1
|
Samani SL, Barlow SC, Freeburg LA, Catherwood GM, Churillo AM, Jones TL, Altomare D, Ji H, Shtutman M, Zile MR, Shazly T, Spinale FG. Heart failure with preserved ejection fraction in pigs causes shifts in posttranscriptional checkpoints. Am J Physiol Heart Circ Physiol 2024; 327:H1272-H1285. [PMID: 39240258 DOI: 10.1152/ajpheart.00551.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/07/2024]
Abstract
Left ventricular pressure overload (LVPO) can lead to heart failure with a preserved ejection fraction (HFpEF) and LV chamber stiffness (LV Kc) is a hallmark. This project tested the hypothesis that the development of HFpEF due to an LVPO stimulus will alter posttranscriptional regulation, specifically microRNAs (miRs). LVPO was induced in pigs (n = 9) by sequential ascending aortic cuff and age- and weight-matched pigs (n = 6) served as controls. LV function was measured by echocardiography and LV Kc by speckle tracking. LV myocardial miRs were quantified using an 84-miR array. Treadmill testing and natriuretic peptide-A (NPPA) mRNA levels in controls and LVPO were performed (n = 10, n = 9, respectively). LV samples from LVPO and controls (n = 6, respectively) were subjected to RNA sequencing. LV mass and Kc increased by over 40% with LVPO (P < 0.05). A total of 30 miRs shifted with LVPO of which 11 miRs correlated to LV Kc (P < 0.05) that mapped to functional domains relevant to Kc such as fibrosis and calcium handling. LVPO resulted in reduced exercise tolerance (oxygen saturation, respiratory effort) and NPPA mRNA levels increased by fourfold (P < 0.05). RNA analysis identified several genes that mapped to specific miRs that were altered with LVPO. In conclusion, a specific set of miRs are changed in a large animal model consistent with the HFpEF phenotype, were related to LV stiffness properties, and several miRs mapped to molecular pathways that may hold relevance in terms of prognosis and therapeutic targets.NEW & NOTEWORTHY Heart failure with preserved ejection fraction (HFpEF) is an ever-growing cause for the HF burden. HFpEF is particularly difficult to treat as the mechanisms responsible for this specific form of HF are poorly understood. Using a relevant large animal model, this study uncovered a unique molecular signature with the development of HFpEF that regulates specific biological pathways relevant to the progression of this ever-growing cause of HF.
Collapse
Affiliation(s)
- Stephanie L Samani
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Columbia Veteran Affairs Health Care System, Columbia, South Carolina, United States
| | - Shayne C Barlow
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Lisa A Freeburg
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Columbia Veteran Affairs Health Care System, Columbia, South Carolina, United States
| | - Grayson M Catherwood
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Amelia M Churillo
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Columbia Veteran Affairs Health Care System, Columbia, South Carolina, United States
| | - Traci L Jones
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Diego Altomare
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States
| | - Hao Ji
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States
| | - Michael Shtutman
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, United States
| | - Michael R Zile
- Division of Cardiology, Ralph H. Johnson Department of Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Tarek Shazly
- College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, United States
| | - Francis G Spinale
- Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, South Carolina, United States
- Columbia Veteran Affairs Health Care System, Columbia, South Carolina, United States
| |
Collapse
|
2
|
Yang G, Khan A, Liang W, Xiong Z, Stegbauer J. Aortic aneurysm: pathophysiology and therapeutic options. MedComm (Beijing) 2024; 5:e703. [PMID: 39247619 PMCID: PMC11380051 DOI: 10.1002/mco2.703] [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: 12/19/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Aortic aneurysm (AA) is an aortic disease with a high mortality rate, and other than surgery no effective preventive or therapeutic treatment have been developed. The renin-angiotensin system (RAS) is an important endocrine system that regulates vascular health. The ACE2/Ang-(1-7)/MasR axis can antagonize the adverse effects of the activation of the ACE/Ang II/AT1R axis on vascular dysfunction, atherosclerosis, and the development of aneurysms, thus providing an important therapeutic target for the prevention and treatment of AA. However, products targeting the Ang-(1-7)/MasR pathway still lack clinical validation. This review will outline the epidemiology of AA, including thoracic, abdominal, and thoracoabdominal AA, as well as current diagnostic and treatment strategies. Due to the highest incidence and most extensive research on abdominal AA (AAA), we will focus on AAA to explain the role of the RAS in its development, the protective function of Ang-(1-7)/MasR, and the mechanisms involved. We will also describe the roles of agonists and antagonists, suggest improvements in engineering and drug delivery, and provide evidence for Ang-(1-7)/MasR's clinical potential, discussing risks and solutions for clinical use. This study will enhance our understanding of AA and offer new possibilities and promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Guang Yang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Institute of Translational Medicine Shenzhen Second People's Hospital The First Affiliated Hospital of Shenzhen University Shenzhen China
- Department of Life Sciences Yuncheng University Yuncheng China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Abbas Khan
- Department of Nutrition and Health Promotion University of Home Economics Lahore Pakistan Lahore Pakistan
| | - Wei Liang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Zibo Xiong
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Johannes Stegbauer
- Department of Nephrology Medical Faculty University Hospital Düsseldorf Heinrich Heine University Düsseldorf Düsseldorf Germany
| |
Collapse
|
3
|
Zhang H, Zhang K, Gu Y, Tu Y, Ouyang C. Roles and Mechanisms of miRNAs in Abdominal Aortic Aneurysm: Signaling Pathways and Clinical Insights. Curr Atheroscler Rep 2024; 26:273-287. [PMID: 38709435 DOI: 10.1007/s11883-024-01204-8] [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] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE OF REVIEW Abdominal aortic aneurysm refers to a serious medical condition that can cause the irreversible expansion of the abdominal aorta, which can lead to ruptures that are associated with up to 80% mortality. Currently, surgical and interventional procedures are the only treatment options available for treating abdominal aortic aneurysm patients. In this review, we focus on the upstream and downstream molecules of the microRNA-related signaling pathways and discuss the roles, mechanisms, and targets of microRNAs in abdominal aortic aneurysm modulation to provide novel insights for precise and targeted drug therapy for the vast number of abdominal aortic aneurysm patients. RECENT FINDINGS Recent studies have highlighted that microRNAs, which are emerging as novel regulators of gene expression, are involved in the biological activities of regulating abdominal aortic aneurysms. Accumulating studies suggested that microRNAs modulate abdominal aortic aneurysm development through various signaling pathways that are yet to be comprehensively summarized. A total of six signaling pathways (NF-κB signaling pathway, PI3K/AKT signaling pathway, MAPK signaling pathway, TGF-β signaling pathway, Wnt signaling pathway, and P53/P21 signaling pathway), and a total of 19 miRNAs are intimately associated with the biological properties of abdominal aortic aneurysm through targeting various essential molecules. MicroRNAs modulate the formation, progression, and rupture of abdominal aortic aneurysm by regulating smooth muscle cell proliferation and phenotype change, vascular inflammation and endothelium function, and extracellular matrix remodeling. Because of the broad crosstalk among signaling pathways, a comprehensive analysis of miRNA-mediated signaling pathways is necessary to construct a well-rounded upstream and downstream regulatory network for future basic and clinical research of AAA therapy.
Collapse
Affiliation(s)
- Haorui Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Ke Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Yuanrui Gu
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Yanxia Tu
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China.
| |
Collapse
|
4
|
Liu Y, Sun X, Gou Z, Deng Z, Zhang Y, Zhao P, Sun W, Bai Y, Jing Y. Epigenetic modifications in abdominal aortic aneurysms: from basic to clinical. Front Cardiovasc Med 2024; 11:1394889. [PMID: 38895538 PMCID: PMC11183338 DOI: 10.3389/fcvm.2024.1394889] [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: 03/11/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Abdominal Aortic Aneurysm (AAA) is a disease characterized by localized dilation of the abdominal aorta, involving multiple factors in its occurrence and development, ultimately leading to vessel rupture and severe bleeding. AAA has a high mortality rate, and there is a lack of targeted therapeutic drugs. Epigenetic regulation plays a crucial role in AAA, and the treatment of AAA in the epigenetic field may involve a series of related genes and pathways. Abnormal expression of these genes may be a key factor in the occurrence of the disease and could potentially serve as promising therapeutic targets. Understanding the epigenetic regulation of AAA is of significant importance in revealing the mechanisms underlying the disease and identifying new therapeutic targets. This knowledge can contribute to offering AAA patients better clinical treatment options beyond surgery. This review systematically explores various aspects of epigenetic regulation in AAA, including DNA methylation, histone modification, non-coding RNA, and RNA modification. The analysis of the roles of these regulatory mechanisms, along with the identification of relevant genes and pathways associated with AAA, is discussed comprehensively. Additionally, a comprehensive discussion is provided on existing treatment strategies and prospects for epigenetics-based treatments, offering insights for future clinical interventions.
Collapse
Affiliation(s)
- YuChen Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - XiaoYun Sun
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Zhen Gou
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - ZhenKun Deng
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - YunRui Zhang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - PingPing Zhao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Wei Sun
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Yang Bai
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - YuChen Jing
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Khayati S, Dehnavi S, Sadeghi M, Tavakol Afshari J, Esmaeili SA, Mohammadi M. The potential role of miRNA in regulating macrophage polarization. Heliyon 2023; 9:e21615. [PMID: 38027572 PMCID: PMC10665754 DOI: 10.1016/j.heliyon.2023.e21615] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Macrophage polarization is a dynamic process determining the outcome of various physiological and pathological situations through inducing pro-inflammatory responses or resolving inflammation via exerting anti-inflammatory effects. The miRNAs are epigenetic regulators of different biologic pathways that target transcription factors and signaling molecules to promote macrophage phenotype transition and regulate immune responses. Modulating the macrophage activation, differentiation, and polarization by miRNAs is crucial for immune responses in response to microenvironmental signals and under various physiological and pathological conditions. In term of clinical significance, regulating macrophage polarization via miRNAs could be utilized for inflammation control. Also, understanding the role of miRNAs in macrophage polarization can provide insights into diagnostic strategies associated with dysregulated miRNAs and for developing macrophage-centered therapeutic methods. In this case, targeting miRNAs to further regulate of macrophage polarization may become an efficient strategy for treating immune-associated disorders. The current review investigated and categorized various miRNAs directly or indirectly involved in macrophage polarization by targeting different transcription factors and signaling pathways. In addition, prospects for regulating macrophage polarization via miRNA as a therapeutic choice that could be implicated in various pathological conditions, including cancer or inflammation-mediated injuries, were discussed.
Collapse
Affiliation(s)
- Shaho Khayati
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Mohammadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Wu X, Zhang Y, Wang P, Li X, Song Z, Wei C, Zhang Q, Luo B, Liu Z, Yang Y, Ren Z, Liu H. Clinical and preclinical evaluation of miR-144-5p as a key target for major depressive disorder. CNS Neurosci Ther 2023; 29:3598-3611. [PMID: 37308778 PMCID: PMC10580367 DOI: 10.1111/cns.14291] [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: 01/01/2023] [Revised: 04/06/2023] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Neuronal abnormalities are closely associated with major depressive disorder (MDD). Available evidence suggests a role for microRNAs (miRNAs) in regulating the expression of genes involved in MDD. Hence, miRNAs that can be potential therapeutic targets need to be identified. METHODS A mouse model of chronic unpredictable stress (CUS) was used to evaluate the function of miRNAs in MDD. miR-144-5p was screened from the hippocampi of CUS mice based on sequencing results. Adenovirus-associated vectors were used to overexpress or knockdown miR-144-5p in mice. BpV(pic) and LY294002 were used to determine the relationship between miR-144-5p target genes PTEN and TLR4 in neuronal impairment caused by miR-144-5p deficiency. Western blotting, immunofluorescence, ELISA immunosorbent assay, and Golgi staining were used to detect neuronal abnormalities. Serum samples from healthy individuals and patients with MDD were used to detect miR-144-5p levels in the serum and serum exosomes using qRT-PCR. RESULTS miR-144-5p expression was significantly decreased within the hippocampal dentate gyrus (DG) of CUS mice. Upregulation of miR-144-5p in the DG ameliorated depression-like behavior in CUS mice and attenuated neuronal abnormalities by directly targeting PTEN and TLR4 expression. Furthermore, miR-144-5p knockdown in normal mice led to depression-like behavior via inducing neuronal abnormalities, including abnormal neurogenesis, neuronal apoptosis, altered synaptic plasticity, and neuroinflammation. miR-144-5p deficiency-mediated neuronal impairment was mediated by PI3K/Akt/FoxO1 signaling. Furthermore, miR-144-5p levels were downregulated in the sera of patients with MDD and associated with depressive symptoms. Consistently, serum exosome-derived miR-144-5p levels were decreased in patients with MDD. CONCLUSION miR-144-5p plays a vital role in regulating neuronal abnormalities in depression. Our findings provide translational evidence that miR-144-5p is a new potential therapeutic target for MDD.
Collapse
Affiliation(s)
- Xiaodong Wu
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Yulong Zhang
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Ping Wang
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Xiaohui Li
- Department of AnatomyAnhui Medical UniversityHefeiChina
| | - Zhen Song
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Chuke Wei
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Qing Zhang
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Bei Luo
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Zhichun Liu
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Yingying Yang
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| | - Zhenhua Ren
- Department of AnatomyAnhui Medical UniversityHefeiChina
| | - Huanzhong Liu
- Department of PsychiatryChaohu Hospital of Anhui Medical UniversityHefeiChina
- Department of Psychiatry, School of Mental Health and Psychological SciencesAnhui Medical UniversityHefeiChina
- Department of Psychiatry, Anhui Psychiatric CenterAnhui Medical UniversityHefeiChina
| |
Collapse
|
7
|
Vyas H, Vohra A, Upadhyay K, Thounaojam M, Jadeja R, Dalvi N, Bartoli M, Devkar R. miR34a-5p impedes CLOCK expression in chronodisruptive C57BL/6J mice and potentiates pro-atherogenic manifestations. PLoS One 2023; 18:e0283591. [PMID: 37561715 PMCID: PMC10414636 DOI: 10.1371/journal.pone.0283591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/11/2023] [Indexed: 08/12/2023] Open
Abstract
INTRODUCTION Altered circadian rhythms underlie manifestation of several cardiovascular disorders, however a little is known about the mediating biomolecules. Multiple transcriptional-translational feedback loops control circadian-clockwork wherein; micro RNAs (miRNAs) are known to manifest post transcriptional regulation. This study assesses miR34a-5p as a mediating biomolecule. METHOD 8-10-week-old male C57BL/6J mice (n = 6/group) were subjected to photoperiodic manipulation induced chronodisruption and thoracic aortae were examined for miRNA, gene (qPCR) and protein (Immunoblot) expression studies. Histomorphological changes were assessed for pro-atherogenic manifestations (fibrillar arrangement, collagen/elastin ratio, intima-media thickening). Computational studies for miRNA-mRNA target prediction were done using TargetScan and miRDB. Correlative in vitro studies were done in serum synchronized HUVEC cells. Time point based studies were done at five time points (ZT 0, 6, 12, 18, 24) in 24h. RESULTS Chronodisruption induced hypomethylation in the promoter region of miR34a-5p, in the thoracic aortae, culminating in elevated miRNA titers. In a software-based detection of circadian-clock-associated targets of miR34a-5p, Clock and Sirt1 genes were identified. Moreover, miR34a-5p exhibited antagonist circadian oscillations to that of its target genes CLOCK and SIRT1 in endothelial cells. Luciferase reporter gene assay further showed that miR34a-5p interacts with the 3'UTR of the Clock gene to lower its expression, disturbing the operation of positive arm of circadian clock system. Elevated miR34a-5p and impeded SIRT1 expression in a chronodisruptive aortae exhibited pro-atherogenic changes observed in form of gene expression, increased collagen/elastin ratio, fibrillar derangement and intimal-media thickening. CONCLUSION The study reports for the first time chronodisruption mediated miR34a-5p elevation, its circadian expression and interaction with the 3'UTR of Clock gene to impede its expression. Moreover, elevated miR34a-5p and lowered SIRT1 expression in the chronodisruptive aortae lead off cause-consequence relationship of chronodisruption mediated proatherogenic changes.
Collapse
Affiliation(s)
- Hitarthi Vyas
- Division of Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Aliasgar Vohra
- Division of Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Kapil Upadhyay
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Menaka Thounaojam
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, United States of America
| | - Ravirajsinh Jadeja
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, United States of America
| | - Nilay Dalvi
- Division of Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Manuela Bartoli
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Ranjitsinh Devkar
- Division of Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
8
|
Zhang MQ, Wang CC, Pang XB, Shi JZ, Li HR, Xie XM, Wang Z, Zhang HD, Zhou YF, Chen JW, Han ZY, Zhao LL, He YY. Role of macrophages in pulmonary arterial hypertension. Front Immunol 2023; 14:1152881. [PMID: 37153557 PMCID: PMC10154553 DOI: 10.3389/fimmu.2023.1152881] [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: 01/28/2023] [Accepted: 03/27/2023] [Indexed: 05/09/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe cardiopulmonary vascular disease characterized by progressive pulmonary artery pressure elevation, increased pulmonary vascular resistance and ultimately right heart failure. Studies have demonstrated the involvement of multiple immune cells in the development of PAH in patients with PAH and in experimental PAH. Among them, macrophages, as the predominant inflammatory cells infiltrating around PAH lesions, play a crucial role in exacerbating pulmonary vascular remodeling in PAH. Macrophages are generally polarized into (classic) M1 and (alternative) M2 phenotypes, they accelerate the process of PAH by secreting various chemokines and growth factors (CX3CR1, PDGF). In this review we summarize the mechanisms of immune cell action in PAH, as well as the key factors that regulate the polarization of macrophages in different directions and their functional changes after polarization. We also summarize the effects of different microenvironments on macrophages in PAH. The insight into the interactions between macrophages and other cells, chemokines and growth factors may provide important clues for the development of new, safe and effective immune-targeted therapies for PAH.
Collapse
Affiliation(s)
- Meng-Qi Zhang
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Chen-Chen Wang
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Xiao-Bin Pang
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Jun-Zhuo Shi
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Hao-Ran Li
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Xin-Mei Xie
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Zhe Wang
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Hong-Da Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun-Feng Zhou
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Ji-Wang Chen
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Zhi-Yan Han
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yang-Yang He, ; Lu-Ling Zhao, ; Zhi-Yan Han,
| | - Lu-Ling Zhao
- School of Pharmacy, Henan University, Kaifeng, Henan, China
- *Correspondence: Yang-Yang He, ; Lu-Ling Zhao, ; Zhi-Yan Han,
| | - Yang-Yang He
- School of Pharmacy, Henan University, Kaifeng, Henan, China
- *Correspondence: Yang-Yang He, ; Lu-Ling Zhao, ; Zhi-Yan Han,
| |
Collapse
|
9
|
Madè A, Greco S, Vausort M, Miliotis M, Schordan E, Baksi S, Zhang L, Baryshnikova E, Ranucci M, Cardani R, Fagherazzi G, Ollert M, Tastsoglou S, Vatsellas G, Hatzigeorgiou A, Firat H, Devaux Y, Martelli F. Association of miR-144 levels in the peripheral blood with COVID-19 severity and mortality. Sci Rep 2022; 12:20048. [PMID: 36414650 PMCID: PMC9681736 DOI: 10.1038/s41598-022-23922-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) can be asymptomatic or lead to a wide symptom spectrum, including multi-organ damage and death. Here, we explored the potential of microRNAs in delineating patient condition and predicting clinical outcome. Plasma microRNA profiling of hospitalized COVID-19 patients showed that miR-144-3p was dynamically regulated in response to COVID-19. Thus, we further investigated the biomarker potential of miR-144-3p measured at admission in 179 COVID-19 patients and 29 healthy controls recruited in three centers. In hospitalized patients, circulating miR-144-3p levels discriminated between non-critical and critical illness (AUCmiR-144-3p = 0.71; p = 0.0006), acting also as mortality predictor (AUCmiR-144-3p = 0.67; p = 0.004). In non-hospitalized patients, plasma miR-144-3p levels discriminated mild from moderate disease (AUCmiR-144-3p = 0.67; p = 0.03). Uncontrolled release of pro-inflammatory cytokines can lead to clinical deterioration. Thus, we explored the added value of a miR-144/cytokine combined analysis in the assessment of hospitalized COVID-19 patients. A miR-144-3p/Epidermal Growth Factor (EGF) combined score discriminated between non-critical and critical hospitalized patients (AUCmiR-144-3p/EGF = 0.81; p < 0.0001); moreover, a miR-144-3p/Interleukin-10 (IL-10) score discriminated survivors from nonsurvivors (AUCmiR-144-3p/IL-10 = 0.83; p < 0.0001). In conclusion, circulating miR-144-3p, possibly in combination with IL-10 or EGF, emerges as a noninvasive tool for early risk-based stratification and mortality prediction in COVID-19.
Collapse
Affiliation(s)
- Alisia Madè
- grid.419557.b0000 0004 1766 7370Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, MI Italy
| | - Simona Greco
- grid.419557.b0000 0004 1766 7370Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, MI Italy
| | - Melanie Vausort
- grid.451012.30000 0004 0621 531XCardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
| | - Marios Miliotis
- grid.418497.7Hellenic Pasteur Institute, 11521 Athens, Greece ,grid.410558.d0000 0001 0035 6670DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece
| | - Eric Schordan
- grid.450762.2Firalis SA, 35 Rue du Fort, 68330 Huningue, France
| | - Shounak Baksi
- grid.451012.30000 0004 0621 531XCardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
| | - Lu Zhang
- grid.451012.30000 0004 0621 531XBioinformatics Platform, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg
| | - Ekaterina Baryshnikova
- grid.419557.b0000 0004 1766 7370Department of Cardiovascular Anesthesia and ICU, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, MI Italy
| | - Marco Ranucci
- grid.419557.b0000 0004 1766 7370Department of Cardiovascular Anesthesia and ICU, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, MI Italy
| | - Rosanna Cardani
- grid.419557.b0000 0004 1766 7370BioCor Biobank, UOC SMEL-1 of Clinical Pathology, Department of Pathology and Laboratory Medicine, IRCCS-Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, MI Italy
| | - Guy Fagherazzi
- grid.451012.30000 0004 0621 531XDeep Digital Phenotyping Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1 A-B Rue Thomas Edison, 1445 Strassen, Luxembourg
| | - Markus Ollert
- grid.451012.30000 0004 0621 531XDepartment of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, 4354 Esch-Sur-Alzette, Luxembourg ,grid.10825.3e0000 0001 0728 0170Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis (ORCA), University of Southern Denmark, 5000 Odense, Denmark
| | - Spyros Tastsoglou
- grid.418497.7Hellenic Pasteur Institute, 11521 Athens, Greece ,grid.410558.d0000 0001 0035 6670DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece
| | - Giannis Vatsellas
- grid.417593.d0000 0001 2358 8802Greek Genome Center, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | - Artemis Hatzigeorgiou
- grid.418497.7Hellenic Pasteur Institute, 11521 Athens, Greece ,grid.410558.d0000 0001 0035 6670DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece
| | - Hüseyin Firat
- grid.450762.2Firalis SA, 35 Rue du Fort, 68330 Huningue, France
| | - Yvan Devaux
- grid.451012.30000 0004 0621 531XCardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
| | - Fabio Martelli
- grid.419557.b0000 0004 1766 7370Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, MI Italy
| |
Collapse
|
10
|
Macrophage polarization in THP-1 cell line and primary monocytes: A systematic review. Differentiation 2022; 128:67-82. [DOI: 10.1016/j.diff.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/27/2022] [Accepted: 10/02/2022] [Indexed: 11/21/2022]
|
11
|
Chen L, Wang S, Wang Z, Liu Y, Xu Y, Yang S, Xue G. Construction and analysis of competing endogenous RNA network and patterns of immune infiltration in abdominal aortic aneurysm. Front Cardiovasc Med 2022; 9:955838. [PMID: 35990982 PMCID: PMC9386163 DOI: 10.3389/fcvm.2022.955838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/12/2022] [Indexed: 11/29/2022] Open
Abstract
Background Various studies have highlighted the role of circular RNAs (circRNAs) as critical molecular regulators in cardiovascular diseases, but its role in abdominal aortic aneurysm (AAA) is unclear. This study explores the potential molecular mechanisms of AAA based on the circRNA-microRNA (miRNA)-mRNA competing endogenous RNA (ceRNA) network and immune cell infiltration patterns. Methods The expression profiles of circRNAs (GSE144431) and mRNAs (GSE57691 and GSE47472) were obtained from the Gene Expression Omnibus (GEO). Then, the differentially expressed circRNAs (DEcircRNAs) and mRNAs (DEmRNAs) between AAA patients and healthy control samples, and the target miRNAs of these DEmRNAs and DEcircRNAs were identified. Based on the miRNA-DEmRNAs and miRNA-DEcircRNAs pairs, the ceRNA network was constructed. Furthermore, the proportion of the 22 immune cell types in AAA patients was assessed using cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm. The expressions of key genes and immune cell infiltration were validated using clinical specimens. Results A total of 214 DEmRNAs were identified in the GSE57691 and GSE47472 datasets, and 517 DEcircRNAs were identified in the GSE144431 dataset. The ceRNA network included 19 circRNAs, 36 mRNAs, and 68 miRNAs. Two key genes, PPARG and FOXO1, were identified among the hub genes of the established protein-protein interaction between mRNAs in the ceRNA network. Moreover, seven types of immune cells were differentially expressed between AAA patients and healthy control samples. Hub genes in ceRNA, such as FOXO1, HSPA8, and RAB5C, positively correlated with resting CD4 memory T cells or M1 macrophages, or both. Conclusion In conclusion, a ceRNA interaction axis was constructed. The composition of infiltrating immune cells was analyzed in the abdominal aorta of AAA patients and healthy control samples. This may help identify potential therapeutic targets for AAA.
Collapse
Affiliation(s)
- Liang Chen
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuangshuang Wang
- Songyuan Central Hospital, Songyuan Children's Hospital, Songyuan, China
| | - Zheyu Wang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yuting Liu
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yi Xu
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- *Correspondence: Shuofei Yang
| | - Guanhua Xue
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Guanhua Xue
| |
Collapse
|
12
|
Chang S, Chang M, Liu G, Xu D, Wang H, Sun R, Feng M. LncRNA OIP5-AS1 reduces renal epithelial cell apoptosis in cisplatin-induced AKI by regulating the miR-144-5p/PKM2 axis. Biomed J 2022; 45:642-653. [PMID: 34311128 PMCID: PMC9486127 DOI: 10.1016/j.bj.2021.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 07/05/2021] [Accepted: 07/19/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The abnormal expression of long non-coding RNA (lncRNA) Opa-interacting protein 5 antisense RNA 1 (OIP5-AS1) has been observed in many human cancers and the underlying mechanisms have been well studied. However, the function of OIP5-AS1 in acute kidney injury (AKI) remains unclear. METHODS To explore the role of OIP5-AS1 in the progression of AKI, the cisplatin-induced AKI mouse and cell model were established. To confirm the potential protective effect of OIP5-AS1 during cisplatin-induced AKI, rescue experiments were performed. Targetscan was used to predict the potential targets of miR-144-5p. To further determine whether the effect of miR-144-5p during cisplatin-induced AKI was mediated by PMK2, the recuse experiments using PMK2 overexpressing vector was applied. RESULTS OIP5-AS1 was significantly downregulated both in cisplatin-induced AKI mice and human renal tubular cell line HK-2 cells. Moreover, overexpression of OIP5-AS1 efficiently promoted cell growth and reduced cisplatin-induced apoptosis of HK-2 cells. Furthermore, OIP5-AS1 was identified as a sponge of miR-144-5p, and upregulation of miR-144-5p could significantly reverse overexpression of OIP5-AS1-induced protective effect on the damage of cisplatin to HK-2 cells. In addition, pyruvate kinase M2 (PKM2) was found to be a direct target of miR-144-5p, and overexpression of PKM2 efficiently reversed the effect of miR-144-5p mimics on the damage in cisplatin-stimulated HK-2 cells. CONCLUSIONS OIP5-AS1 reduced the apoptosis of cisplatin-stimulated renal epithelial cells by targeting the miR-144-5p/PKM2 axis, which extended the regulatory network of lncRNAs in cisplatin-induced AKI and also provided a novel therapeutic target for AKI treatment.
Collapse
Affiliation(s)
- Siyuan Chang
- Department of SICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, PR China
| | - Mingyang Chang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, PR China
| | - Gang Liu
- Department of SICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, PR China
| | - Daqian Xu
- Department of SICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, PR China
| | - Haili Wang
- Department of SICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, PR China
| | - Rongqing Sun
- Department of SICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, PR China
| | - Min Feng
- Department of SICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, PR China.
| |
Collapse
|
13
|
Onuoha CP, Ipe J, Simpson E, Liu Y, Skaar T, Kreutz RP. MicroRNA
sequencing in patients with coronary artery disease – considerations for use as biomarker for thrombotic risk. Clin Transl Sci 2022; 15:1946-1958. [PMID: 35643946 PMCID: PMC9372418 DOI: 10.1111/cts.13307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/24/2022] [Accepted: 04/30/2022] [Indexed: 12/05/2022] Open
Abstract
MicroRNAs (miRNAs) are small RNAs integral in the regulation of gene expression. Analysis of circulating miRNA levels may identify patients with coronary artery disease (CAD) at risk for recurrent myocardial infarction (MI) after percutaneous coronary interventions (PCIs). Subjects with CAD were selected from the GENCATH cardiac catheterization biobank. Subjects with recurrent MI after PCI were compared with those without recurrent MI during follow‐up in the initial (n = 48) and replication cohort (n = 67). Next generation MiRNA sequencing was performed on plasma samples and whole blood samples fixed with PAXGENE tubes upon collection. Overall, 164 miRNAs derived from whole blood were differentially expressed in the replication cohort between subjects with and without recurrent MI events (p < 0.05), with 69 remaining significant after false‐discovery rate (FDR) correction. None of the miRNAs in plasma was significantly different by FDR among subjects with and without MI. Overall, correlation between direction of effects between plasma and whole blood assays was variable, and only two miRNAs were concordant and significant in both. Associations of miRNA with vascular disease, MI, and thrombosis were further explored. MiRNA profiling has potential as the future biomarker for disease prognosis and treatment response marker in secondary treatment of patients with CAD after PCI. Whole blood may be the preferred sample source as compared to plasma.
Collapse
Affiliation(s)
- Chimnonso P. Onuoha
- Department of Medicine/Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Joseph Ipe
- Department of Medicine/Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Edward Simpson
- Center for Medical Genomics Indiana University School of Medicine Indianapolis Indiana USA
| | - Yunlong Liu
- Center for Medical Genomics Indiana University School of Medicine Indianapolis Indiana USA
| | - Todd C. Skaar
- Department of Medicine/Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Rolf P. Kreutz
- Department of Medicine/Cardiology Indiana University School of Medicine Indianapolis Indiana USA
| |
Collapse
|
14
|
Zhou F, Zheng Z, Zha Z, Xiong T, Pan Y. Nuclear Paraspeckle Assembly Transcript 1 Enhances Hydrogen Peroxide-Induced Human Vascular Smooth Muscle Cell Injury by Regulating miR-30d-5p/A Disintegrin and Metalloprotease 10. Circ J 2022; 86:1007-1018. [PMID: 34880199 DOI: 10.1253/circj.cj-21-0042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Nuclear paraspeckle assembly transcript 1 (NEAT1) has been reported to be involved in the progression of many cancers; however, the role and mechanisms underlying NEAT1 in abdominal aortic aneurysm (AAA) remain unclear. METHODS AND RESULTS The expression of NEAT1, miR-30d-5p and A disintegrin and metalloprotease 10 (ADAM10) was measured by qRT-PCR and western blot. Functional experiments were conducted by using a CCK-8 assay, EDU assay, flow cytometry, western blot, ELISA, and commercial kits. The target relation was confirmed by dual-luciferase reporter assay and the RIP assay. It was then found that NEAT1 was upregulated in peripheral blood of AAA patients ~3.46-fold, smooth muscle cells (SMCs) isolated from AAA tissues ~2.6-fold and in a hydrogen peroxide (H2O2)-induced injury model of human vascular SMC (HVSMCs) ~2.0- and 3.9-fold at 50 µmol/L and 200 µmol/L H2O2treatment, respectively. NEAT1 deletion attenuated H2O2-induced cell proliferation promotion (40.0% vs. 74.3%), apoptosis inhibition (25.0% vs. 13.5%), and reduction of inflammatory response and oxidative stress in HVSMCs. Mechanistically, NEAT1 targeted miR-30d-5p to prevent the degradation of its target, ADAM10, in HVSMCs. Further rescue experiments suggested miR-30d-5p inhibition mitigated the effects of NEAT1 deletion on H2O2-induced HVSMCs. Moreover, ADAM10 overexpression counteracted the inhibitory functions of miR-30d-5p on H2O2-evoked HVSMC injury. CONCLUSIONS NEAT1 promoted H2O2-induced HVSMC injury by inducing cell apoptosis, inflammation and oxidative stress through miR-30d-5p/ADAM10 axis, indicating the possible involvement of NEAT1 in the pathogenesis of AAA.
Collapse
Affiliation(s)
- Fushuo Zhou
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Zhi Zheng
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Zhengbiao Zha
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Tianxin Xiong
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Youmin Pan
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| |
Collapse
|
15
|
Song H, Yang Y, Sun Y, Wei G, Zheng H, Chen Y, Cai D, Li C, Ma Y, Lin Z, Shi X, Liao W, Liao Y, Zhong L, Bin J. Circular RNA Cdyl promotes abdominal aortic aneurysm formation by inducing M1 macrophage polarization and M1-type inflammation. Mol Ther 2022; 30:915-931. [PMID: 34547461 PMCID: PMC8821928 DOI: 10.1016/j.ymthe.2021.09.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/09/2021] [Accepted: 09/14/2021] [Indexed: 02/04/2023] Open
Abstract
Macrophage polarization plays a crucial role in regulating abdominal aortic aneurysm (AAA) formation. Circular RNAs (circRNAs) are important regulators of macrophage polarization during the development of cardiovascular diseases. How-ever, the roles of circRNAs in regulating AAA formation through modulation of macrophage polarization remain unknown. In the present study, we compared circRNA microarray data under two distinct polarizing conditions (M1 and M2 macrophages) and identified an M1-enriched circRNA, circCdyl. Loss- and gain-of-function assay results demonstrated that circCdyl overexpression accelerated angiotensin II (Ang II)- and calcium chloride (CaCl2)-induced AAA formation by promoting M1 polarization and M1-type inflammation, while circCdyl deficiency showed the opposite effects. RNA pulldown, mass spectrometry analysis, and RNA immunoprecipitation (RIP) assays were conducted to elucidate the underlying mechanisms by which circCdyl regulates AAA formation and showed that circCdyl promotes vascular inflammation and M1 polarization by inhibiting interferon regulatory factor 4 (IRF4) entry into the nucleus, significantly inducing AAA formation. In addition, circCdyl was shown to act as a let-7c sponge, promoting C/EBP-δ expression in macrophages to induce M1 polarization. Our results indicate an important role for circCdyl-mediated macrophage polarization in AAA formation and provide a potent therapeutic target for AAA treatment.
Collapse
Affiliation(s)
- Haoyu Song
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Wards of Cadres, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China
| | - Yang Yang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Yili Sun
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Guoquan Wei
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Hao Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Yijin Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Donghua Cai
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Chuling Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Yusheng Ma
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Zhongqiu Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Xiaoran Shi
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Lintao Zhong
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China,Corresponding author: Lintao Zhong, MD, PhD, Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China.
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China,Corresponding author: Jianping Bin, MD, PhD, Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
16
|
Si K, Lu D, Tian J. Integrated analysis and the identification of a circRNA-miRNA-mRNA network in the progression of abdominal aortic aneurysm. PeerJ 2022; 9:e12682. [PMID: 35036156 PMCID: PMC8711282 DOI: 10.7717/peerj.12682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/02/2021] [Indexed: 11/20/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a disease commonly seen in the elderly. The aneurysm diameter increases yearly, and the larger the AAA the higher the risk of rupture, increasing the risk of death. However, there are no current effective interventions in the early stages of AAA. Methods Four gene expression profiling datasets, including 23 normal artery (NOR) tissue samples and 97 AAA tissue samples, were integrated in order to explore potential molecular biological targets for early intervention. After preprocessing, differentially expressed genes (DEGs) between AAA and NOR were identified using LIMMA package. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis were conducted using the DAVID database. The protein-protein interaction network was constructed and hub genes were identified using the STRING database and plugins in Cytoscape. A circular RNA (circRNA) profile of four NOR tissues versus four AAA tissues was then reanalyzed. A circRNA-miRNA-mRNA interaction network was constructed after predictions were made using the Targetscan and Circinteractome databases. Results A total of 440 DEGs (263 up-regulated and 177 down-regulated) were identified in the AAA group, compared with the NOR group. The majority were associated with the extracellular matrix, tumor necrosis factor-α, and transforming growth factor-β. Ten hub gene-encoded proteins (namely IL6, RPS27A, JUN, UBC, UBA52, FOS, IL1B, MMP9, SPP1 and CCL2) coupled with a higher degree of connectivity hub were identified after protein‐protein interaction network analysis. Our results, in combination with the results of previous studies revealed that miR-635, miR-527, miR-520h, miR-938 and miR-518a-5p may be affected by circ_0005073 and impact the expression of hub genes such as CCL2, SPP1 and UBA52. The miR-1206 may also be affected by circ_0090069 and impact RPS27A expression. Conclusions This circRNA-miRNA-mRNA network may perform critical roles in AAA and may be a novel target for early intervention.
Collapse
Affiliation(s)
- Ke Si
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Da Lu
- Department of Vascular Surgery, Shanghai General Hospital, Shanghai, People's Republic of China
| | - Jianbo Tian
- Institute of Information Engineering, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
17
|
Huang Y, Ren L, Li J, Zou H. Long non-coding RNA PVT1/microRNA miR-3127-5p/NCK-associated protein 1-like axis participates in the pathogenesis of abdominal aortic aneurysm by regulating vascular smooth muscle cells. Bioengineered 2021; 12:12583-12596. [PMID: 34898354 PMCID: PMC8810122 DOI: 10.1080/21655979.2021.2010384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The long non-coding RNA plasmacytoma variant translocation 1 (lncRNA PVT1) has been implicated in the progression of abdominal aortic aneurysms (AAA). However, the detailed mechanism requires further analysis. Our study was aimed at interrogating the mechanism of PVT1 in an H2O2-induced AAA model in vitro. The expression of lncRNA PVT1, microRNA miR-3127-5p, and NCK-associated protein 1-like (NCKAP1L) was examined in AAA tissues and H2O2-treated vascular smooth muscle cells (VSMCs). Cell proliferation was assayed using Cell Counting Kit-8 (CCK8) and 5-Bromodeoxyuridine (BrdU) assays. Meanwhile, 5-Ethynyl-2'-deoxyuridine (EdU) staining was performed to assess cell apoptosis and caspase-3 activity. IL-1β and caspase-1 expression was also assessed using Western blotting to determine inflammasome activation in H2O2-treated VSMCs. Luciferase reporter assays addressed the possible interaction between miR-3127-5p and PVT1 or NCKAP1L, which was predicted by starBase analysis. PVT1 and NCKAP1L expression was elevated in AAA tissues and induced the AAA model in vitro, whereas miR-3127-5p showed the opposite trend. Functionally, PVT1 silencing promoted cell proliferation and reduced the apoptotic rate and inflammasome activation in H2O2-treated VSMCs. Mechanical investigation demonstrated that PVT1 acted as a sponge of miR-3127-5p to modulate NCKAP1L expression, resulting in suppression of VSMC proliferation, induction of apoptosis, and activation of inflammation. In conclusion, PVT1 participates in AAA progression through the miR-3127-5p/NCKAP1L axis and may be a promising biosignature and therapeutic target for AAA.
Collapse
Affiliation(s)
- Youjin Huang
- Department of Vascular Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Li Ren
- Department of Vascular Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Jiajia Li
- Intensive Care Unit, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Haibo Zou
- Department of Hepatobiliary Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Yang G, Qin H, Liu B, Zhao X, Yin H. Mesenchymal stem cells-derived exosomes modulate vascular endothelial injury via miR-144-5p/PTEN in intracranial aneurysm. Hum Cell 2021; 34:1346-1359. [PMID: 34240392 DOI: 10.1007/s13577-021-00571-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/22/2021] [Indexed: 12/27/2022]
Abstract
Phosphatase and tensin homolog (PTEN) is known to be involved in the pathogenesis of intracranial aneurysm (IA). This study investigated the molecular mechanism of exosomal miR-144-5p (ex-miR-144-5p) and PTEN in IA. Ex-miR-144-5p expression was assessed in serum from individuals with ruptured intracranial aneurysm (RA) or unruptured intracranial aneurysm (UA), and healthy controls (HC). Vascular endothelial cells (VECs) were co-cultured with exosomes isolated from mesenchymal stem cells (MSCs) with transfection of miR-144-5p mimic or miR-144-5p inhibitor. IA rats were induced by combing systemic hypertension and intrathecal elastase injection. VECs were transfected with miR-144-5p mimic or inhibitor to verify the impacts of miR-144-5p on cell viability and proliferation. The connection between miR-144-5p and PTEN was verified by luciferase activity assay. Our data proved that ex-miR-144-5p was decreased in both UA and RA patients. MiR-144-5p overexpression in MSCs-derived exosome promoted VEC viability, inhibited VEC proliferation of VEs, and decreased the protein levels of matrix metalloproteinase-9 (MMP-9), proliferating cell nuclear antigen (PCNA) and osteopontin (OPN). IA rats injected with ex-miR-144-5p mimic showed significant luminal dilation, declined smooth muscle layers, and thinned vascular wall. Besides, inhibited cell apoptosis and decreased protein expressions were also observed. However, ex-miR-144-5p inhibitor had the opposite effects both in vivo and in vitro. We validated that miR-144-5p directly targeted PTEN. MiR-144-5p mimic increased cell viability and proliferation and reduced protein expressions, which could be blunted by PTEN overexpression. This study suggests that miR-144-5p elevates PTEN expression, thereby boosting apoptosis and attenuating viability of VECs in IA.
Collapse
Affiliation(s)
- Guojun Yang
- Department of Neurosurgery, Affiliated Hospital of Chengde Medical University, Chengde City, Hebei Province, 067000, People's Republic of China
| | - Hao Qin
- Department of Neurosurgery, Zaozhuang Municipal Hospital, No. 41 Longtou Middle Road, Shizhong District, Zaozhuang City, Shandong Province, 277100, People's Republic of China
| | - Bing Liu
- Department of Neurosurgery, Affiliated Hospital of Chengde Medical University, Chengde City, Hebei Province, 067000, People's Republic of China
| | - Xinhong Zhao
- Pharmacy Department, Affiliated Hospital of Chengde Medical University, Chengde City, Hebei Province, 067000, People's Republic of China
| | - Hang Yin
- Department of Neurosurgery, Zaozhuang Municipal Hospital, No. 41 Longtou Middle Road, Shizhong District, Zaozhuang City, Shandong Province, 277100, People's Republic of China.
| |
Collapse
|
19
|
Law YY, Lee WF, Hsu CJ, Lin YY, Tsai CH, Huang CC, Wu MH, Tang CH, Liu JF. miR-let-7c-5p and miR-149-5p inhibit proinflammatory cytokine production in osteoarthritis and rheumatoid arthritis synovial fibroblasts. Aging (Albany NY) 2021; 13:17227-17236. [PMID: 34198264 PMCID: PMC8312412 DOI: 10.18632/aging.203201] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are two of the most common types of arthritis. Both are characterized by the infiltration of a number of proinflammatory cytokines into the joint microenvironment. miRNAs play critical roles in the disease processes of arthritic disorders. However, little is known about the effects of miRNAs on critical inflammatory cytokine production with OA and RA progression. Here, we found higher levels of proinflammatory cytokines including interleukin 1 beta (IL-1β), interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) in human OA and RA synovial fibroblasts (SFs) compared with normal SFs. Searches of open-source microRNA (miRNA) software determined that miR-let-7c-5p and miR-149-5p interfere with IL-1β, IL-6 and TNF-α transcription; levels of all three proinflammatory cytokines were lower in human OA and RA patients compared with normal controls. Anti-inflammatory agents dexamethasone, celecoxib and indomethacin reduced proinflammatory cytokine production by promoting the expression of miR-let-7c-5p and miR-149-5p. Similarly, ibuprofen and methotrexate also enhanced miR-let-7c-5p and miR-149-5p expression in human SFs. The evidence suggests that increasing miR-let-7c-5p and miR-149-5p expression is a novel strategy for OA and RA.
Collapse
Affiliation(s)
- Yat-Yin Law
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wei-Fang Lee
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Jung Hsu
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yen-You Lin
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan.,Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Chien-Chung Huang
- School of Medicine, China Medical University, Taichung, Taiwan.,Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Min-Huan Wu
- Bachelor of Science in Senior Wellness and Sports Science, Tunghai University, Taichung, Taiwan.,Tunghai University Sports Recreation and Health Management Degree Program, Tunghai University, Taichung, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
20
|
Yang D, Yang L, Cai J, Hu X, Li H, Zhang X, Zhang X, Chen X, Dong H, Nie H, Li Y. A sweet spot for macrophages: Focusing on polarization. Pharmacol Res 2021; 167:105576. [PMID: 33771700 DOI: 10.1016/j.phrs.2021.105576] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/21/2022]
Abstract
Macrophages are a type of functionally plastic cells that can create a pro-/anti-inflammatory microenvironment for organs by producing different kinds of cytokines, chemokines, and growth factors to regulate immunity and inflammatory responses. In addition, they can also be induced to adopt different phenotypes in response to extracellular and intracellular signals, a process defined as M1/M2 polarization. Growing evidence indicates that glycobiology is closely associated with this polarization process. In this research, we review studies of the roles of glycosylation, glucose metabolism, and key lectins in the regulation of macrophages function and polarization to provide a new perspective for immunotherapies for multiple diseases.
Collapse
Affiliation(s)
- Depeng Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lijun Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110000, China
| | - Xibo Hu
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huaxin Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaoqing Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaohan Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xinghe Chen
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Haiyang Dong
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huan Nie
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| | - Yu Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
21
|
Emerging roles of non-coding RNAs in the metabolic reprogramming of tumor-associated macrophages. Immunol Lett 2021; 232:27-34. [PMID: 33577913 DOI: 10.1016/j.imlet.2021.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 01/09/2023]
Abstract
Macrophages are the most common immune cells in the tumor microenvironment, and tumor-associated macrophages play an important role in cancer development. Metabolic reprogramming is important for the functional plasticity of macrophages. Studies investigating the relevance of non-coding RNAs (ncRNAs) in human cancer found that ncRNAs can regulate the metabolism of cancer cells and tumor-associated macrophages. NcRNAs include short ncRNAs, long ncRNAs (lncRNAs), and circular RNAs (circRNAs). The most common short ncRNAs are microRNAs, which regulate glucose, lipid, and amino acid metabolism in macrophages by acting on metabolism-related pathways and targeting metabolism-related enzymes and proteins, and are therefore involved in cancer progression. The role of lncRNAs and circRNAs in the metabolism of tumor-associated macrophages remains unclear. LncRNAs affect the glucose metabolism of macrophages, whereas their role in lipid and amino acid metabolism is not clear. CircRNAs regulate amino acid metabolism in macrophages. The roles of ncRNAs in energy metabolism and the underlying mechanisms need to be investigated further. Here, we summarize recent findings on the involvement of ncRNAs in metabolic reprogramming in tumor-associated macrophages, which affect the tumor microenvironment and play important roles in the development of cancer. Improving our understanding of the effects of ncRNAs on metabolic reprogramming of tumor-associated macrophages may facilitate the development of effective clinical therapies.
Collapse
|
22
|
Shi X, Wang J, Dai S, Qin L, Zhou J, Chen Y. Apolipoprotein C1 (APOC1): A Novel Diagnostic and Prognostic Biomarker for Cervical Cancer. Onco Targets Ther 2020; 13:12881-12891. [PMID: 33364782 PMCID: PMC7751697 DOI: 10.2147/ott.s280690] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Previous reports showed that APOC1 was associated with several cancers but the function of APOC1 in cervical cancer was unknown. This study aimed to investigate the clinical effect and function of APOC1 in cervical cancer. MATERIALS AND METHODS In this study, the relative expression of APOC1 in cervical cancer was detected by RT-qPCR. In order to determine the cell proliferation and migration and invading ability and apoptosis more accurately, we used CCK8 assay, Edu assay, wound healing assay, migration and invasion assay, flow cytometry assay, co-immunoprecipitation, proteomics and Western blot by silencing and overexpressing APOC1, respectively. The role of APOC1 on tumor progression was explored in vitro and vivo. RESULTS The relative expression of APOC1 in cervical cancer tissues was up-regulated (P<0.05). In cervical cancer cell lines, silencing of APOC1 restrained cell progression and EMT, while over-expression of APOC1 accelerated cell progression and EMT in vivo and vitro (P<0.05). CONCLUSION APOC1 acts as an oncogene in cervical cancers and knockdown of APOC1 inhibited cervical cancer cells growth in vitro and in vivo. There is a close relationship between the relative expression of APOC1 and clinical outcome in cervical cancer patients.
Collapse
Affiliation(s)
- Xiu Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
- Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Juan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Shouqian Dai
- Department of Emergency Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Lingyan Qin
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| |
Collapse
|
23
|
Lin K, Luo W, Yan J, Shen S, Shen Q, Wang J, Guan X, Wu G, Huang W, Liang G. TLR2 regulates angiotensin II-induced vascular remodeling and EndMT through NF-κB signaling. Aging (Albany NY) 2020; 13:2553-2574. [PMID: 33318302 PMCID: PMC7880316 DOI: 10.18632/aging.202290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
Excessive vascular remodeling has been shown in hypertensive patients. In experimental models of hypertensive vascular injury, such as angiotensin II (Ang II) challenged mice, toll like receptor 2 (TLR2) initiates inflammatory responses. More recently, studies have reported atypical endothelial to mesenchymal transition (EndMT) in vascular injuries and inflammatory conditions. Here, we aimed to investigate whether TLR2 mediates Ang II-induced vascular inflammation and initiates EndMT. In a mouse model of angiotensin II-induced hypertension, we show that aortas exhibit increased medial thickening, fibrosis, and features of EndMT. These alterations were not observed in TLR2 knockout mice in response to Ang II. TLR2 silencing in cultured endothelial cells confirmed the essential role of TLR2 in Ang II-induced inflammatory factor induction, and EndMT-associated phenotypic change. Mechanistically, we found Ang II activates nuclear factor-κB signaling, inducing pro-inflammatory cytokine production, and mediates EndMT in both cultured endothelial cells and in mice. These studies illustrate a novel role of TLR2 in regulating Ang II-induced deleterious vascular remodeling through the induction of EndMT. The studies also suggest that TLR2 may be targeted to alleviate hypertension-associated vascular injury.
Collapse
Affiliation(s)
- Ke Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.,Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jueqian Yan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Siyuan Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.,Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Qirui Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jun Wang
- Department of Cardiology, Wenzhou Central Hospital and Affiliated Dingli Clinical Institute, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Xinfu Guan
- Affiliated Cangnan Hospital, Wenzhou Medical University, Cangnan 325800, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.,Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.,Affiliated Cangnan Hospital, Wenzhou Medical University, Cangnan 325800, Zhejiang, China
| |
Collapse
|
24
|
MiR-126-5p promotes contractile switching of aortic smooth muscle cells by targeting VEPH1 and alleviates Ang II-induced abdominal aortic aneurysm in mice. J Transl Med 2020; 100:1564-1574. [PMID: 32612287 DOI: 10.1038/s41374-020-0454-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 12/25/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a potential lethal disease that is defined by an irreversible dilatation (>50%) of the aorta. During AAA expansion, the aortic wall is often remodeled, which is featured by extracellular matrix (ECM) degeneration, medial and adventitial inflammation, depletion and phenotypic switching of vascular smooth muscle cells (SMCs). Recent studies have suggested microRNAs as vital regulators for vascular SMC function. Our earlier work demonstrated an anti-AAA role of miR-126-5p in ApoE-/- mice infused with angiotensin (Ang) II. The present study aimed to further elucidate its role in AAA pathogenesis with a focus on aortic SMC phenotypic switching. Ventricular zone expressed PH domain containing 1 (VEPH1) was identified as a novel negative regulator for vascular SMC differentiation by our group, and its expression was negatively correlated to miR-126-5p in mouse abdominal aortas based on the present microarray data. In vivo, in addition attenuating Ang II infusion-induced aortic dilation and elastin degradation, miR-126-5p agomirs also significantly reduced the expression of VEPH1. In vitro, to induce synthetic transition of human aortic smooth muscle cells (hAoSMCs), cells were stimulated with 1 μM Ang II for 24 h. Ectopic overexpression of miR-126-5p restored the differentiation of hAoSMCs-the expression of contractile/differentiated SMC markers, MYH11, and α-SMA, increased, whilst that of synthetic/dedifferentiated SMC markers, PCNA and Vimentin, decreased. Both mus and homo VEPH1 genes were validated as direct targets for miR-126-5p. VEPH1 re-expression impaired miR-126-5p-induced differentiation of hAoSMCs. In addition, Ang II-induced upregulation in matrix metalloproteinase (MMP)-9 and MMP2, two key proteases responsible for ECM degradation, in mouse aortas and hAoSMCs was reduced by miR-126-5p overexpression as well. Collectively, these results reveal an important, but previously unexplored, role of miR-126-5p in inhibiting AAA development-associated aortic SMC dedifferentiation.
Collapse
|
25
|
Xin T, Lu C, Zhang J, Wen J, Yan S, Li C, Zhang F, Zhang J. Oxidized LDL Disrupts Metabolism and Inhibits Macrophage Survival by Activating a miR-9/Drp1/Mitochondrial Fission Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8848930. [PMID: 33204400 PMCID: PMC7655251 DOI: 10.1155/2020/8848930] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/04/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022]
Abstract
Mitochondrial dysfunction is associated with macrophage damage, but the role of mitochondrial fission in macrophage cholesterol metabolism is not fully understood. In this study, we explored the influences of miR-9 and mitochondrial fission on macrophage viability and cholesterol metabolism. Macrophages were incubated with oxidized low-density lipoprotein (ox-LDL) in vitro, after which mitochondrial fission, cell viability, and cholesterol metabolism were examined using qPCR, ELISAs, and immunofluorescence. ox-LDL treatment significantly increased Drp1-associated mitochondrial fission. Transfection of Drp1 siRNA significantly reduced cell death, attenuated oxidative stress, and inhibited inflammatory responses in ox-LDL-treated macrophages. Interestingly, inhibition of Drp1-related mitochondrial fission also improved cholesterol metabolism by balancing the transcription of cholesterol influx/efflux enzymes. We also found that miR-9 was downregulated in ox-LDL-treated macrophages, and administration of a miR-9 mimic decreased Drp1 transcription and mitochondrial fission, as well as its effects. These results indicate that signaling via the novel miR-9/Drp1/mitochondrial fission axis is a key determinant of macrophage viability and cholesterol metabolism.
Collapse
Affiliation(s)
- Ting Xin
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Chengzhi Lu
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Jing Zhang
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Jiaxin Wen
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Shuangbin Yan
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Chao Li
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Feng Zhang
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Jin Zhang
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| |
Collapse
|
26
|
Golledge J, Krishna SM, Wang Y. Mouse models for abdominal aortic aneurysm. Br J Pharmacol 2020; 179:792-810. [PMID: 32914434 DOI: 10.1111/bph.15260] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/25/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) rupture is estimated to cause 200,000 deaths each year. Currently, the only treatment for AAA is surgical repair; however, this is only indicated for large asymptomatic, symptomatic or ruptured aneurysms, is not always durable, and is associated with a risk of serious perioperative complications. As a result, patients with small asymptomatic aneurysms or who are otherwise unfit for surgery are treated conservatively, but up to 70% of small aneurysms continue to grow, increasing the risk of rupture. There is thus an urgent need to develop drug therapies effective at slowing AAA growth. This review describes the commonly used mouse models for AAA. Recent research in these models highlights key roles for pathways involved in inflammation and cell turnover in AAA pathogenesis. There is also evidence for long non-coding RNAs and thrombosis in aneurysm pathology. Further well-designed research in clinically relevant models is expected to be translated into effective AAA drugs.
Collapse
Affiliation(s)
- Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia.,The Department of Vascular and Endovascular Surgery, The Townsville University Hospital, Townsville, Queensland, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Smriti Murali Krishna
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia.,The Department of Vascular and Endovascular Surgery, The Townsville University Hospital, Townsville, Queensland, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Yutang Wang
- Discipline of Life Sciences, School of Health and Life Sciences, Federation University Australia, Ballarat, Victoria, Australia
| |
Collapse
|