Collapse
Affiliation(s)
- Ziravard N Tonyan
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Saint-Petersburg 199034, Russia
| | - Yulia A Nasykhova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Saint-Petersburg 199034, Russia
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, Saint-Petersburg 199034, Russia
| | - Maria M Danilova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Saint-Petersburg 199034, Russia
| | - Andrey S Glotov
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Saint-Petersburg 199034, Russia
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, Saint-Petersburg 199034, Russia
| |
Collapse
|
32
|
Shi Z, Zheng Z, Lin X, Ma H. Long Noncoding RNA MALAT1 Regulates the Progression of Atherosclerosis by miR-330-5p/NF-κB Signal Pathway. J Cardiovasc Pharmacol 2021; 78:235-246. [PMID: 34554676 DOI: 10.1097/fjc.0000000000001061] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/28/2021] [Indexed: 01/04/2023]
Abstract
ABSTRACT Long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was reported to be related to atherosclerosis (AS) progression. However, the underlying mechanism of MALAT1 in AS remains unknown. Quantitative real-time polymerase chain reaction was performed to detect the expression of MALAT1 and miR-330-5p. Western blot was applied to assess the protein levels of cluster of differentiation 36, interleukin-1β, interleukin-6 and tumor necrosis factor-α, phosphorylation of nuclear factor kappa-B inhibitor alpha and phosphorylation of p65. Flow cytometry assay, cell counting kit 8 assay, triglyceride, and total cholesterol detection assays were used to detect the apoptosis, viability, and lipid indexes of THP-1 macrophages-derived foam cells. Online database starbasev2.0 was used to predict the binding sequences between MALAT1 and miR-330-5p and it was verified by dual-luciferase reporter system and RNA immunoprecipitation assay. Besides, an AS mice model was used to evaluate the effect of MALAT1 in vivo. As a result, MALAT1 was overexpressed, whereas miR-330-5p was downregulated in THP-1 macrophages-derived foam cells. MiR-330-5p was a target of MALAT1. MALAT1 depletion inhibited cell formation, apoptosis, and inflammation in THP-1 macrophages-derived foam cells. Besides, MALAT1 overexpression promoted the inflammation in AS mice model, which promoted the pathogenesis of AS. Furthermore, miR-330-5p regulated the nuclear factor kappa light chain enhancer of activated B cells (NF-κB) pathway in THP-1 macrophages-derived foam cells. Moreover, MALAT1 regulated NF-κB signal pathway to mediate the pathogenesis of AS by sponging miR-330-5p. MALAT1 sponges miR-330-5p to activate NF-κB signal pathway in THP-1 macrophages-derived foam cells. This finding may provide a novel biomarker for AS diagnosis.
Collapse
Affiliation(s)
- Zhifeng Shi
- Department of Neurology, MinDong Hospital of Ningde City, Fuan City, Fujian Province, China
| | | | | | | |
Collapse
|
33
|
Zhu Q, Zhao C, Wang Y, Li X, Xue Y, Ma C. LncRNA NEAT1 Promote Inflammatory Responses in Coronary Slow Flow Through Regulating miR-148b-3p/ICAM-1 Axis. J Inflamm Res 2021; 14:2445-2463. [PMID: 34135616 PMCID: PMC8200141 DOI: 10.2147/jir.s312583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/13/2021] [Indexed: 12/25/2022] Open
Abstract
Background Coronary slow flow (CSF) is an angiographic phenomenon characterized by delayed coronary opacification with normal or near-normal epicardial coronary arteries. The pathogenesis of CSF is closely related to inflammatory response. Accumulating evidence shows that long non-coding RNAs (lncRNAs) play an important role in cardiovascular disease. However, the mechanism underlying the influence of the lncRNA nuclear enriched abundant transcripts 1 (NEAT1) on CSF is still unknown. Patients and Methods Forty CSF patients and forty control subjects were included in the study and underwent coronary angiography, Seattle angina questionnaire (SAQ) and echocardiography. The plasma levels of the inflammatory factors soluble intercellular adhesion molecule-1 (sICAM-1), interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) were determined by ELISA. The expression levels of NEAT1, miR-148b-3p and ICAM-1 in cells were measured by qRT-PCR or Western blotting. Cell proliferation was measured by 5‐Ethynyl‐2ʹ‐deoxyuridine (EdU) and Cell Counting Kit-8 (CCK-8) assay. Cell apoptosis was detected by apoptosis assay. The relationship between NEAT1 and miR-148b-3p was verified by luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay and avidin-biotin pull-down assay. The relationship between ICAM-1 and miR-148b-3p was verified by luciferase reporter gene assay and avidin-biotin pull-down assay. Results This study showed that plasma sICAM-1, miR-148b-3p, and NEAT1 as independent predictors of a CSF diagnosis. Furthermore, plasma NEAT1 level showed superior diagnostic ability for CSF compared with sICAM-1 and miR-148b-3p. It was also shown that high expression of NEAT1 in oxygen-glucose deprivation (OGD)-treated human umbilical vein endothelial cells (HUVECs) functions as a competing endogenous RNA (ceRNA). By specifically binding miR-148b-3p, it weakened the negative regulatory effects of miR-148b-3p on the ICAM-1 target gene leading to upregulated expression of ICAM-1. This interaction was also shown to inhibit HUVEC proliferation and enhance apoptosis. Conclusion This study demonstrated for the first time the important mechanism of action of the NEAT1/miR-148b-3p/ICAM-1 axis in the progression of CSF disease, and indicated the potential of NEAT1, miR-148b-3p, and ICAM-1 as a new target for the diagnosis and treatment of CSF.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Cuiting Zhao
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yonghuai Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xinxin Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, People's Republic of China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
34
|
Deng X, Zuo M, Pei Z, Xie Y, Yang Z, Zhang Z, Jiang M, Kuang D. MicroRNA-455-5p Contributes to Cholangiocarcinoma Growth and Mediates Galangin's Anti-Tumor Effects. J Cancer 2021; 12:4710-4721. [PMID: 34149934 PMCID: PMC8210562 DOI: 10.7150/jca.58873] [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: 01/31/2021] [Accepted: 05/22/2021] [Indexed: 01/06/2023] Open
Abstract
Fully understanding the mechanism of how Cholangiocarcinoma (CCA) development and discovering promising therapeutic drugs are important to improve patients' survival time. This study identifies that microRNA-455-5p (miR-455-5p) targets protein phosphatase 1 regulatory subunit 12A (PPP1R12A), an effect that represses mitogen-activated protein kinase (MAPK) and PI3K/AKT pathway activation, thereby controlling CCA cells survival and metastasis. Moreover, miR-455-5p expression is reduced in CCA tissues and negative correlation with PPP1R12A and PPP1R12A knockdown phenotypic mimics miR-455-5p' effects on CCA cells. Furthermore, we demonstrate that galangin inhibits CCA growth both in vitro and in vivo, which is associated with increased miR-455-5p and repressed PPP1R12A expression. In support, overexpression of miR-455-5p abrogates those galangin-mediated anti-CCA effects. These findings establish an essential role of miR-455-5p in CCA development and galangin may provide a potential therapeutic adjuvant agent for anti-CCA treatment.
Collapse
Affiliation(s)
- Xu Deng
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Meiling Zuo
- Department of Pharmacy, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zhifang Pei
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuanlin Xie
- Department of Pharmacy, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zhongbao Yang
- Department of Pharmacy, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zhihui Zhang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Minna Jiang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dabin Kuang
- Department of Pharmacy, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
35
|
Sivasubramaniyam T, Yang J, Cheng HS, Zyla A, Li A, Besla R, Dotan I, Revelo XS, Shi SY, Le H, Schroer SA, Dodington DW, Park YJ, Kim MJ, Febbraro D, Ruel I, Genest J, Kim RH, Mak TW, Winer DA, Robbins CS, Woo M. Dj1 deficiency protects against atherosclerosis with anti-inflammatory response in macrophages. Sci Rep 2021; 11:4723. [PMID: 33633277 PMCID: PMC7907332 DOI: 10.1038/s41598-021-84063-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/08/2021] [Indexed: 11/09/2022] Open
Abstract
Inflammation is a key contributor to atherosclerosis with macrophages playing a pivotal role through the induction of oxidative stress and cytokine/chemokine secretion. DJ1, an anti-oxidant protein, has shown to paradoxically protect against chronic and acute inflammation. However, the role of DJ1 in atherosclerosis remains elusive. To assess the role of Dj1 in atherogenesis, we generated whole-body Dj1-deficient atherosclerosis-prone Apoe null mice (Dj1-/-Apoe-/-). After 21 weeks of atherogenic diet, Dj1-/- Apoe-/-mice were protected against atherosclerosis with significantly reduced plaque macrophage content. To assess whether haematopoietic or parenchymal Dj1 contributed to atheroprotection in Dj1-deficient mice, we performed bone-marrow (BM) transplantation and show that Dj1-deficient BM contributed to their attenuation in atherosclerosis. To assess cell-autonomous role of macrophage Dj1 in atheroprotection, BM-derived macrophages from Dj1-deficient mice and Dj1-silenced macrophages were assessed in response to oxidized low-density lipoprotein (oxLDL). In both cases, there was an enhanced anti-inflammatory response which may have contributed to atheroprotection in Dj1-deficient mice. There was also an increased trend of plasma DJ-1 levels from individuals with ischemic heart disease compared to those without. Our findings indicate an atheropromoting role of Dj1 and suggests that targeting Dj1 may provide a novel therapeutic avenue for atherosclerosis treatment or prevention.
Collapse
Affiliation(s)
- Tharini Sivasubramaniyam
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Jiaqi Yang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Henry S Cheng
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Alexandra Zyla
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Angela Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Rickvinder Besla
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Idit Dotan
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Xavier S Revelo
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Sally Yu Shi
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Helen Le
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Stephanie A Schroer
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - David W Dodington
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Yoo Jin Park
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Min Jeong Kim
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Korea
| | - Daniella Febbraro
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Isabelle Ruel
- Research Institute of the McGill University Health Centre, Royal Victoria Hospital, Montreal, QC, H4A 3J1, Canada
| | - Jacques Genest
- Research Institute of the McGill University Health Centre, Royal Victoria Hospital, Montreal, QC, H4A 3J1, Canada.,Department of Medicine, McGill University, Royal Victoria Hospital, Montreal, QC, H4A 3J1, Canada
| | - Raymond H Kim
- Department of Medicine, University Health Network/Sinai Health System, University of Toronto, Toronto, ON, M5G 2C4, Canada
| | - Tak W Mak
- Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Daniel A Winer
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 2M9, Canada.,Department of Pathology, University Health Network, Toronto, M5G 2C4, Canada
| | - Clinton S Robbins
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, M5G 2M9, Canada. .,Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada. .,Department of Medicine, University Health Network/Sinai Health System, University of Toronto, Toronto, ON, M5G 2C4, Canada. .,Division of Endocrinology and Metabolism, University Health Network/Sinai Health System, University of Toronto, Toronto, ON, M5G 2C4, Canada. .,MaRS Centre, Toronto Medical Discovery Tower, 101 College Street, 10th floor, Room 10-361, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
36
|
Águila S, de los Reyes-García AM, Fernández-Pérez MP, Reguilón-Gallego L, Zapata-Martínez L, Ruiz-Lorente I, Vicente V, González-Conejero R, Martínez C. MicroRNAs as New Regulators of Neutrophil Extracellular Trap Formation. Int J Mol Sci 2021; 22:ijms22042116. [PMID: 33672737 PMCID: PMC7924615 DOI: 10.3390/ijms22042116] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/27/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are formed after neutrophils expelled their chromatin content in order to primarily capture and eliminate pathogens. However, given their characteristics due in part to DNA and different granular proteins, NETs may induce a procoagulant response linking inflammation and thrombosis. Unraveling NET formation molecular mechanisms as well as the intracellular elements that regulate them is relevant not only for basic knowledge but also to design diagnostic and therapeutic tools that may prevent their deleterious effects observed in several inflammatory pathologies (e.g., cardiovascular and autoimmune diseases, cancer). Among the potential elements involved in NET formation, several studies have investigated the role of microRNAs (miRNAs) as important regulators of this process. miRNAs are small non-coding RNAs that have been involved in the control of almost all physiological processes in animals and plants and that are associated with the development of several pathologies. In this review, we give an overview of the actual knowledge on NETs and their implication in pathology with a special focus in cardiovascular diseases. We also give a brief overview on miRNA biology to later focus on the different miRNAs implicated in NET formation and the perspectives opened by the presented data.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rocío González-Conejero
- Correspondence: (R.G.-C.); (C.M.); Tel.: +34-968341990 (R.G.-C. & C.M.); Fax: +34-968261914 (R.G.-C. & C.M.)
| | - Constantino Martínez
- Correspondence: (R.G.-C.); (C.M.); Tel.: +34-968341990 (R.G.-C. & C.M.); Fax: +34-968261914 (R.G.-C. & C.M.)
| |
Collapse
|
37
|
Arroyo AB, Águila S, Fernández-Pérez MP, Reyes-García AMDL, Reguilón-Gallego L, Zapata-Martínez L, Vicente V, Martínez C, González-Conejero R. miR-146a in Cardiovascular Diseases and Sepsis: An Additional Burden in the Inflammatory Balance? Thromb Haemost 2020; 121:1138-1150. [PMID: 33352593 DOI: 10.1055/a-1342-3648] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The new concept of thrombosis associated with an inflammatory process is called thromboinflammation. Indeed, both thrombosis and inflammation interplay one with the other in a feed forward manner amplifying the whole process. This pathological reaction in response to a wide variety of sterile or non-sterile stimuli eventually causes acute organ damage. In this context, neutrophils, mainly involved in eliminating pathogens as an early barrier to infection, form neutrophil extracellular traps (NETs) that are antimicrobial structures responsible of deleterious side effects such as thrombotic complications. Although NETosis mechanisms are being unraveled, there are still many regulatory elements that have to be discovered. Micro-ribonucleic acids (miRNAs) are important modulators of gene expression implicated in human pathophysiology almost two decades ago. Among the different miRNAs implicated in inflammation, miR-146a is of special interest because: (1) it regulates among others, Toll-like receptors/nuclear factor-κB axis which is of paramount importance in inflammatory processes, (2) it regulates the formation of NETs by modifying their aging phenotype, and (3) it has expression levels that may decrease among individuals up to 50%, controlled in part by the presence of several polymorphisms. In this article, we will review the main characteristics of miR-146a biology. In addition, we will detail how miR-146a is implicated in the development of two paradigmatic diseases in which thrombosis and inflammation interact, cardiovascular diseases and sepsis, and their association with the presence of miR-146a polymorphisms and the use of miR-146a as a marker of cardiovascular diseases and sepsis.
Collapse
Affiliation(s)
- Ana B Arroyo
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| | - Sonia Águila
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| | - María P Fernández-Pérez
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| | - Ascensión M de Los Reyes-García
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| | - Laura Reguilón-Gallego
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| | - Laura Zapata-Martínez
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| | - Vicente Vicente
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| | - Constantino Martínez
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| | - Rocío González-Conejero
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Centro Regional de Hemodonación, Universidad de Murcia, IMIB, Murcia, Spain
| |
Collapse
|
38
|
An association study between MiR-146a and INSR gene polymorphisms and hypertensive disorders of pregnancy in Northeastern Han Chinese population. Placenta 2020; 104:94-101. [PMID: 33310299 DOI: 10.1016/j.placenta.2020.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/22/2020] [Accepted: 11/27/2020] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Hypertensive disorders of pregnancy(HDP) is a complex and challenging group of pregnancy complications that is one of the leading causes of maternal and fetal death worldwide. Recent studies have shown that the single nucleotide polymorphism(SNP) may play a role in the pathogenesis of HDP. This study aimed to investigate the association of MiR-146a rs2910164 and insulin receptor(INSR) rs2059806 SNPs with HDP and their associated complications in the Han population of Northeast China. METHODS A total of 240 HDP patients and 380 healthy controls were selected for genotype determination. For the most special and high incidence of HDP, we also studied the SNPs in association with pre-eclampsia(PE) patients. In addition, HDP complicated with gestational diabetes mellitus(GDM) patients was further analyzed to identify the association between SNPs and HDP-related complications. Multivariate logical regression analysis combined with 10, 000 permutation test corrections was used to analyze the association of MiR-146a and INSR SNPs with HDP. RESULTS After adjusting for relevant factors, MiR-146a rs2910164 or INSR rs2059806 SNPs were not significantly different between HDP or PE patients and healthy controls(P>0.05). Meanwhile, MiR146a rs2910164 and INSR rs2059806 SNPs were not significantly different between HDP complicated with GDM and control group. DISCUSSION Our data indicates that MiR-146a rs2910164 and INSR rs2059806 SNPs may not be significantly related with HDP in the Han population of Northeast China living in Heilongjiang Province.
Collapse
|
39
|
Guo W, Li XN, Li J, Lu J, Wu J, Zhu WF, Qin P, Xu NZ, Zhang Q. Increased plasma miR-146a levels are associated with subclinical atherosclerosis in newly diagnosed type 2 diabetes mellitus. J Diabetes Complications 2020; 34:107725. [PMID: 32981813 DOI: 10.1016/j.jdiacomp.2020.107725] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/02/2020] [Accepted: 08/23/2020] [Indexed: 11/29/2022]
Abstract
AIMS The association between circulating miR-146a and subclinical atherosclerosis in type 2 diabetes mellitus (T2DM) remains poorly understood. This study aimed to investigate the correlation between plasma miR-146a levels and subclinical atherosclerosis as measured by the carotid intima-media thickness (CIMT) and brachial-ankle pulse wave velocity (baPWV) in patients with newly diagnosed T2DM. METHODS We studied 100 patients with newly diagnosed T2DM. Subclinical atherosclerosis was defined as a thickened CIMT (≥1.0 mm) and high baPWV defined as a value greater than the 75th percentile. Plasma miR-146a levels and metabolic parameters were measured. RESULTS Patients with thickened CIMT had higher plasma miR-146a levels than those without thickened CIMT (3.36 ± 1.32 vs 1.38 ± 1.11, P < 0.001). Patients in the high baPWV group had higher plasma miR-146a levels than those in the normal baPWV group (3.43 ± 1.32 vs 1.98 ± 1.48, P < 0.001). Both CIMT (β = 0.569, P < 0.001) and baPWV (β = 0.274, P = 0.001) positively correlated with plasma miR-146a levels after adjustment for confounding factors by multiple stepwise regression. On binary logistic regression, plasma miR-146a level was an independent risk factor for thickened CIMT (OR = 3.890, 95% CI 1.415-7.698, P = 0.008) and high baPWV (OR = 1.954, 95% CI 1.256-3.040, P = 0.002) after adjustment for established cardiovascular risk factors. The area under the receiver operating characteristics curve (AUROC) of plasma miR-146a level for predicting thickened CIMT was 0.795 (95%CI 0.708-0.883, P < 0.001) and for predicting high baPWV was 0.773 (95%CI 0.679-0.867, P < 0.001). CONCLUSION Plasma miR-146a levels correlate with CIMT and baPWV and could act as a biomarker for early diagnosis and as a therapeutic target for atherosclerosis in T2DM.
Collapse
Affiliation(s)
- Wen Guo
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Na Li
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jie Li
- Department of Endocrinology, Nanjing Central Hospital, Nanjing 210018, China
| | - Jing Lu
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Juan Wu
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wen-Fang Zhu
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Pei Qin
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| | - Nian-Zhen Xu
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Qun Zhang
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
40
|
Karunakaran D, Nguyen MA, Geoffrion M, Vreeken D, Lister Z, Cheng HS, Otte N, Essebier P, Wyatt H, Kandiah JW, Jung R, Alenghat FJ, Mompeon A, Lee R, Pan C, Gordon E, Rasheed A, Lusis AJ, Liu P, Matic LP, Hedin U, Fish JE, Guo L, Kolodgie F, Virmani R, van Gils JM, Rayner KJ. RIPK1 Expression Associates With Inflammation in Early Atherosclerosis in Humans and Can Be Therapeutically Silenced to Reduce NF-κB Activation and Atherogenesis in Mice. Circulation 2020; 143:163-177. [PMID: 33222501 DOI: 10.1161/circulationaha.118.038379] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic activation of the innate immune system drives inflammation and contributes directly to atherosclerosis. We previously showed that macrophages in the atherogenic plaque undergo RIPK3 (receptor-interacting serine/threonine-protein kinase 3)-MLKL (mixed lineage kinase domain-like protein)-dependent programmed necroptosis in response to sterile ligands such as oxidized low-density lipoprotein and damage-associated molecular patterns and that necroptosis is active in advanced atherosclerotic plaques. Upstream of the RIPK3-MLKL necroptotic machinery lies RIPK1 (receptor-interacting serine/threonine-protein kinase 1), which acts as a master switch that controls whether the cell undergoes NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells)-dependent inflammation, caspase-dependent apoptosis, or necroptosis in response to extracellular stimuli. We therefore set out to investigate the role of RIPK1 in the development of atherosclerosis, which is driven largely by NF-κB-dependent inflammation at early stages. We hypothesize that, unlike RIPK3 and MLKL, RIPK1 primarily drives NF-κB-dependent inflammation in early atherogenic lesions, and knocking down RIPK1 will reduce inflammatory cell activation and protect against the progression of atherosclerosis. METHODS We examined expression of RIPK1 protein and mRNA in both human and mouse atherosclerotic lesions, and used loss-of-function approaches in vitro in macrophages and endothelial cells to measure inflammatory responses. We administered weekly injections of RIPK1 antisense oligonucleotides to Apoe-/- mice fed a cholesterol-rich (Western) diet for 8 weeks. RESULTS We find that RIPK1 expression is abundant in early-stage atherosclerotic lesions in both humans and mice. Treatment with RIPK1 antisense oligonucleotides led to a reduction in aortic sinus and en face lesion areas (47.2% or 58.8% decrease relative to control, P<0.01) and plasma inflammatory cytokines (IL-1α [interleukin 1α], IL-17A [interleukin 17A], P<0.05) in comparison with controls. RIPK1 knockdown in macrophages decreased inflammatory genes (NF-κB, TNFα [tumor necrosis factor α], IL-1α) and in vivo lipopolysaccharide- and atherogenic diet-induced NF-κB activation. In endothelial cells, knockdown of RIPK1 prevented NF-κB translocation to the nucleus in response to TNFα, where accordingly there was a reduction in gene expression of IL1B, E-selectin, and monocyte attachment. CONCLUSIONS We identify RIPK1 as a central driver of inflammation in atherosclerosis by its ability to activate the NF-κB pathway and promote inflammatory cytokine release. Given the high levels of RIPK1 expression in human atherosclerotic lesions, our study suggests RIPK1 as a future therapeutic target to reduce residual inflammation in patients at high risk of coronary artery disease.
Collapse
Affiliation(s)
- Denuja Karunakaran
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Dianne Vreeken
- Leiden University Medical Center, The Netherlands (D.V., J.M.v.G.)
| | - Zachary Lister
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Henry S Cheng
- Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C.)
| | - Nicola Otte
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Patricia Essebier
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Hailey Wyatt
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Joshua W Kandiah
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Richard Jung
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Francis J Alenghat
- Cardiology, Department of Medicine, University of Chicago, IL (F.J.A., J.E.F.)
| | - Ana Mompeon
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Richard Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, CA (R.L.)
| | - Calvin Pan
- David Geffen School of Medicine, University of California Los Angeles (C.P., A.J.L.)
| | - Emma Gordon
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Adil Rasheed
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Aldons J Lusis
- David Geffen School of Medicine, University of California Los Angeles (C.P., A.J.L.)
| | - Peter Liu
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Ljubica Perisic Matic
- Vascular Surgery Division, Department of Molecular Medicine and Surgery, Karolinska Institute, Sweden (L.P.M.)
| | | | - Jason E Fish
- Cardiology, Department of Medicine, University of Chicago, IL (F.J.A., J.E.F.)
| | - Liang Guo
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | - Frank Kolodgie
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | - Renu Virmani
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | | | - Katey J Rayner
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.)
| |
Collapse
|
41
|
Fernández-García V, González-Ramos S, Martín-Sanz P, Castrillo A, Boscá L. Contribution of Extramedullary Hematopoiesis to Atherosclerosis. The Spleen as a Neglected Hub of Inflammatory Cells. Front Immunol 2020; 11:586527. [PMID: 33193412 PMCID: PMC7649205 DOI: 10.3389/fimmu.2020.586527] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/06/2020] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) incidence is becoming higher. This fact is promoted by metabolic disorders such as obesity, and aging. Atherosclerosis is the underlying cause of most of these pathologies. It is a chronic inflammatory disease that begins with the progressive accumulation of lipids and fibrotic materials in the blood-vessel wall, which leads to massive leukocyte recruitment. Rupture of the fibrous cap of the atherogenic cusps is responsible for tissue ischemic events, among them myocardial infarction. Extramedullary hematopoiesis (EMH), or blood cell production outside the bone marrow (BM), occurs when the normal production of these cells is impaired (chronic hematological and genetic disorders, leukemia, etc.) or is altered by metabolic disorders, such as hypercholesterolemia, or after myocardial infarction. Recent studies indicate that the main EMH tissues (spleen, liver, adipose and lymph nodes) complement the hematopoietic function of the BM, producing circulating inflammatory cells that infiltrate into the atheroma. Indeed, the spleen, which is a secondary lymphopoietic organ with high metabolic activity, contains a reservoir of myeloid progenitors and monocytes, constituting an important source of inflammatory cells to the atherosclerotic lesion. Furthermore, the spleen also plays an important role in lipid homeostasis and immune-cell selection. Interestingly, clinical evidence from splenectomized subjects shows that they are more susceptible to developing pathologies, such as dyslipidemia and atherosclerosis due to the loss of immune selection. Although CVDs represent the leading cause of death worldwide, the mechanisms involving the spleen-atherosclerosis-heart axis cross-talk remain poorly characterized.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Unidad de Biomedicina, (Unidad Asociada al CSIC), Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Universidad de Las Palmas, Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Grupo de Investigación Medio Ambiente y Salud, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Unidad de Biomedicina, (Unidad Asociada al CSIC), Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Universidad de Las Palmas, Gran Canaria, Spain
| |
Collapse
|
42
|
Sun D, Xiang G, Wang J, Li Y, Mei S, Ding H, Yan J. miRNA 146b-5p protects against atherosclerosis by inhibiting vascular smooth muscle cell proliferation and migration. Epigenomics 2020; 12:2189-2204. [PMID: 33084403 DOI: 10.2217/epi-2020-0155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim: To explore the potentially important role of miRNA 146b-5p (miR-146b) during the development of atherosclerosis. Materials & methods: Proliferation, migration and luciferase assays and mouse models were used to determine the functions of miR-146b. Results: miR-146b was identified as substantially upregulated in the aortic plaques of ApoE-/- mice as well as in response to inflammatory cytokines. Overexpression of miR-146b repressed proliferation and migration of vascular smooth muscle cells by downregulating Bag1 and Mmp16, respectively. Adeno-associated virus-mediated miR-146b overexpression inhibited neointima formation after carotid injury and suppressed atherosclerotic plaque formation in western diet-induced ApoE-/- mice. Conclusion: miR-146b is a novel regulator of vascular smooth muscle cell function induced by inflammatory response, specifically in neointima formation, and offers a novel therapeutic strategy for treating atherosclerosis.
Collapse
Affiliation(s)
- Dating Sun
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, PR China
| | - Gui Xiang
- Department of Physiology & Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, PR China
| | - Jing Wang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, PR China
| | - Yuanyuan Li
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, PR China
| | - Shuai Mei
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, PR China
| | - Hu Ding
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, PR China
| | - Jiangtao Yan
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, PR China
| |
Collapse
|
43
|
Inhibition of microRNA-146a attenuated heart failure in myocardial infarction rats. Biosci Rep 2020; 39:221324. [PMID: 31763669 PMCID: PMC6928527 DOI: 10.1042/bsr20191732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/15/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to determine the roles of microRNA (miR)-146a on myocardial infarction (MI)-induced heart failure and cardiac remodeling. Experiments were carried out in Sprague-Dawley rats treated with ligation of left coronary artery to induce heart failure, and in primary neonatal rat cardiac fibroblasts (CFs) and cardiomyocytes treated with angiotensin (Ang) II. Four weeks after MI, rats were injected with miR-146a antagomiR or agomiR via tail vein. After 2 weeks of injection, the rats were killed. In MI rats, left ventricle (LV) ejection fraction and fractional shortening were reduced, and LV volumes in diastole and systole were increased, which were reversed by miR-146a antagomiR, and further exacerbated after miR-146a agomiR treatment. Administration of miR-146a antagomiR improved the decreases of LV ±dp/dtmax and LV systolic pressure (LVSP), and the increase in LV end-diastolic pressure (LVEDP) of MI rats, but miR-146a agomiR deteriorated the LV ±dp/dtmax, LVSP and LVEDP. The increases in the levels of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), collagen I and collagen III in the heart, and ST2 and norepinephrine in the serum of MI rats were inhibited by miR-146a antagomiR, but aggravated after miR-146a agomiR treatment. The increases of collagen I and collagen III levels induced by Ang II in CFs, and the increases of ANP and BNP levels induced by Ang II in cardiomyocytes were inhibited by miR-146a antagomiR, but aggravated by miR-146a agomiR. These results demonstrated that inhibition of miR-146a improved cardiac dysfunction and cardiac remodeling in heart failure rats.
Collapse
|
44
|
Liao Y, Li H, Cao H, Dong Y, Gao L, Liu Z, Ge J, Zhu H. Therapeutic silencing miR-146b-5p improves cardiac remodeling in a porcine model of myocardial infarction by modulating the wound reparative phenotype. Protein Cell 2020; 12:194-212. [PMID: 32845445 PMCID: PMC7895884 DOI: 10.1007/s13238-020-00750-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/29/2020] [Indexed: 12/18/2022] Open
Abstract
Fibrotic remodeling is an adverse consequence of immune response-driven phenotypic modulation of cardiac cells following myocardial infarction (MI). MicroRNA-146b (miR-146b) is an active regulator of immunomodulation, but its function in the cardiac inflammatory cascade and its clinical implication in fibrotic remodeling following MI remain largely unknown. Herein, miR-146b-5p was found to be upregulated in the infarcted myocardium of mice and the serum of myocardial ischemia patients. Gain- and loss-of-function experiments demonstrated that miR-146b-5p was a hypoxia-induced regulator that governed the pro-fibrotic phenotype transition of cardiac cells. Overexpression of miR-146b-5p activated fibroblast proliferation, migration, and fibroblast-to-myofibroblast transition, impaired endothelial cell function and stress survival, and disturbed macrophage paracrine signaling. Interestingly, the opposite effects were observed when miR-146b-5p expression was inhibited. Luciferase assays and rescue studies demonstrated that the miR-146b-5p target genes mediating the above phenotypic modulations included interleukin 1 receptor associated kinase 1 (IRAK1) and carcinoembryonic antigen related cell adhesion molecule 1 (CEACAM1). Local delivery of a miR-146b-5p antagomir significantly reduced fibrosis and cell death, and upregulated capillary and reparative macrophages in the infarcted myocardium to restore cardiac remodeling and function in both mouse and porcine MI models. Local inhibition of miR-146b-5p may represent a novel therapeutic approach to treat cardiac fibrotic remodeling and dysfunction following MI.
Collapse
Affiliation(s)
- Yiteng Liao
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hao Li
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hao Cao
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yun Dong
- Department of Ultrasound in Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Lei Gao
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China. .,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
| | - Hongming Zhu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
45
|
Mortazavi-Jahromi SS, Aslani M, Mirshafiey A. A comprehensive review on miR-146a molecular mechanisms in a wide spectrum of immune and non-immune inflammatory diseases. Immunol Lett 2020; 227:8-27. [PMID: 32810557 DOI: 10.1016/j.imlet.2020.07.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/10/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are single-strand endogenous and non-coding RNA molecules with a length of about 22 nucleotides, which regulate genes expression, through modulating the translation and stability of their target mRNAs. miR-146a is one of the most studied miRNAs, due to its central role in immune system homeostasis and control of the innate and acquired immune responses. Accordingly, abnormal expression or function of miR-146a results in the incidence and progression of immune and non-immune inflammatory diseases. Its deregulated expression pattern and inefficient function have been reported in a wide spectrum of these illnesses. Based on the existing evidence, this miRNA qualifies as an ideal biomarker for diagnosis, prognosis, and activity evaluation of immune and non-immune inflammatory disorders. Moreover, much attention has recently been paid to therapeutic potential of miR-146a and several researchers have assessed the effects of different drugs on expression and function of this miRNA at diverse experimental, animal, besides human levels, reporting motivating results in the treatment of the diseases. Here, in this comprehensive review, we provide an overview of miR-146a role in the pathogenesis and progression of several immune and non-immune inflammatory diseases such as Rheumatoid arthritis, Systemic lupus erythematosus, Inflammatory bowel disease, Multiple sclerosis, Psoriasis, Graves' disease, Atherosclerosis, Hepatitis, Chronic obstructive pulmonary disease, etc., discuss about its eligibility for being a desirable biomarker for these disorders, and also highlight its therapeutic potential. Understanding these mechanisms underlies the selecting and designing the proper therapeutic targets and medications, which eventually facilitate the treatment process.
Collapse
Affiliation(s)
| | - Mona Aslani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Fasolo F, Di Gregoli K, Maegdefessel L, Johnson JL. Non-coding RNAs in cardiovascular cell biology and atherosclerosis. Cardiovasc Res 2020; 115:1732-1756. [PMID: 31389987 DOI: 10.1093/cvr/cvz203] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/14/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis underlies the predominant number of cardiovascular diseases and remains a leading cause of morbidity and mortality worldwide. The development, progression and formation of clinically relevant atherosclerotic plaques involves the interaction of distinct and over-lapping mechanisms which dictate the roles and actions of multiple resident and recruited cell types including endothelial cells, vascular smooth muscle cells, and monocyte/macrophages. The discovery of non-coding RNAs (ncRNAs) including microRNAs, long non-coding RNAs, and circular RNAs, and their identification as key mechanistic regulators of mRNA and protein expression has piqued interest in their potential contribution to atherosclerosis. Accruing evidence has revealed ncRNAs regulate pivotal cellular and molecular processes during all stages of atherosclerosis including cell invasion, growth, and survival; cellular uptake and efflux of lipids, expression and release of pro- and anti-inflammatory intermediaries, and proteolytic balance. The expression profile of ncRNAs within atherosclerotic lesions and the circulation have been determined with the aim of identifying individual or clusters of ncRNAs which may be viable therapeutic targets alongside deployment as biomarkers of atherosclerotic plaque progression. Consequently, numerous in vivo studies have been convened to determine the effects of moderating the function or expression of select ncRNAs in well-characterized animal models of atherosclerosis. Together, clinicopathological findings and studies in animal models have elucidated the multifaceted and frequently divergent effects ncRNAs impose both directly and indirectly on the formation and progression of atherosclerosis. From these findings' potential novel therapeutic targets and strategies have been discovered which may pave the way for further translational studies and possibly taken forward for clinical application.
Collapse
Affiliation(s)
- Francesca Fasolo
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar-Technical University Munich, Biedersteiner Strasse 29, Munich, Germany
| | - Karina Di Gregoli
- Laboratory of Cardiovascular Pathology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar-Technical University Munich, Biedersteiner Strasse 29, Munich, Germany.,Molecular Vascular Medicine, Karolinska Institute, Center for Molecular Medicine L8:03, 17176 Stockholm, Sweden.,German Center for Cardiovascular Research (DZHK), Partner Site Munich (Munich Heart Alliance), Munich, Germany
| | - Jason L Johnson
- Laboratory of Cardiovascular Pathology, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
47
|
Keller M, Gebhardt C, Huth S, Schleinitz D, Heyne H, Scholz M, Stumvoll M, Böttcher Y, Tönjes A, Kovacs P. Genetically programmed changes in transcription of the novel progranulin regulator. J Mol Med (Berl) 2020; 98:1139-1148. [PMID: 32620998 PMCID: PMC7399677 DOI: 10.1007/s00109-020-01942-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 01/02/2023]
Abstract
Abstract Progranulin is a glycoprotein marking chronic inflammation in obesity and type 2 diabetes. Previous studies suggested PSRC1 (proline and serine rich coiled-coil 1) to be a target of genetic variants associated with serum progranulin levels. We aimed to identify potentially functional variants and characterize their role in regulation of PSRC1. Phylogenetic module complexity analysis (PMCA) prioritized four polymorphisms (rs12740374, rs629301, rs660240, rs7528419) altering transcription factor binding sites with an overall score for potential regulatory function of Sall > 7.0. The effects of these variants on transcriptional activity and binding of transcription factors were tested by luciferase reporter and electrophoretic mobility shift assays (EMSA). In parallel, blood DNA promoter methylation of two regions was tested in subjects with a very high (N = 100) or a very low (N = 100) serum progranulin. Luciferase assays revealed lower activities in vectors carrying the rs629301-A compared with the C allele. Moreover, EMSA indicated a different binding pattern for the two rs629301 alleles, with an additional prominent band for the A allele, which was finally confirmed with the supershift for the Yin Yang 1 transcription factor (YY1). Subjects with high progranulin levels manifested a significantly higher mean DNA methylation (P < 1 × 10−7) in one promoter region, which was in line with a significantly lower PSRC1 mRNA expression levels in blood (P = 1 × 10−3). Consistently, rs629301-A allele was associated with lower PSRC1 mRNA expression (P < 1 × 10−7). Our data suggest that the progranulin-associated variant rs629301 modifies the transcription of PSRC1 through alteration of YY1 binding capacity. DNA methylation studies further support the role of PSRC1 in regulation of progranulin serum levels. Key messages PSRC1 (proline and serine rich coiled-coil 1) SNPs are associated with serum progranulin levels. rs629301 regulates PSRC1 expression by affecting Yin Yang 1 transcription factor (YY1) binding. PSRC1 is also epigenetically regulated in subjects with high progranulin levels.
Electronic supplementary material The online version of this article (10.1007/s00109-020-01942-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria Keller
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, University Hospital Leipzig, University of Leipzig, 04103, Leipzig, Germany.,Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, 20502, Malmö, Sweden.,Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Claudia Gebhardt
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, University Hospital Leipzig, University of Leipzig, 04103, Leipzig, Germany
| | - Sandra Huth
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103, Leipzig, Germany
| | - Dorit Schleinitz
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Henrike Heyne
- Institute of Human Genetics, University of Leipzig, 04103, Leipzig, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107, Leipzig, Germany
| | - Michael Stumvoll
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, University Hospital Leipzig, University of Leipzig, 04103, Leipzig, Germany.,Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Yvonne Böttcher
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Clinical Molecular Biology, Akershus Universitetssykehus, Lørenskog, Norway
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103, Leipzig, Germany.
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103, Leipzig, Germany.
| |
Collapse
|
48
|
Ntelios D, Efthimiadis G, Zegkos T, Didagelos M, Katopodi T, Meditskou S, Parcharidou D, Karvounis H, Tzimagiorgis G. Correlation of miR-146a-5p plasma levels and rs2910164 polymorphism with left ventricle outflow tract obstruction in hypertrophic cardiomyopathy. Hellenic J Cardiol 2020; 62:349-354. [PMID: 32389629 DOI: 10.1016/j.hjc.2020.04.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE Hypertrophic cardiomyopathy (HCM) is a genetic disease of the myocardium that is characterized by phenotypic variability among patients. miR-146a is a small non-coding RNA that is well known for its role in inflammation and myocardial hypertrophy. The aim of this study is to evaluate the role of miR-146a as a candidate genetic factor influencing HCM phenotype. METHODS In this study, 140 HCM patients and 112 control individuals were genotyped for the rs2910164 single nucleotide polymorphism (SNP) in the MIR146A gene; using this data, the correlation between different genotypes and clinical features of the disease were determined. Additionally, plasma levels of miR-146a-5p were determined in 50 HCM patients and 30 control individuals by using qPCR. RESULTS The incidence of GC and CC genotypes were significantly lower in HCM patients (odds ratio (OR) = 0.5 [0.3-0.8], p = 0.007). The GC/CC genotypes in the dominant genetic model positively correlated with the presence of left ventricle outflow tract (LVOT) obstruction (OR = 2.3 [1.2-4.7] and p = 0.018), a higher left ventricle mass index (118 ± 47 g/m2 vs 92 ± 42 g/m2 and p = 0.02), and increased left ventricle end-diastolic diameter (4.66 ± 0.64cm vs 4.39 ± 0.7cm and p = 0.026). Atrial fibrillation was significantly higher in patients homozygous for the C allele (OR = 10.6 [2-55], p = 0.003). Interestingly, the plasma levels of miR-146a-5p were significantly increased in HCM patients with LVOT obstruction. CONCLUSION Our findings indicate that the C allele of the rs2910164 SNP might be under negative selection in HCM patients. Additionally, plasma levels of miR-146a-5p and GC/CC genotypes are indicative of the obstructive phenotype in HCM patients.
Collapse
Affiliation(s)
- Dimitrios Ntelios
- Laboratory of Biological Chemistry, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece; First Department of Cardiology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Efthimiadis
- First Department of Cardiology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Thomas Zegkos
- First Department of Cardiology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Matthaios Didagelos
- First Department of Cardiology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theodora Katopodi
- Laboratory of Biology, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Soultana Meditskou
- Laboratory of Histology and Embryology, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Despoina Parcharidou
- First Department of Cardiology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Haralampos Karvounis
- First Department of Cardiology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Tzimagiorgis
- Laboratory of Biological Chemistry, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
49
|
Bao Y, Li S, Ding Y, Du X, Zhang M, Tang W, Zhou S. MiRNA: a potential target for gene diagnosis and treatment of atherosclerotic stroke. Int J Neurosci 2020; 131:283-288. [PMID: 32129115 DOI: 10.1080/00207454.2020.1738428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Stroke is one of the major diseases that endanger the physical health life of middle-aged and old people. It has the characteristics of high incidence, mortality and disability rate. Atherosclerosis is the main intervention target for stroke prevention and treatment. MiRNAs are highly expressed in the cerebral vasculature and play an important regulatory role in the pathogenesis of neuronal damage and ischemic stroke. This article reviews the mechanism of action between miRNAs and atherosclerosis, stroke, ischemia-reperfusion injury, collateral circulation and circRNA molecular networks, providing theoretical support for miRNA in gene diagnosis and drug therapy of atherosclerotic stroke.
Collapse
Affiliation(s)
- Yi Bao
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China.,Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Sijing Li
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Yayong Ding
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Xinyu Du
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Miao Zhang
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Wanjuan Tang
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Siqin Zhou
- Department of Neurology, Hubei University of Medicine, Shiyan City, Hubei Province, China
| |
Collapse
|
50
|
Abstract
Anthracycline-associated cardiomyopathy and peripartum cardiomyopathy are nonischemic cardiomyopathies that often afflict previously healthy young patients; both diseases have been well described since at least the 1970s and both occur in the settings of predictable stressors (ie, cancer treatment and pregnancy). Despite this, the precise mechanisms and the ability to reliably predict who exactly will go on to develop cardiomyopathy and heart failure in the face of anthracycline exposure or childbirth have proven elusive. For both cardiomyopathies, recent advances in basic and molecular sciences have illuminated the complex balance between cardiomyocyte and endothelial homeostasis via 3 broad pathways: reactive oxidative stress, interference in apoptosis/growth/metabolism, and angiogenic imbalance. These advances have already shown potential for specific, disease-altering therapies, and as our mechanistic knowledge continues to evolve, further clinical successes are expected to follow.
Collapse
Affiliation(s)
- Joshua A Cowgill
- From the Department of Cardiovascular Medicine, Maine Medical Center, Portland
| | - Sanjeev A Francis
- From the Department of Cardiovascular Medicine, Maine Medical Center, Portland
| | - Douglas B Sawyer
- From the Department of Cardiovascular Medicine, Maine Medical Center, Portland
| |
Collapse
|