1
|
Mogi M, Kukida M. Factors associated with blood pressure regulation in vascular smooth muscle cells. Hypertens Res 2025; 48:1684-1686. [PMID: 40055496 DOI: 10.1038/s41440-025-02170-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 02/16/2025] [Indexed: 03/12/2025]
Affiliation(s)
- Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Ehime, Japan.
| | - Masayoshi Kukida
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
2
|
Shilenok V, Kobzeva K, Bushueva O. "SERBP1 (Hero45) is a Novel Link with Ischemic Heart Disease Risk: Associations with Coronary Arteries Occlusion, Blood Coagulation and Lipid Profile". Cell Biochem Biophys 2025:10.1007/s12013-025-01736-z. [PMID: 40175693 DOI: 10.1007/s12013-025-01736-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Ischemic heart disease (IHD), stemming from coronary atherosclerosis, involves pathological processes in which chaperone proteins play an essential role. SERBP1 (Hero45), an RNA-binding protein, has recently been ascribed to the newly discovered class of Hero proteins with chaperone-like activity, making it particularly relevant in atherosclerosis-related diseases. In this study, 2164 subjects (836 IHD patients and 1328 controls) were genotyped for five common single nucleotide polymorphisms (SNPs) of SERBP1 using probe-based PCR. Here, we report that SNPs of SERBP1 are associated with reduced risk of left coronary artery atherosclerosis: rs4655707 (effect allele [EA] T, OR = 0.63, 95% CI 0.43-0.93, p = 0.02), (EA C, OR = 0.63, 95% CI 0.42-0.95, p = 0.02), rs12561767 (EA G, OR = 0.65, 95% CI 0.45-0.96, p = 0.03), rs6702742 (EA A, OR = 0.63, 95% CI 0.43-0.94, p = 0.02). Additionally, SERBP1 loci are linked to lower coronary artery stenosis (rs1058074), improved blood lipid profiles (rs1058074), and favorable blood coagulation parameters (rs4655707, rs6702742, rs1058074, rs12561767). Together, our study is the first to provide evidence that SERBP1 is involved in lipid metabolism and coagulation regulation, modulating IHD risk.
Collapse
Affiliation(s)
- Vladislav Shilenok
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
- Cardiology Department with the intensive care unit, Kursk Emergency Hospital, Kursk, Russia
| | - Ksenia Kobzeva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
| | - Olga Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia.
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russia.
| |
Collapse
|
3
|
Chen Q, Rong H, Zhang L, Wang Y, Bian Q, Zheng J. KLF2 Orchestrates Pathological Progression of Infantile Hemangioma through Hemangioma Stem Cell Fate Decisions. J Invest Dermatol 2024; 144:1850-1864.e9. [PMID: 38382868 DOI: 10.1016/j.jid.2024.01.029] [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: 08/10/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
Infantile hemangioma (IH) is the most prevalent vascular tumor during infancy, characterized by a rapid proliferation phase of disorganized blood vessels and spontaneous involution. IH possibly arises from a special type of multipotent stem cells called hemangioma stem cells (HemSCs), which could differentiate into endothelial cells, pericytes, and adipocytes. However, the underlying mechanisms that regulate the cell fate determination of HemSCs remain elusive. In this study, we unveil KLF2 as a candidate transcription factor involved in the control of HemSCs differentiation. KLF2 exhibits high expression in endothelial cells in proliferating IH but diminishes in adipocytes in involuting IH. Using a combination of in vitro culture of patient-derived HemSCs and HemSCs implantation mouse models, we show that KLF2 governs the proliferation, apoptosis, and cell cycle progression of HemSCs. Importantly, KLF2 acts as a crucial determinant of HemSC fate, directing their differentiation toward endothelial cells while inhibiting adipogenesis. Knockdown of KLF2 induces a proadipogenic transcriptome in HemSCs, leading to impaired blood vessel formation and accelerated adipocyte differentiation. Collectively, our findings highlight KLF2 as a critical regulator controlling the progression and involution of IH by modulating HemSC fate decisions.
Collapse
Affiliation(s)
- Qiming Chen
- Department of Oromaxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hao Rong
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Zhang
- Department of Oromaxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yanan Wang
- Department of Oromaxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Qian Bian
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiawei Zheng
- Department of Oromaxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China.
| |
Collapse
|
4
|
Fan J, Zhu T, Tian X, Liu S, Zhang SL. Exploration of ferroptosis and necroptosis-related genes and potential molecular mechanisms in psoriasis and atherosclerosis. Front Immunol 2024; 15:1372303. [PMID: 39072329 PMCID: PMC11272566 DOI: 10.3389/fimmu.2024.1372303] [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: 01/17/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
Objective Ferroptosis and necroptosis are two recently identified forms of non-apoptotic cell death. Their dysregulation plays a critical role in the development and progression of Psoriasis (PsD) and Atherosclerosis (AS). This study explores shared Ferroptosis and necroptosis-related genes and elucidates their molecular mechanisms in PsD and AS through the analysis of public databases. Methods Data sets for PsD (GSE30999) and AS (GSE28829) were retrieved from the GEO database. Differential gene expression (DEG) and weighted gene co-expression network analysis (WGCNA) were performed. Machine learning algorithms identified candidate biomarkers, whose diagnostic values were assessed using Receiver Operating Characteristic (ROC) curve analysis. Additionally, the expression levels of these biomarkers in cell models of AS and PsD were quantitatively measured using Western Blot (WB) and real-time quantitative PCR (RT-qPCR). Furthermore, CIBERSORT evaluated immune cell infiltration in PsD and AS tissues, highlighting the correlation between characteristic genes and immune cells. Predictive analysis for candidate drugs targeting characteristic genes was conducted using the DGIdb database, and an lncRNA-miRNA-mRNA network related to these genes was constructed. Results We identified 44 differentially expressed ferroptosis-related genes (DE-FRGs) and 30 differentially expressed necroptosis-related genes (DE-NRGs). GO and KEGG enrichment analyses revealed significant enrichment of these genes in immune-related and inflammatory pathways, especially in NOD-like receptor and TNF signaling pathways. Two ferroptosis-related genes (NAMPT, ZFP36) and eight necroptosis-related genes (C7, CARD6, CASP1, CTSD, HMOX1, NOD2, PYCARD, TNFRSF21) showed high sensitivity and specificity in ROC curve analysis. These findings were corroborated in external validation datasets and cell models. Immune infiltration analysis revealed increased levels of T cells gamma delta, Macrophages M0, and Macrophages M2 in PsD and AS samples. Additionally, we identified 43 drugs targeting 5 characteristic genes. Notably, the XIST-miR-93-5p-ZFP36/HMOX1 and NEAT1-miR-93-5p-ZFP36/HMOX1 pathways have been identified as promising RNA regulatory pathways in AS and PsD. Conclusion The two ferroptosis-related genes (NAMPT, ZFP36) and eight necroptosis-related genes (C7, CARD6, CASP1, CTSD, HMOX1, NOD2, PYCARD, TNFRSF21) are potential key biomarkers for PsD and AS. These genes significantly influence the pathogenesis of PsD and AS by modulating macrophage activity, participating in immune regulation, and mediating inflammatory responses.
Collapse
Affiliation(s)
- Jilin Fan
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tingting Zhu
- Department of Neurosurgery Ward 5, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaoling Tian
- Department of Neurosurgery Ward 5, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Sijia Liu
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Shi-Liang Zhang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
5
|
Zhang M, Zhang X, Niu J, Hua C, Liu P, Zhong G. Integrated analysis of single-cell RNA sequencing and bulk RNA data reveals gene regulatory networks and targets in dilated cardiomyopathy. Sci Rep 2024; 14:13942. [PMID: 38886541 PMCID: PMC11183045 DOI: 10.1038/s41598-024-64693-2] [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: 11/17/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a common cause of heart failure, thromboembolism, arrhythmias, and sudden cardiac death. The quality of life and long-term survival rates of patients with dilated DCM have greatly improved in recent decades. Nevertheless, the clinical prognosis for DCM patients remains unfavorable. The primary driving factors underlying the pathogenesis of DCM remain incompletely understood. The present study aimed to identify driving factors underlying the pathogenesis of DCM from the perspective of gene regulatory networks. Single-cell RNA sequencing data and bulk RNA data were obtained from the Gene Expression Omnibus (GEO) database. Differential gene analysis, single-cell genomics analysis, and functional enrichment analysis were conducted using R software. The construction of Gene Regulatory Networks was performed using Python. We used the pySCENIC method to analyze the single-cell data and identified 401 regulons. Through variance decomposition, we selected 19 regulons that showed significant responsiveness to DCM. Next, we employed the ssGSEA method to assess regulons in two bulk RNA datasets. Significant statistical differences were observed in 9 and 13 regulons in each dataset. By intersecting these differentiated regulons and identifying shared targets that appeared at least twice, we successfully pinpointed three differentially expressed targets across both datasets. In this study, we assessed and identified 19 gene regulatory networks that were responsive to the disease. Furthermore, we validated these networks using two bulk RNA datasets of DCM. The elucidation of dysregulated regulons and targets (CDKN1A, SAT1, ZFP36) enhances the molecular understanding of DCM, aiding in the development of tailored therapies for patients.
Collapse
Affiliation(s)
- Min Zhang
- Department of Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Xin Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiayin Niu
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Cuncun Hua
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Pengfei Liu
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guangzhen Zhong
- Department of Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China.
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Völkers M, Preiss T, Hentze MW. RNA-binding proteins in cardiovascular biology and disease: the beat goes on. Nat Rev Cardiol 2024; 21:361-378. [PMID: 38163813 DOI: 10.1038/s41569-023-00958-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
Cardiac development and function are becoming increasingly well understood from different angles, including signalling, transcriptional and epigenetic mechanisms. By contrast, the importance of the post-transcriptional landscape of cardiac biology largely remains to be uncovered, building on the foundation of a few existing paradigms. The discovery during the past decade of hundreds of additional RNA-binding proteins in mammalian cells and organs, including the heart, is expected to accelerate progress and has raised intriguing possibilities for better understanding the intricacies of cardiac development, metabolism and adaptive alterations. In this Review, we discuss the progress and new concepts on RNA-binding proteins and RNA biology and appraise them in the context of common cardiovascular clinical conditions, from cell and organ-wide perspectives. We also discuss how a better understanding of cardiac RNA-binding proteins can fill crucial knowledge gaps in cardiology and might pave the way to developing better treatments to reduce cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Mirko Völkers
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg and Mannheim, Germany
| | - Thomas Preiss
- Shine-Dalgarno Centre for RNA Innovation, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Matthias W Hentze
- European Molecular Biology Laboratory, Heidelberg, Germany.
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany.
| |
Collapse
|
7
|
Gibson Hughes TA, Dona MSI, Sobey CG, Pinto AR, Drummond GR, Vinh A, Jelinic M. Aortic Cellular Heterogeneity in Health and Disease: Novel Insights Into Aortic Diseases From Single-Cell RNA Transcriptomic Data Sets. Hypertension 2024; 81:738-751. [PMID: 38318714 DOI: 10.1161/hypertensionaha.123.20597] [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] [Indexed: 02/07/2024]
Abstract
Aortic diseases such as atherosclerosis, aortic aneurysms, and aortic stiffening are significant complications that can have significant impact on end-stage cardiovascular disease. With limited pharmacological therapeutic strategies that target the structural changes in the aorta, surgical intervention remains the only option for some patients with these diseases. Although there have been significant contributions to our understanding of the cellular architecture of the diseased aorta, particularly in the context of atherosclerosis, furthering our insight into the cellular drivers of disease is required. The major cell types of the aorta are well defined; however, the advent of single-cell RNA sequencing provides unrivaled insights into the cellular heterogeneity of each aortic cell type and the inferred biological processes associated with each cell in health and disease. This review discusses previous concepts that have now been enhanced with recent advances made by single-cell RNA sequencing with a focus on aortic cellular heterogeneity.
Collapse
Affiliation(s)
- Tayla A Gibson Hughes
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Malathi S I Dona
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., A.R.P.)
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Alexander R Pinto
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., A.R.P.)
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| |
Collapse
|
8
|
Saemann L, Wächter K, Gharpure N, Pohl S, Hoorn F, Korkmaz-Icöz S, Karck M, Veres G, Simm A, Szabó G. HTK vs. HTK-N for Coronary Endothelial Protection during Hypothermic, Oxygenated Perfusion of Hearts Donated after Circulatory Death. Int J Mol Sci 2024; 25:2262. [PMID: 38396938 PMCID: PMC10889240 DOI: 10.3390/ijms25042262] [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: 12/22/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Protection of the coronary arteries during donor heart maintenance is pivotal to improve results and prevent the development of coronary allograft vasculopathy. The effect of hypothermic, oxygenated perfusion (HOP) with the traditional HTK and the novel HTK-N solution on the coronary microvasculature of donation-after-circulatory-death (DCD) hearts is known. However, the effect on the coronary macrovasculature is unknown. Thus, we maintained porcine DCD hearts by HOP with HTK or HTK-N for 4 h, followed by transplantation-equivalent reperfusion with blood for 2 h. Then, we removed the left anterior descending coronary artery (LAD) and compared the endothelial-dependent and -independent vasomotor function of both groups using bradykinin and sodium-nitroprusside (SNP). We also determined the transcriptome of LAD samples using microarrays. The endothelial-dependent relaxation was significantly better after HOP with HTK-N. The endothelial-independent relaxation was comparable between both groups. In total, 257 genes were expressed higher, and 668 genes were expressed lower in the HTK-N group. Upregulated genes/pathways were involved in endothelial and vascular smooth muscle cell preservation and heart development. Downregulated genes were related to ischemia/reperfusion injury, oxidative stress, mitochondrion organization, and immune reaction. The novel HTK-N solution preserves the endothelial function of DCD heart coronary arteries more effectively than traditional HTK.
Collapse
Affiliation(s)
- Lars Saemann
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Kristin Wächter
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
| | - Nitin Gharpure
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
| | - Sabine Pohl
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
| | - Fabio Hoorn
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Sevil Korkmaz-Icöz
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Matthias Karck
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Gábor Veres
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Andreas Simm
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
| | - Gábor Szabó
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
9
|
Schutt WR, Conde JN, Mladinich MC, Himmler GE, Mackow ER. ZIKV induction of tristetraprolin in endothelial and Sertoli cells post-transcriptionally inhibits IFNβ/λ expression and promotes ZIKV persistence. mBio 2023; 14:e0174223. [PMID: 37707056 PMCID: PMC10653947 DOI: 10.1128/mbio.01742-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 09/15/2023] Open
Abstract
IMPORTANCE Our findings define a novel role for ZIKV-induced TTP expression in regulating IFNβ/IFNλ production in primary hBMECs and Sertoli cells. These cells comprise key physiological barriers subverted by ZIKV to access brain and testicular compartments and serve as reservoirs for persistent replication and dissemination. We demonstrate for the first time that the ARE-binding protein TTP is virally induced and post-transcriptionally regulates IFNβ/IFNλ secretion. In ZIKV-infected hBMEC and Sertoli cells, TTP knockout increased IFNβ/IFNλ secretion, while TTP expression blocked IFNβ/IFNλ secretion. The TTP-directed blockade of IFN secretion permits ZIKV spread and persistence in hBMECs and Sertoli cells and may similarly augment ZIKV spread across IFNλ-protected placental barriers. Our work highlights the importance of post-transcriptional ZIKV regulation of IFN expression and secretion in cells that regulate viral access to protected compartments and defines a novel mechanism of ZIKV-regulated IFN responses which may facilitate neurovirulence and sexual transmission.
Collapse
Affiliation(s)
- William R. Schutt
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
| | - Jonas N. Conde
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
- Molecular and Cell Biology Program, Stony Brook University, Stony Brook, New York, USA
| | - Megan C. Mladinich
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
- Molecular and Cell Biology Program, Stony Brook University, Stony Brook, New York, USA
| | - Grace E. Himmler
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
- Molecular and Cell Biology Program, Stony Brook University, Stony Brook, New York, USA
| | - Erich R. Mackow
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Disease, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
10
|
Zheng Y, Wen S, Jiang S, He S, Qiao W, Liu Y, Yang W, Zhou J, Wang B, Li D, Lin J. CircRNA/lncRNA-miRNA-mRNA network and gene landscape in calcific aortic valve disease. BMC Genomics 2023; 24:419. [PMID: 37491214 PMCID: PMC10367311 DOI: 10.1186/s12864-023-09441-y] [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: 02/04/2023] [Accepted: 06/11/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a common valve disease with an increasing incidence, but no effective drugs as of yet. With the development of sequencing technology, non-coding RNAs have been found to play roles in many diseases as well as CAVD, but no circRNA/lncRNA-miRNA-mRNA interaction axis has been established. Moreover, valve interstitial cells (VICs) and valvular endothelial cells (VECs) play important roles in CAVD, and CAVD differed between leaflet phenotypes and genders. This work aims to explore the mechanism of circRNA/lncRNA-miRNA-mRNA network in CAVD, and perform subgroup analysis on the important characteristics of CAVD, such as key cells, leaflet phenotypes and genders. RESULTS We identified 158 differentially expressed circRNAs (DEcircRNAs), 397 DElncRNAs, 45 DEmiRNAs and 167 DEmRNAs, and constructed a hsa-circ-0073813/hsa-circ-0027587-hsa-miR-525-5p-SPP1/HMOX1/CD28 network in CAVD after qRT-PCR verification. Additionally, 17 differentially expressed genes (DEGs) in VICs, 9 DEGs in VECs, 7 DEGs between different leaflet phenotypes and 24 DEGs between different genders were identified. Enrichment analysis suggested the potentially important pathways in inflammation and fibro-calcification during the pathogenesis of CAVD, and immune cell patterns in CAVD suggest that M0 macrophages and memory B cells memory were significantly increased, and many genes in immune cells were also differently expressed. CONCLUSIONS The circRNA/lncRNA-miRNA-mRNA interaction axis constructed in this work and the DEGs identified between different characteristics of CAVD provide a direction for a deeper understanding of CAVD and provide possible diagnostic markers and treatment targets for CAVD in the future.
Collapse
Affiliation(s)
- Yuqi Zheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuyu Wen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shijiu Jiang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Cardiology, The First Affiliated Hospital, Shihezi University, Shihezi, 832000, China
| | - Shaolin He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenling Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jin Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Boyuan Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dazhu Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jibin Lin
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
11
|
Sachse M, Tual-Chalot S, Ciliberti G, Amponsah-Offeh M, Stamatelopoulos K, Gatsiou A, Stellos K. RNA-binding proteins in vascular inflammation and atherosclerosis. Atherosclerosis 2023; 374:55-73. [PMID: 36759270 DOI: 10.1016/j.atherosclerosis.2023.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/01/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the major cause of premature death and disability worldwide, even when patients with an established manifestation of atherosclerotic heart disease are optimally treated according to the clinical guidelines. Apart from the epigenetic control of transcription of the genetic information to messenger RNAs (mRNAs), gene expression is tightly controlled at the post-transcriptional level before the initiation of translation. Although mRNAs are traditionally perceived as the messenger molecules that bring genetic information from the nuclear DNA to the cytoplasmic ribosomes for protein synthesis, emerging evidence suggests that processes controlling RNA metabolism, driven by RNA-binding proteins (RBPs), affect cellular function in health and disease. Over the recent years, vascular endothelial cell, smooth muscle cell and immune cell RBPs have emerged as key co- or post-transcriptional regulators of several genes related to vascular inflammation and atherosclerosis. In this review, we provide an overview of cell-specific function of RNA-binding proteins involved in all stages of ASCVD and how this knowledge may be used for the development of novel precision medicine therapeutics.
Collapse
Affiliation(s)
- Marco Sachse
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Cardiovascular Surgery, University Heart Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Giorgia Ciliberti
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Michael Amponsah-Offeh
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Aikaterini Gatsiou
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Konstantinos Stellos
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany; Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany.
| |
Collapse
|
12
|
Li Z, Solomonidis EG, Berkeley B, Tang MNH, Stewart KR, Perez-Vicencio D, McCracken IR, Spiroski AM, Gray GA, Barton AK, Sellers SL, Riley PR, Baker AH, Brittan M. Multi-species meta-analysis identifies transcriptional signatures associated with cardiac endothelial responses in the ischaemic heart. Cardiovasc Res 2023; 119:136-154. [PMID: 36082978 PMCID: PMC10022865 DOI: 10.1093/cvr/cvac151] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/04/2022] [Accepted: 08/10/2022] [Indexed: 11/12/2022] Open
Abstract
AIM Myocardial infarction remains the leading cause of heart failure. The adult human heart lacks the capacity to undergo endogenous regeneration. New blood vessel growth is integral to regenerative medicine necessitating a comprehensive understanding of the pathways that regulate vascular regeneration. We sought to define the transcriptomic dynamics of coronary endothelial cells following ischaemic injuries in the developing and adult mouse and human heart and to identify new mechanistic insights and targets for cardiovascular regeneration. METHODS AND RESULTS We carried out a comprehensive meta-analysis of integrated single-cell RNA-sequencing data of coronary vascular endothelial cells from the developing and adult mouse and human heart spanning healthy and acute and chronic ischaemic cardiac disease. We identified species-conserved gene regulatory pathways aligned to endogenous neovascularization. We annotated injury-associated temporal shifts of the endothelial transcriptome and validated four genes: VEGF-C, KLF4, EGR1, and ZFP36. Moreover, we showed that ZFP36 regulates human coronary endothelial cell proliferation and defined that VEGF-C administration in vivo enhances clonal expansion of the cardiac vasculature post-myocardial infarction. Finally, we constructed a coronary endothelial cell meta-atlas, CrescENDO, to empower future in-depth research to target pathways associated with coronary neovascularization. CONCLUSION We present a high-resolution single-cell meta-atlas of healthy and injured coronary endothelial cells in the mouse and human heart, revealing a suite of novel targets with great potential to promote vascular regeneration, and providing a rich resource for therapeutic development.
Collapse
Affiliation(s)
- Ziwen Li
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Emmanouil G Solomonidis
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Bronwyn Berkeley
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Michelle Nga Huen Tang
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Katherine Ross Stewart
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Daniel Perez-Vicencio
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ian R McCracken
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ana-Mishel Spiroski
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gillian A Gray
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Anna K Barton
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Stephanie L Sellers
- Division of Cardiology, Centre for Heart Lung Innovation, Providence Research, University of British Columbia, Vancouver, Canada
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | |
Collapse
|
13
|
RBP-RNA interactions in the control of autoimmunity and autoinflammation. Cell Res 2023; 33:97-115. [PMID: 36599968 PMCID: PMC9892603 DOI: 10.1038/s41422-022-00752-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/07/2022] [Indexed: 01/06/2023] Open
Abstract
Autoimmunity and autoinflammation arise from aberrant immunological and inflammatory responses toward self-components, contributing to various autoimmune diseases and autoinflammatory diseases. RNA-binding proteins (RBPs) are essential for immune cell development and function, mainly via exerting post-transcriptional regulation of RNA metabolism and function. Functional dysregulation of RBPs and abnormities in RNA metabolism are closely associated with multiple autoimmune or autoinflammatory disorders. Distinct RBPs play critical roles in aberrant autoreactive inflammatory responses via orchestrating a complex regulatory network consisting of DNAs, RNAs and proteins within immune cells. In-depth characterizations of RBP-RNA interactomes during autoimmunity and autoinflammation will lead to a better understanding of autoimmune pathogenesis and facilitate the development of effective therapeutic strategies. In this review, we summarize and discuss the functions of RBP-RNA interactions in controlling aberrant autoimmune inflammation and their potential as biomarkers and therapeutic targets.
Collapse
|
14
|
Kang D, Baek Y, Lee JS. Mechanisms of RNA and Protein Quality Control and Their Roles in Cellular Senescence and Age-Related Diseases. Cells 2022; 11:cells11244062. [PMID: 36552825 PMCID: PMC9777292 DOI: 10.3390/cells11244062] [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: 11/10/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Cellular senescence, a hallmark of aging, is defined as irreversible cell cycle arrest in response to various stimuli. It plays both beneficial and detrimental roles in cellular homeostasis and diseases. Quality control (QC) is important for the proper maintenance of cellular homeostasis. The QC machineries regulate the integrity of RNA and protein by repairing or degrading them, and are dysregulated during cellular senescence. QC dysfunction also contributes to multiple age-related diseases, including cancers and neurodegenerative, muscle, and cardiovascular diseases. In this review, we describe the characters of cellular senescence, discuss the major mechanisms of RNA and protein QC in cellular senescence and aging, and comprehensively describe the involvement of these QC machineries in age-related diseases. There are many open questions regarding RNA and protein QC in cellular senescence and aging. We believe that a better understanding of these topics could propel the development of new strategies for addressing age-related diseases.
Collapse
Affiliation(s)
- Donghee Kang
- Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Yurim Baek
- Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Jae-Seon Lee
- Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
- Correspondence: ; Tel.: +82-32-860-9832; Fax: +82-32-885-8302
| |
Collapse
|
15
|
Plasmodium falciparum and TNF-α Differentially Regulate Inflammatory and Barrier Integrity Pathways in Human Brain Endothelial Cells. mBio 2022; 13:e0174622. [PMID: 36036514 PMCID: PMC9601155 DOI: 10.1128/mbio.01746-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cerebral malaria is a severe complication of Plasmodium falciparum infection characterized by the loss of blood-brain barrier (BBB) integrity, which is associated with brain swelling and mortality in patients. P. falciparum-infected red blood cells and inflammatory cytokines, like tumor necrosis factor alpha (TNF-α), have been implicated in the development of cerebral malaria, but it is still unclear how they contribute to the loss of BBB integrity. Here, a combination of transcriptomic analysis and cellular assays detecting changes in barrier integrity and endothelial activation were used to distinguish between the effects of P. falciparum and TNF-α on a human brain microvascular endothelial cell (HBMEC) line and in primary human brain microvascular endothelial cells. We observed that while TNF-α induced high levels of endothelial activation, it only caused a small increase in HBMEC permeability. Conversely, P. falciparum-infected red blood cells (iRBCs) led to a strong increase in HBMEC permeability that was not mediated by cell death. Distinct transcriptomic profiles of TNF-α and P. falciparum in HBMECs confirm the differential effects of these stimuli, with the parasite preferentially inducing an endoplasmic reticulum stress response. Our results establish that there are fundamental differences in the responses induced by TNF-α and P. falciparum on brain endothelial cells and suggest that parasite-induced signaling is a major component driving the disruption of the BBB during cerebral malaria, proposing a potential target for much needed therapeutics.
Collapse
|
16
|
Cornelius VA, Naderi-Meshkin H, Kelaini S, Margariti A. RNA-Binding Proteins: Emerging Therapeutics for Vascular Dysfunction. Cells 2022; 11:2494. [PMID: 36010571 PMCID: PMC9407011 DOI: 10.3390/cells11162494] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 12/02/2022] Open
Abstract
Vascular diseases account for a significant number of deaths worldwide, with cardiovascular diseases remaining the leading cause of mortality. This ongoing, ever-increasing burden has made the need for an effective treatment strategy a global priority. Recent advances in regenerative medicine, largely the derivation and use of induced pluripotent stem cell (iPSC) technologies as disease models, have provided powerful tools to study the different cell types that comprise the vascular system, allowing for a greater understanding of the molecular mechanisms behind vascular health. iPSC disease models consequently offer an exciting strategy to deepen our understanding of disease as well as develop new therapeutic avenues with clinical translation. Both transcriptional and post-transcriptional mechanisms are widely accepted to have fundamental roles in orchestrating responses to vascular damage. Recently, iPSC technologies have increased our understanding of RNA-binding proteins (RBPs) in controlling gene expression and cellular functions, providing an insight into the onset and progression of vascular dysfunction. Revelations of such roles within vascular disease states have therefore allowed for a greater clarification of disease mechanisms, aiding the development of novel therapeutic interventions. Here, we discuss newly discovered roles of RBPs within the cardio-vasculature aided by iPSC technologies, as well as examine their therapeutic potential, with a particular focus on the Quaking family of isoforms.
Collapse
Affiliation(s)
| | | | | | - Andriana Margariti
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| |
Collapse
|
17
|
Lu F, Hu F, Qiu B, Zou H, Xu J. Identification of novel biomarkers in septic cardiomyopathy via integrated bioinformatics analysis and experimental validation. Front Genet 2022; 13:929293. [PMID: 35957694 PMCID: PMC9358039 DOI: 10.3389/fgene.2022.929293] [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: 04/26/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
Purpose: Septic cardiomyopathy (SCM) is an important world public health problem with high morbidity and mortality. It is necessary to identify SCM biomarkers at the genetic level to identify new therapeutic targets and strategies. Method: DEGs in SCM were identified by comprehensive bioinformatics analysis of microarray datasets (GSE53007 and GSE79962) downloaded from the GEO database. Subsequently, bioinformatics analysis was used to conduct an in-depth exploration of DEGs, including GO and KEGG pathway enrichment analysis, PPI network construction, and key gene identification. The top ten Hub genes were identified, and then the SCM model was constructed by treating HL-1 cells and AC16 cells with LPS, and these top ten Hub genes were examined using qPCR. Result: STAT3, SOCS3, CCL2, IL1R2, JUNB, S100A9, OSMR, ZFP36, and HAMP were significantly elevated in the established SCM cells model. Conclusion: After bioinformatics analysis and experimental verification, it was demonstrated that STAT3, SOCS3, CCL2, IL1R2, JUNB, S100A9, OSMR, ZFP36, and HAMP might play important roles in SCM.
Collapse
Affiliation(s)
- Feng Lu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Baiquan Qiu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongpeng Zou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianjun Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Jianjun Xu,
| |
Collapse
|
18
|
Wu Y, Chen H, Li L, Zhang L, Dai K, Wen T, Peng J, Peng X, Zheng Z, Jiang T, Xiong W. Construction of Novel Gene Signature-Based Predictive Model for the Diagnosis of Acute Myocardial Infarction by Combining Random Forest With Artificial Neural Network. Front Cardiovasc Med 2022; 9:876543. [PMID: 35694667 PMCID: PMC9174464 DOI: 10.3389/fcvm.2022.876543] [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: 02/15/2022] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
Background Acute myocardial infarction (AMI) is one of the most common causes of mortality around the world. Early diagnosis of AMI contributes to improving prognosis. In our study, we aimed to construct a novel predictive model for the diagnosis of AMI using an artificial neural network (ANN), and we verified its diagnostic value via constructing the receiver operating characteristic (ROC). Methods We downloaded three publicly available datasets (training sets GSE48060, GSE60993, and GSE66360) from Gene Expression Omnibus (GEO) database, and differentially expressed genes (DEGs) were identified between 87 AMI and 78 control samples. We applied the random forest (RF) and ANN algorithms to further identify novel gene signatures and construct a model to predict the possibility of AMI. Besides, the diagnostic value of our model was further validated in the validation sets GSE61144 (7 AMI patients and 10 controls), GSE34198 (49 AMI patients and 48 controls), and GSE97320 (3 AMI patients and 3 controls). Results A total of 71 DEGs were identified, of which 68 were upregulated and 3 were downregulated. Firstly, 11 key genes in 71 DEGs were screened with RF classifier for the classification of AMI and control samples. Then, we calculated the weight of each key gene using ANN. Furthermore, the diagnostic model was constructed and named neuralAMI, with significant predictive power (area under the curve [AUC] = 0.980). Finally, our model was validated with the independent datasets GSE61144 (AUC = 0.900), GSE34198 (AUC = 0.882), and GSE97320 (AUC = 1.00). Conclusion Machine learning was used to develop a reliable predictive model for the diagnosis of AMI. The results of our study provide potential gene biomarkers for early disease screening.
Collapse
Affiliation(s)
- Yanze Wu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hui Chen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lei Li
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liuping Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kai Dai
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tong Wen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingtian Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zeqi Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Jiang
- Department of Hospital Infection Control, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Ting Jiang,
| | - Wenjun Xiong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Wenjun Xiong,
| |
Collapse
|
19
|
In heart failure reactivation of RNA-binding proteins is associated with the expression of 1,523 fetal-specific isoforms. PLoS Comput Biol 2022; 18:e1009918. [PMID: 35226669 PMCID: PMC8912908 DOI: 10.1371/journal.pcbi.1009918] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 03/10/2022] [Accepted: 02/10/2022] [Indexed: 01/03/2023] Open
Abstract
Reactivation of fetal-specific genes and isoforms occurs during heart failure. However, the underlying molecular mechanisms and the extent to which the fetal program switch occurs remains unclear. Limitations hindering transcriptome-wide analyses of alternative splicing differences (i.e. isoform switching) in cardiovascular system (CVS) tissues between fetal, healthy adult and heart failure have included both cellular heterogeneity across bulk RNA-seq samples and limited availability of fetal tissue for research. To overcome these limitations, we have deconvoluted the cellular compositions of 996 RNA-seq samples representing heart failure, healthy adult (heart and arteria), and fetal-like (iPSC-derived cardiovascular progenitor cells) CVS tissues. Comparison of the expression profiles revealed that reactivation of fetal-specific RNA-binding proteins (RBPs), and the accompanied re-expression of 1,523 fetal-specific isoforms, contribute to the transcriptome differences between heart failure and healthy adult heart. Of note, isoforms for 20 different RBPs were among those that reverted in heart failure to the fetal-like expression pattern. We determined that, compared with adult-specific isoforms, fetal-specific isoforms encode proteins that tend to have more functions, are more likely to harbor RBP binding sites, have canonical sequences at their splice sites, and contain typical upstream polypyrimidine tracts. Our study suggests that compared with healthy adult, fetal cardiac tissue requires stricter transcriptional regulation, and that during heart failure reversion to this stricter transcriptional regulation occurs. Furthermore, we provide a resource of cardiac developmental stage-specific and heart failure-associated genes and isoforms, which are largely unexplored and can be exploited to investigate novel therapeutics for heart failure. Heart failure is a chronic condition in which the heart does not pump enough blood. It has been shown that in heart failure, the adult heart reverts to a fetal-like metabolic state and oxygen consumption. Additionally, genes and isoforms that are expressed in the heart only during fetal development (i.e. not in the healthy adult heart) are turned on in heart failure. However, the underlying molecular mechanisms and the extent to which the switch to a fetal gene program occurs remains unclear. In this study, we initially characterized the differences between the fetal and adult heart transcriptomes (entire set of expressed genes and isoforms). We found that RNA binding proteins (RBPs), a family of genes that regulate multiple aspects of a transcript’s maturation, including transcription, splicing and post-transcriptional modifications, play a central role in the differences between fetal and adult heart tissues. We observed that many RBPs that are only expressed in the heart during fetal development become reactivated in heart failure, resulting in the expression of 1,523 fetal-specific isoforms. These findings suggest that reactivation of fetal-specific RBPs in heart failure drives a transcriptome-wide switch to expression of fetal-specific isoforms; and hence that RBPs could potentially serve as novel therapeutic targets.
Collapse
|
20
|
Yildirim Z, Baboo S, Hamid SM, Dogan AE, Tufanli O, Robichaud S, Emerton C, Diedrich JK, Vatandaslar H, Nikolos F, Gu Y, Iwawaki T, Tarling E, Ouimet M, Nelson DL, Yates JR, Walter P, Erbay E. Intercepting IRE1 kinase-FMRP signaling prevents atherosclerosis progression. EMBO Mol Med 2022; 14:e15344. [PMID: 35191199 PMCID: PMC8988208 DOI: 10.15252/emmm.202115344] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/15/2022] Open
Abstract
Fragile X Mental Retardation protein (FMRP), widely known for its role in hereditary intellectual disability, is an RNA‐binding protein (RBP) that controls translation of select mRNAs. We discovered that endoplasmic reticulum (ER) stress induces phosphorylation of FMRP on a site that is known to enhance translation inhibition of FMRP‐bound mRNAs. We show ER stress‐induced activation of Inositol requiring enzyme‐1 (IRE1), an ER‐resident stress‐sensing kinase/endoribonuclease, leads to FMRP phosphorylation and to suppression of macrophage cholesterol efflux and apoptotic cell clearance (efferocytosis). Conversely, FMRP deficiency and pharmacological inhibition of IRE1 kinase activity enhances cholesterol efflux and efferocytosis, reducing atherosclerosis in mice. Our results provide mechanistic insights into how ER stress‐induced IRE1 kinase activity contributes to macrophage cholesterol homeostasis and suggests IRE1 inhibition as a promising new way to counteract atherosclerosis.
Collapse
Affiliation(s)
- Zehra Yildirim
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Molecular Biology and Genetics, National Nanotechnology Center, Bilkent University, Ankara, Turkey
| | - Sabyasachi Baboo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Syed M Hamid
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asli E Dogan
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Molecular Biology and Genetics, National Nanotechnology Center, Bilkent University, Ankara, Turkey
| | - Ozlem Tufanli
- Lagone Medical Center, New York University, New York, NY, USA
| | - Sabrina Robichaud
- Department of Biochemistry, Microbiology and Immunology, Heart Institute, University of Ottawa, Ottawa, ON, Canada
| | - Christina Emerton
- Department of Biochemistry, Microbiology and Immunology, Heart Institute, University of Ottawa, Ottawa, ON, Canada
| | - Jolene K Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Hasan Vatandaslar
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH), Zürich, Switzerland
| | - Fotis Nikolos
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yanghong Gu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Takao Iwawaki
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Elizabeth Tarling
- Department of Biochemistry and Biophysics, Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA, USA
| | - Mireille Ouimet
- Department of Biochemistry, Microbiology and Immunology, Heart Institute, University of Ottawa, Ottawa, ON, Canada
| | - David L Nelson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Peter Walter
- Department of Biochemistry and Biophysics, Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA, USA
| | - Ebru Erbay
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
21
|
Brandt KJ, Burger F, Baptista D, Roth A, Fernandes da Silva R, Montecucco F, Mach F, Miteva K. Single-Cell Analysis Uncovers Osteoblast Factor Growth Differentiation Factor 10 as Mediator of Vascular Smooth Muscle Cell Phenotypic Modulation Associated with Plaque Rupture in Human Carotid Artery Disease. Int J Mol Sci 2022; 23:1796. [PMID: 35163719 PMCID: PMC8836240 DOI: 10.3390/ijms23031796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Vascular smooth muscle cells (VSMCs) undergo a complex phenotypic switch in response to atherosclerosis environmental triggers, contributing to atherosclerosis disease progression. However, the complex heterogeneity of VSMCs and how VSMC dedifferentiation affects human carotid artery disease (CAD) risk has not been clearly established. (2) Method: A single-cell RNA sequencing analysis of CD45- cells derived from the atherosclerotic aorta of Apolipoprotein E-deficient (Apoe-/-) mice on a normal cholesterol diet (NCD) or a high cholesterol diet (HCD), respecting the site-specific predisposition to atherosclerosis was performed. Growth Differentiation Factor 10 (GDF10) role in VSMCs phenotypic switch was investigated via flow cytometry, immunofluorescence in human atherosclerotic plaques. (3) Results: scRNAseq analysis revealed the transcriptomic profile of seven clusters, five of which showed disease-relevant gene signature of VSMC macrophagic calcific phenotype, VSMC mesenchymal chondrogenic phenotype, VSMC inflammatory and fibro-phenotype and VSMC inflammatory phenotype. Osteoblast factor GDF10 involved in ossification and osteoblast differentiation emerged as a hallmark of VSMCs undergoing phenotypic switch. Under hypercholesteremia, GDF10 triggered VSMC osteogenic switch in vitro. The abundance of GDF10 expressing osteogenic-like VSMCs cells was linked to the occurrence of carotid artery disease (CAD) events. (4) Conclusions: Taken together, these results provide evidence about GDF10-mediated VSMC osteogenic switch, with a likely detrimental role in atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Karim J. Brandt
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Rafaela Fernandes da Silva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 6627, Brazil
- Swiss Institute for Translational and Entrepreneurial Medicine, Freiburgstrasse 3, 3010 Bern, Switzerland
| | - Fabrizio Montecucco
- Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy;
- First Clinic of Internal Medicine, Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Francois Mach
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine Specialized Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, CH-1211 Geneva 4, Switzerland; (K.J.B.); (F.B.); (D.B.); (A.R.); (R.F.d.S.); (F.M.)
| |
Collapse
|
22
|
Ding Y, Yin R, Zhang S, Xiao Q, Zhao H, Pan X, Zhu X. The Combined Regulation of Long Non-coding RNA and RNA-Binding Proteins in Atherosclerosis. Front Cardiovasc Med 2021; 8:731958. [PMID: 34796209 PMCID: PMC8592911 DOI: 10.3389/fcvm.2021.731958] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/07/2021] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is a complex disease closely related to the function of endothelial cells (ECs), monocytes/macrophages, and vascular smooth muscle cells (VSMCs). Despite a good understanding of the pathogenesis of atherosclerosis, the underlying molecular mechanisms are still only poorly understood. Therefore, atherosclerosis continues to be an important clinical issue worthy of further research. Recent evidence has shown that long non-coding RNAs (lncRNAs) and RNA-binding proteins (RBPs) can serve as important regulators of cellular function in atherosclerosis. Besides, several studies have shown that lncRNAs are partly dependent on the specific interaction with RBPs to exert their function. This review summarizes the important contributions of lncRNAs and RBPs in atherosclerosis and provides novel and comprehensible interaction models of lncRNAs and RBPs.
Collapse
Affiliation(s)
- Yuanyuan Ding
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruihua Yin
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shuai Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Xiao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongqin Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
23
|
lv L, Qin T, Huang Q, Jiang H, Chen F, Long F, Ren L, Liu J, Xie Y, Zeng M. Targeting Tristetraprolin Expression or Functional Activity Regulates Inflammatory Response Induced by MSU Crystals. Front Immunol 2021; 12:675534. [PMID: 34335573 PMCID: PMC8322984 DOI: 10.3389/fimmu.2021.675534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
The RNA-binding protein tristetraprolin (TTP) is an anti-inflammatory factor that prompts the mRNA decay of target mRNAs and is involved in inflammatory diseases such as rheumatoid arthritis (RA). TTP is regulated by phosphorylation, and protein phosphatase 2A (PP2A) can dephosphorylate TTP to activate its mRNA-degrading function. Some small molecules can enhance PP2A activation. Short interfering RNA (siRNA) targeting TTP expression or PP2A agonist (Arctigenin) was administered to monosodium urate (MSU) crystal-induced J774A.1 cells, and the expression of inflammatory related genes was detected by RT-PCR and Western blot assays. The effects of Arctigenin in mouse models of acute inflammation induced by MSU crystals, including peritonitis and arthritis, were evaluated. The data indicated that TTP expression levels and endogenous PP2A activity were increased in MSU-crystal treated J774A.1 cells. TTP knockdown exacerbated inflammation-related genes expression and NLRP3 inflammasome activation. However, PP2A agonist treatment (Arctigenin) suppressed MSU crystal-induced inflammation in J774A.1 cells. Arctigenin also relieved mitochondrial reactive oxygen species (mtROS) production and improved lysosomal membrane permeability in MSU crystal-treated J774A.1 cells. Moreover, TTP knockdown reversed the anti-inflammatory and antioxidant effects of Arctigenin. Oral administration of Arctigenin significantly alleviated foot pad swelling, the number of inflammatory cells in peritoneal lavage fluids and the production of IL-1β in the mouse model of inflammation induced by MSU crystals. Collectively, these data imply that targeting TTP expression or functional activity may provide a potential therapeutic strategy for inflammation caused by MSU crystals.
Collapse
Affiliation(s)
- Linxi lv
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Biology Group of Preclinical School of North SiChuan Medical College, Nanchong, China
| | - Ting Qin
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qiushi Huang
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Biology Group of Preclinical School of North SiChuan Medical College, Nanchong, China
| | - Hui Jiang
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Biology Group of Preclinical School of North SiChuan Medical College, Nanchong, China
| | - Feng Chen
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Biology Group of Preclinical School of North SiChuan Medical College, Nanchong, China
| | - Fan Long
- Clinical Lab of The Fifth People’s Hospital of Nanchong City, Nanchong, China
| | - Long Ren
- Clinical Lab of The Fifth People’s Hospital of Nanchong City, Nanchong, China
| | - Jianpin Liu
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yongen Xie
- Biology Group of Preclinical School of North SiChuan Medical College, Nanchong, China
| | - Mei Zeng
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Biology Group of Preclinical School of North SiChuan Medical College, Nanchong, China
- Clinical Lab of The Fifth People’s Hospital of Nanchong City, Nanchong, China
- Medical Imaging Key Laboratory of Sichuan, North SiChuan Medical College, Nanchong, China
- Academician (Expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
24
|
Cornelissen A, Guo L, Fernandez R, Kelly MC, Janifer C, Kuntz S, Sakamoto A, Jinnouchi H, Sato Y, Paek KH, Kolodgie FD, Romero ME, Surve D, Virmani R, Finn AV. Endothelial Recovery in Bare Metal Stents and Drug-Eluting Stents on a Single-Cell Level. Arterioscler Thromb Vasc Biol 2021; 41:2277-2292. [PMID: 34162228 DOI: 10.1161/atvbaha.121.316472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anne Cornelissen
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.).,Department of Cardiology, University Hospital RWTH Aachen, Germany (A.C.)
| | - Liang Guo
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Raquel Fernandez
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Michael C Kelly
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD (M.C.K.)
| | - Christine Janifer
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Salome Kuntz
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Atsushi Sakamoto
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Hiroyuki Jinnouchi
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Yu Sato
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Ka Hyun Paek
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Frank D Kolodgie
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Maria E Romero
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Dipti Surve
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Renu Virmani
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Aloke V Finn
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.).,University of Maryland, School of Medicine, Baltimore (A.V.F.)
| |
Collapse
|
25
|
Tristetraprolin, Inflammation, and Metabolic Syndrome in Arab Adults: A Case Control Study. BIOLOGY 2021; 10:biology10060550. [PMID: 34207463 PMCID: PMC8235193 DOI: 10.3390/biology10060550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 01/06/2023]
Abstract
Simple Summary Metabolic syndrome (MetS) is a common disorder characterized as a low-grade chronic inflammatory state. The association of tristetraprolin (TTP), a novel anti-inflammatory protein, and MetS remains to be explored. We evaluated circulating TTP in a group of adult males and females with and without MetS. Serum levels of TTP were higher in the MetS group than in controls. In all subjects, serum TTP was also correlated with MetS components (e.g., glucose, lipids, and obesity indices). These findings suggest that TTP may be a promising biomarker for MetS. Abstract Tristetraprolin (TTP) is an mRNA binding protein suggested to have a substantial role in regulating the mRNA expression of numerous inflammatory factors, but data on TTP and its association with metabolic syndrome (MetS), a chronic low-grade inflammatory disorder, are scarce. We hypothesize that TTP may modulate MetS and its components. A total of 200 Saudi adults (aged 38.6 ± 8.3 years) were included in this cross-sectional study. Anthropometrics data were collected and fasting blood glucose taken for the assessment of glycemic, lipids and inflammatory markers using commercially available assays. The National Cholesterol Education Program Adult Treatment Panel (NCEP ATP III) criteria were used to define MetS. Results showed significantly higher levels of TTP in the MetS group than in controls [288.1 pg/mL vs. 150.9 pg/mL, p < 0.001]. Circulating TTP was significantly associated with tumor necrosis factor alpha [TNF-α, R = 0.30, p < 0.05], interleukin 1β [IL-1β, R = 0.41, p < 0.01] and C-reactive protein [CRP, R = 0.36, p < 0.01], adiponectin [R = 0.36, p < 0.05], insulin [R = 0.37, p < 0.05], and insulin resistance [HOMA-IR, R = 0.40, p < 0.05]. Receiver operating characteristics (ROC) suggest a potential use of TTP as diagnostic biomarker for MetS [AUC = 0.819, p < 0.001]. The findings suggest that TTP is associated with inflammation and glycemia, which may influence MetS. TTP is a promising diagnostic biomarker for MetS which can be confirmed in larger cohorts.
Collapse
|
26
|
Bertesi M, Fantini S, Alecci C, Lotti R, Martello A, Parenti S, Carretta C, Marconi A, Grande A, Pincelli C, Zanocco-Marani T. Promoter Methylation Leads to Decreased ZFP36 Expression and Deregulated NLRP3 Inflammasome Activation in Psoriatic Fibroblasts. Front Med (Lausanne) 2021; 7:579383. [PMID: 33585499 PMCID: PMC7874095 DOI: 10.3389/fmed.2020.579383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
The mRNA-destabilizing protein tristetraprolin (TTP), encoded by the ZFP36 gene, is known to be able to end inflammatory responses by directly targeting and destabilizing mRNAs encoding pro-inflammatory cytokines. We analyzed its role in psoriasis, a disease characterized by chronic inflammation. We observed that TTP is downregulated in fibroblasts deriving from psoriasis patients compared to those deriving from healthy individuals and that psoriatic fibroblasts exhibit abnormal inflammasome activity compared to their physiological counterpart. This phenomenon depends on TTP downregulation. In fact, following restoration, TTP is capable of directly targeting for degradation NLRP3 mRNA, thereby drastically decreasing inflammasome activation. Moreover, we provide evidence that ZFP36 undergoes methylation in psoriasis, by virtue of the presence of long stretches of CpG dinucleotides both in the promoter and the coding region. Besides confirming that a perturbation of TTP expression might underlie the pathogenesis of psoriasis, we suggest that deregulated inflammasome activity might play a role in the disease alongside deregulated cytokine expression.
Collapse
Affiliation(s)
- Matteo Bertesi
- Laboratory of Applied Biology, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sebastian Fantini
- Department of Life Sciences, Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy
| | - Claudia Alecci
- Laboratory of Applied Biology, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberta Lotti
- Laboratory of Cutaneous Biology, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Martello
- University College London, Institute of Ophthalmology London, London, United Kingdom
| | - Sandra Parenti
- Department of Life Sciences, Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Carretta
- Department of Life Sciences, Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Marconi
- Laboratory of Cutaneous Biology, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alexis Grande
- Laboratory of Applied Biology, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Pincelli
- Laboratory of Cutaneous Biology, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Tommaso Zanocco-Marani
- Laboratory of Applied Biology, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
27
|
Rappl P, Brüne B, Schmid T. Role of Tristetraprolin in the Resolution of Inflammation. BIOLOGY 2021; 10:biology10010066. [PMID: 33477783 PMCID: PMC7832405 DOI: 10.3390/biology10010066] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 12/18/2022]
Abstract
Simple Summary Chronic inflammatory diseases account for up to 60% of deaths worldwide and, thus, are considered a great threat for human health by the World Health Organization. Nevertheless, acute inflammatory reactions are an integral part of the host defense against invading pathogens or injuries. To avoid excessive damage due to the persistence of a highly reactive environment, inflammations need to resolve in a coordinate and timely manner, ensuring for the immunological normalization of the affected tissues. Since post-transcriptional regulatory mechanisms are essential for effective resolution, the present review discusses the key role of the RNA-binding and post-transcriptional regulatory protein tristetraprolin in establishing resolution of inflammation. Abstract Inflammation is a crucial part of immune responses towards invading pathogens or tissue damage. While inflammatory reactions are aimed at removing the triggering stimulus, it is important that these processes are terminated in a coordinate manner to prevent excessive tissue damage due to the highly reactive inflammatory environment. Initiation of inflammatory responses was proposed to be regulated predominantly at a transcriptional level, whereas post-transcriptional modes of regulation appear to be crucial for resolution of inflammation. The RNA-binding protein tristetraprolin (TTP) interacts with AU-rich elements in the 3′ untranslated region of mRNAs, recruits deadenylase complexes and thereby facilitates degradation of its targets. As TTP regulates the mRNA stability of numerous inflammatory mediators, it was put forward as a crucial post-transcriptional regulator of inflammation. Here, we summarize the current understanding of the function of TTP with a specific focus on its role in adding to resolution of inflammation.
Collapse
Affiliation(s)
- Peter Rappl
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (P.R.); (B.B.)
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (P.R.); (B.B.)
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular and Applied Ecology, 60596 Frankfurt, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (P.R.); (B.B.)
- Correspondence:
| |
Collapse
|
28
|
Li X, He X, Wang J, Wang D, Cong P, Zhu A, Chen W. The Regulation of Exosome-Derived miRNA on Heterogeneity of Macrophages in Atherosclerotic Plaques. Front Immunol 2020; 11:2175. [PMID: 33013913 PMCID: PMC7511579 DOI: 10.3389/fimmu.2020.02175] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/10/2020] [Indexed: 11/13/2022] Open
Abstract
Exosomes are nanosized vesicles secreted by most cells, which can deliver a variety of functional lipids, proteins, and RNAs into the target cells to participate in complex intercellular communications. Cells respond to certain physical, chemical, and biological stimuli by releasing exosomes. Exosomes are rich in small molecules of RNA, including miRNAs and mRNAs, which have been demonstrated to have certain functions in recipient cells. Recent studies on single-cell RNA sequences have revealed the transcription and the heterogeneity of macrophages in Ldlr-/-mice fed with a high-fat diet. Five macrophage populations were found in the atherosclerotic plaques. It is worth noting that these subset populations of macrophages seem to be endowed with different functions in lipid metabolism and catabolism. A total of 100 differentially expressed mRNAs were selected for these subset populations. Importantly, these macrophage populations were also present in human advanced atherosclerosis. To clarify the specific functions and the regulatory mechanism of these macrophage populations, we extracted exosome RNAs from the plasma of patients with chronic coronary artery disease (CAD) and performed RNA sequencing analysis. Compared with the healthy control, a total of 14 miRNAs were significantly expressed in these patients. A total of 5,248 potential mRNAs were predicted by the bioinformatics platform. Next, we determined the outcome of the intersection of these predicted mRNAs with 100 mRNAs expressed in the above-mentioned five macrophage populations. Based on the screening of miRNA-mRNA pairs, a co-expression network was drawn to find out the key RNAs. Three down-regulated miRNAs and five up-regulated mRNAs were selected for validation by real-time RT-PCR. The results showed that the expression of miR-4498 in plasma exosomes was lower than that in the healthy control, and the expressions of Ctss, Ccr2 and Trem2 mRNA in peripheral blood mononuclear cells isolated from CAD patients were higher. In order to clarify the regulatory mechanism, we established a co-culture system in vitro. Studies have shown that the uptake of exosomes from CAD patients can up-regulate the expression of Ctss, Trem2, and Ccr2 mRNA in THP-1 cells induced by lipopolysaccharide. Our findings revealed a unique relationship between the transcriptional signature and the phenotypic heterogeneity of macrophage in the atherosclerotic microenvironment.
Collapse
Affiliation(s)
- Ximing Li
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xinyong He
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Junyan Wang
- The First Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan Wang
- Department of Medical Science of Laboratory, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Peiwei Cong
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Aisong Zhu
- Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenna Chen
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China.,Department of Medical Science of Laboratory, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
29
|
Hu YX, Zhu RF, Qin YW, Zhao XX, Jing Q. Zfp36l1b protects angiogenesis through Notch1b/Dll4 and Vegfa regulation in zebrafish. Atherosclerosis 2020; 309:56-64. [PMID: 32882641 DOI: 10.1016/j.atherosclerosis.2020.07.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 06/17/2020] [Accepted: 07/23/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Angiogenesis is a key process for establishing functional vasculature during embryogenesis and involves different signaling mechanisms. The RNA binding protein Zfp36l1 was reported to be involved in various diseases in different species, including cardiovascular diseases. However, whether Zfp36l1b, one of the 2 paralogs of Zfp36l1 in zebrafish, works like mammalian Zfp36l1, and if the molecular mechanisms are different remains unclear. Here, we show that Zfp36l1b plays a crucial protective role in angiogenesis of zebrafish embryos. METHODS We used transparent transgenic and wild-type zebrafish larvae to dynamically investigate the early stage of angiogenesis with confocal in vivo, after the knockdown of Zfp36l1b by morpholinos (MOs). In situ hybridization and fluorescence-activated cell sorting were performed to detect Zfp36l1b expression. mRNA rescue and CRISPR/Cas9 knockdown, and luciferase reporter experiments were performed to further explore the role of Zfp36l1b in angiogenesis. RESULTS We found that knockdown of Zfp36l1b led to defected angiogenesis in intersomitic vessels and sub-intestinal veins (SIVs), which could be rescued by the addition of Zfp36l1b mRNA. Moreover, knockdown of Zfp36l1b suppressed Notch1b expression, while knockdown of Notch1b resulted in a partial relief of angiogenesis defects induced by Zfp36l1b down-regulation. Besides, Zfp36l1b knockdown alleviated the excessive branch of SIVs caused by Vegfa over-expression. CONCLUSIONS Our results show that Zfp36l1b is responsible for establishing normal vessel circuits by affecting the extension of endothelial tip cells filopodia and the proliferation of endothelial cells partly through Notch1b/Fll4 suppression and synergistic function with Vegfa.
Collapse
Affiliation(s)
- Yang-Xi Hu
- Department of Cardiology, Changhai Hospital, Shanghai, 200433, China
| | - Rong-Fang Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yong-Wen Qin
- Department of Cardiology, Changhai Hospital, Shanghai, 200433, China
| | - Xian-Xian Zhao
- Department of Cardiology, Changhai Hospital, Shanghai, 200433, China
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China; Department of Cardiology, Changhai Hospital, Shanghai, 200433, China.
| |
Collapse
|
30
|
Saaoud F, Wang J, Iwanowycz S, Wang Y, Altomare D, Shao Y, Liu J, Blackshear PJ, Lessner SM, Murphy EA, Wang H, Yang X, Fan D. Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice. Redox Biol 2020; 37:101609. [PMID: 32591281 PMCID: PMC7767740 DOI: 10.1016/j.redox.2020.101609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/30/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022] Open
Abstract
Tristetraprolin (TTP), an mRNA binding and decaying protein, plays a significant role in controlling inflammation by decaying mRNAs encoding inflammatory cytokines such as TNFalpha. We aimed to test a hypothesis that TTP in bone marrow (BM) cells regulates atherogenesis by modulating inflammation and lipid metabolism through the modulation of oxidative stress pathways by TTP target genes. In a BM transplantation study, lethally irradiated atherogenic LDLR-/- mice were reconstituted with BM cells from either wild type (TTP+/+) or TTP knockout (TTP-/-) mice, and fed a Western diet for 12 weeks. We made the following observations: (1) TTP-/- BM recipients display a significantly higher systemic and multi-organ inflammation than TTP+/+ BM recipients; (2) BM TTP deficiency modulates hepatic expression of genes, detected by microarray, involved in lipid metabolism, inflammatory responses, and oxidative stress; (3) TTP-/- BM derived macrophages increase production of mitochondrial reactive oxygen species (mtROS); (4) BM-TTP-/- mice display a significant reduction in serum VLDL/LDL levels, and attenuated hepatic steatosis compared to controls; and (5) Reduction of serum VLDL/LDL levels offsets the increased inflammation, resulting in no changes in atherosclerosis. These findings provide a novel mechanistic insight into the roles of TTP-mediated mRNA decay in bone marrow-derived cells in regulating systemic inflammation, oxidative stress, and liver VLDL/LDL biogenesis.
Collapse
Affiliation(s)
- Fatma Saaoud
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, 29209, USA; Centers for Inflammation, Translational & Clinical Lung Research, Departments of Microbiology and Immunology and Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 14190, USA
| | - Junfeng Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | - Stephen Iwanowycz
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | - Diego Altomare
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Ying Shao
- Centers for Inflammation, Translational & Clinical Lung Research, Departments of Microbiology and Immunology and Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 14190, USA
| | - Jianguo Liu
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Susan M Lessner
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | - E Angela Murphy
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29209, USA
| | - Hong Wang
- Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Departments of Microbiology and Immunology, and Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 14190, USA
| | - Xiaofeng Yang
- Centers for Inflammation, Translational & Clinical Lung Research, Departments of Microbiology and Immunology and Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 14190, USA; Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Departments of Microbiology and Immunology, and Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 14190, USA.
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, 29209, USA.
| |
Collapse
|
31
|
Zhu H, Li Y, Wang MX, Wang JH, Du WX, Zhou F. Analysis of cardiovascular disease-related NF-κB-regulated genes and microRNAs in TNFα-treated primary mouse vascular endothelial cells. J Zhejiang Univ Sci B 2020; 20:803-815. [PMID: 31489800 DOI: 10.1631/jzus.b1800631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Activated nuclear factor-κB (NF-κB) plays an important role in the development of cardiovascular disease (CVD) through its regulated genes and microRNAs (miRNAs). However, the gene regulation profile remains unclear. In this study, primary mouse vascular endothelial cells (pMVECs) were employed to detect CVD-related NF-κB-regulated genes and miRNAs. Genechip assay identified 77 NF-κB-regulated genes, including 45 upregulated and 32 downregulated genes, in tumor necrosis factor α (TNFα)-treated pMVECs. Ten of these genes were also found to be regulated by NF-κB in TNFα-treated HeLa cells. Quantitative real-time PCR (qRT-PCR) assay confirmed the up-regulation of Egr1, Tnf, and Btg2 by NF-κB in the TNFα-treated pMVECs. The functional annotation revealed that many NF-κB-regulated genes identified in pMVECs were clustered into classical NF-κB-involved biological processes. Genechip assay also identified 26 NF-κB-regulated miRNAs, of which 21 were upregulated and 5 downregulated, in the TNFα-treated pMVECs. Further analysis showed that nine of the identified genes are regulated by seven of these miRNAs. Finally, among the identified NF-κB-regulated genes and miRNAs, 5 genes and 12 miRNAs were associated with CVD by miRWalk and genetic association database analysis. Taken together, these findings show an intricate gene regulation network raised by NF-κB in TNFα-treated pMVECs. The network provides new insights for understanding the molecular mechanism underlying the progression of CVD.
Collapse
Affiliation(s)
- Hui Zhu
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| | - Yun Li
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| | - Mao-Xian Wang
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| | - Ju-Hong Wang
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| | - Wen-Xin Du
- Shandong Center for Drug and Food Evaluation & Certification, Jinan 250014, China
| | - Fei Zhou
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou 521041, China
| |
Collapse
|
32
|
CXCL4 is a driver of cytokine mRNA stability in monocyte-derived dendritic cells. Mol Immunol 2019; 114:524-534. [DOI: 10.1016/j.molimm.2019.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/16/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022]
|
33
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
34
|
Cardiovascular inflammation: RNA takes the lead. J Mol Cell Cardiol 2019; 129:247-256. [PMID: 30880251 DOI: 10.1016/j.yjmcc.2019.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/19/2022]
Abstract
Inflammation has recently gained tremendous attention as a key contributor in several chronic diseases. While physiological inflammation is essential to counter a wide variety of damaging stimuli and to improve wound healing, dysregulated inflammation such as in the myocardium and vasculature can promote cardiovascular diseases. Given the high severity, prevalence, and economic burden of these diseases, understanding the factors involved in the regulation of physiological inflammation is essential. Like other complex biological phenomena, RNA-based processes are emerging as major regulators of inflammatory responses. Among such processes are cis-regulatory elements in the mRNA of inflammatory genes, noncoding RNAs directing the production or localization of inflammatory cytokines/chemokines, or pathogenic RNA driving inflammatory responses. In this review, we describe several specific RNA-based molecular mechanisms by which physiological inflammation pertaining to cardiovascular diseases is regulated. These include the role of AU-rich element-containing mRNAs, long non-coding RNAs, microRNAs, and viral RNAs.
Collapse
|
35
|
Veerman K, Tardiveau C, Martins F, Coudert J, Girard JP. Single-Cell Analysis Reveals Heterogeneity of High Endothelial Venules and Different Regulation of Genes Controlling Lymphocyte Entry to Lymph Nodes. Cell Rep 2019; 26:3116-3131.e5. [PMID: 30865898 DOI: 10.1016/j.celrep.2019.02.042] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/25/2019] [Accepted: 02/11/2019] [Indexed: 12/15/2022] Open
Abstract
High-endothelial venules (HEVs) are specialized blood vessels allowing recirculation of naive lymphocytes through lymphoid organs. Here, using full-length, single-cell RNA sequencing, RNA fluorescence in situ hybridization (FISH), flow cytometry, and immunohistofluorescence, we reveal the heterogeneity of HEVs in adult mouse peripheral lymph nodes (PLNs) under conditions of homeostasis, antigenic stimulation, and after inhibition of lymphotoxin-β receptor (LTβR) signaling. We demonstrate that HEV endothelial cells are in an activated state during homeostasis, and we identify the genes characteristic of the differentiated HEV phenotype. We show that LTβR signaling regulates many HEV genes and pathways in resting PLNs and that immune stimulation induces a global and temporary inflammatory phenotype in HEVs without compromising their ability to recruit naive lymphocytes. Most importantly, we uncover differences in the regulation of genes controlling lymphocyte trafficking, Glycam1, Fut7, Gcnt1, Chst4, B3gnt3, and Ccl21a, that have implications for HEV function and regulation in health and disease.
Collapse
Affiliation(s)
- Krystle Veerman
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Claire Tardiveau
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Frédéric Martins
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), UMR1048, INSERM, UPS, Toulouse, France; Plateforme Genome et Transcriptome (GeT), Genopole Toulouse, Toulouse, France
| | - Juliette Coudert
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
36
|
Caracciolo V, Young J, Gonzales D, Ni Y, Flowers SJ, Summer R, Waldman SA, Kim JK, Jung DY, Noh HL, Kim T, Blackshear PJ, O'Connell D, Bauer RC, Kallen CB. Myeloid-specific deletion of Zfp36 protects against insulin resistance and fatty liver in diet-induced obese mice. Am J Physiol Endocrinol Metab 2018; 315:E676-E693. [PMID: 29509432 PMCID: PMC6230714 DOI: 10.1152/ajpendo.00224.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Obesity is associated with adipose tissue inflammation that contributes to insulin resistance. Zinc finger protein 36 (Zfp36) is an mRNA-binding protein that reduces inflammation by binding to cytokine transcripts and promoting their degradation. We hypothesized that myeloid-specific deficiency of Zfp36 would lead to increased adipose tissue inflammation and reduced insulin sensitivity in diet-induced obese mice. As expected, wild-type (Control) mice became obese and diabetic on a high-fat diet, and obese mice with myeloid-specific loss of Zfp36 [knockout (KO)] demonstrated increased adipose tissue and liver cytokine mRNA expression compared with Control mice. Unexpectedly, in glucose tolerance testing and hyperinsulinemic-euglycemic clamp studies, myeloid Zfp36 KO mice demonstrated improved insulin sensitivity compared with Control mice. Obese KO and Control mice had similar macrophage infiltration of the adipose depots and similar peripheral cytokine levels, but lean and obese KO mice demonstrated increased Kupffer cell (KC; the hepatic macrophage)-expressed Mac2 compared with lean Control mice. Insulin resistance in obese Control mice was associated with enhanced Zfp36 expression in KCs. Compared with Control mice, KO mice demonstrated increased hepatic mRNA expression of a multitude of classical (M1) inflammatory cytokines/chemokines, and this M1-inflammatory hepatic milieu was associated with enhanced nuclear localization of IKKβ and the p65 subunit of NF-κB. Our data confirm the important role of innate immune cells in regulating hepatic insulin sensitivity and lipid metabolism, challenge-prevailing models in which M1 inflammatory responses predict insulin resistance, and indicate that myeloid-expressed Zfp36 modulates the response to insulin in mice.
Collapse
Affiliation(s)
- Valentina Caracciolo
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Jeanette Young
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Donna Gonzales
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Yingchun Ni
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Stephen J Flowers
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ross Summer
- Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Dae Young Jung
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Hye Lim Noh
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Taekyoon Kim
- Program in Molecular Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences , Research Triangle Park, North Carolina
| | - Danielle O'Connell
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Robert C Bauer
- Department of Medicine, Columbia University , New York, New York
| | - Caleb B Kallen
- Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
37
|
Yang C, Kelaini S, Caines R, Margariti A. RBPs Play Important Roles in Vascular Endothelial Dysfunction Under Diabetic Conditions. Front Physiol 2018; 9:1310. [PMID: 30294283 PMCID: PMC6158626 DOI: 10.3389/fphys.2018.01310] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/30/2018] [Indexed: 12/17/2022] Open
Abstract
Diabetes is one of the major health care problems worldwide leading to huge suffering and burden to patients and society. Diabetes is also considered as a cardiovascular disorder because of the correlation between diabetes and an increased incidence of cardiovascular disease. Vascular endothelial cell dysfunction is a major mediator of diabetic vascular complications. It has been established that diabetes contributes to significant alteration of the gene expression profile of vascular endothelial cells. Post-transcriptional regulation by RNA binding proteins (RBPs) plays an important role in the alteration of gene expression profile under diabetic conditions. The review focuses on the roles and mechanisms of critical RBPs toward diabetic vascular endothelial dysfunction. Deeper understanding of the post- transcriptional regulation by RBPs could lead to new therapeutic strategies against diabetic manifestation in the future.
Collapse
Affiliation(s)
- Chunbo Yang
- Centre for Experimental Medicine, Queens University Belfast, Belfast, United Kingdom
| | - Sophia Kelaini
- Centre for Experimental Medicine, Queens University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- Centre for Experimental Medicine, Queens University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- Centre for Experimental Medicine, Queens University Belfast, Belfast, United Kingdom
| |
Collapse
|
38
|
Elkahloun AG, Rodriguez Y, Alaiyed S, Wenzel E, Saavedra JM. Telmisartan Protects a Microglia Cell Line from LPS Injury Beyond AT1 Receptor Blockade or PPARγ Activation. Mol Neurobiol 2018; 56:3193-3210. [PMID: 30105672 DOI: 10.1007/s12035-018-1300-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/02/2018] [Indexed: 01/12/2023]
Abstract
The Angiotensin II Receptor Blocker (ARB) Telmisartan reduces inflammation through Angiotensin II AT1 receptor blockade and peroxisome proliferator-activated receptor gamma (PPARγ) activation. However, in a mouse microglia-like BV2 cell line, imitating primary microglia responses with high fidelity and devoid of AT1 receptor gene expression or PPARγ activation, Telmisartan reduced gene expression of pro-injury factors, enhanced that of anti-inflammatory genes, and prevented LPS-induced increase in inflammatory markers. Using global gene expression profiling and pathways analysis, we revealed that Telmisartan normalized the expression of hundreds of genes upregulated by LPS and linked with inflammation, apoptosis and neurodegenerative disorders, while downregulating the expression of genes associated with oncological, neurodegenerative and viral diseases. The PPARγ full agonist Pioglitazone had no neuroprotective effects. Surprisingly, the PPARγ antagonists GW9662 and T0070907 were neuroprotective and enhanced Telmisartan effects. GW9226 alone significantly reduced LPS toxic effects and enhanced Telmisartan neuroprotection, including downregulation of pro-inflammatory TLR2 gene expression. Telmisartan and GW9662 effects on LPS injury negatively correlated with pro-inflammatory factors and upstream regulators, including TLR2, and positively with known neuroprotective factors and upstream regulators. Gene Set Enrichment Analysis (GSEA) of the Telmisartan and GW9662 data revealed negative correlations with sets of genes associated with neurodegenerative and metabolic disorders and toxic treatments in cultured systems, while demonstrating positive correlations with gene sets associated with neuroprotection and kinase inhibition. Our results strongly suggest that novel neuroprotective effects of Telmisartan and GW9662, beyond AT1 receptor blockade or PPARγ activation, include downregulation of the TLR2 signaling pathway, findings that may have translational relevance.
Collapse
Affiliation(s)
- Abdel G Elkahloun
- Microarray Core, Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, 50 South Dr, MSC 4435, Bethesda, MD, 20892-4435, USA
| | - Yara Rodriguez
- Laboratory of Neuroprotection, Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA
| | - Seham Alaiyed
- Laboratory of Neuroprotection, Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA
| | - Erin Wenzel
- Laboratory of Neuroprotection, Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA
| | - Juan M Saavedra
- Laboratory of Neuroprotection, Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA.
| |
Collapse
|
39
|
Nutter CA, Kuyumcu-Martinez MN. Emerging roles of RNA-binding proteins in diabetes and their therapeutic potential in diabetic complications. WILEY INTERDISCIPLINARY REVIEWS-RNA 2017; 9. [PMID: 29280295 DOI: 10.1002/wrna.1459] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/19/2017] [Accepted: 11/05/2017] [Indexed: 12/11/2022]
Abstract
Diabetes is a debilitating health care problem affecting 422 million people around the world. Diabetic patients suffer from multisystemic complications that can cause mortality and morbidity. Recent advancements in high-throughput next-generation RNA-sequencing and computational algorithms led to the discovery of aberrant posttranscriptional gene regulatory programs in diabetes. However, very little is known about how these regulatory programs are mis-regulated in diabetes. RNA-binding proteins (RBPs) are important regulators of posttranscriptional RNA networks, which are also dysregulated in diabetes. Human genetic studies provide new evidence that polymorphisms and mutations in RBPs are linked to diabetes. Therefore, we will discuss the emerging roles of RBPs in abnormal posttranscriptional gene expression in diabetes. Questions that will be addressed are: Which posttranscriptional mechanisms are disrupted in diabetes? Which RBPs are responsible for such changes under diabetic conditions? How are RBPs altered in diabetes? How does dysregulation of RBPs contribute to diabetes? Can we target RBPs using RNA-based methods to restore gene expression profiles in diabetic patients? Studying the evolving roles of RBPs in diabetes is critical not only for a comprehensive understanding of diabetes pathogenesis but also to design RNA-based therapeutic approaches for diabetic complications. WIREs RNA 2018, 9:e1459. doi: 10.1002/wrna.1459 This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Processing > Splicing Regulation/Alternative Splicing Translation > Translation Regulation.
Collapse
Affiliation(s)
- Curtis A Nutter
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas
| | - Muge N Kuyumcu-Martinez
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas.,Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
40
|
Singh AK, Aryal B, Zhang X, Fan Y, Price NL, Suárez Y, Fernández-Hernando C. Posttranscriptional regulation of lipid metabolism by non-coding RNAs and RNA binding proteins. Semin Cell Dev Biol 2017; 81:129-140. [PMID: 29183708 DOI: 10.1016/j.semcdb.2017.11.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/14/2017] [Accepted: 11/20/2017] [Indexed: 12/14/2022]
Abstract
Alterations in lipoprotein metabolism enhance the risk of cardiometabolic disorders including type-2 diabetes and atherosclerosis, the leading cause of death in Western societies. While the transcriptional regulation of lipid metabolism has been well characterized, recent studies have uncovered the importance of microRNAs (miRNAs), long-non-coding RNAs (lncRNAs) and RNA binding proteins (RBP) in regulating the expression of lipid-related genes at the posttranscriptional level. Work from several groups has identified a number of miRNAs, including miR-33, miR-122 and miR-148a, that play a prominent role in controlling cholesterol homeostasis and lipoprotein metabolism. Importantly, dysregulation of miRNA expression has been associated with dyslipidemia, suggesting that manipulating the expression of these miRNAs could be a useful therapeutic approach to ameliorate cardiovascular disease (CVD). The role of lncRNAs in regulating lipid metabolism has recently emerged and several groups have demonstrated their regulation of lipoprotein metabolism. However, given the high abundance of lncRNAs and the poor-genetic conservation between species, much work will be needed to elucidate the specific role of lncRNAs in controlling lipoprotein metabolism. In this review article, we summarize recent findings in the field and highlight the specific contribution of lncRNAs and RBPs in regulating lipid metabolism.
Collapse
Affiliation(s)
- Abhishek K Singh
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510, USA
| | - Binod Aryal
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510, USA
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510, USA
| | - Yuhua Fan
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510, USA; College of Pharmacy, Harbin Medical University -Daqing, 163000, PR China
| | - Nathan L Price
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510, USA
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510, USA.
| |
Collapse
|
41
|
The control of inflammation via the phosphorylation and dephosphorylation of tristetraprolin: a tale of two phosphatases. Biochem Soc Trans 2017; 44:1321-1337. [PMID: 27911715 PMCID: PMC5095909 DOI: 10.1042/bst20160166] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 12/14/2022]
Abstract
Twenty years ago, the first description of a tristetraprolin (TTP) knockout mouse highlighted the fundamental role of TTP in the restraint of inflammation. Since then, work from several groups has generated a detailed picture of the expression and function of TTP. It is a sequence-specific RNA-binding protein that orchestrates the deadenylation and degradation of several mRNAs encoding inflammatory mediators. It is very extensively post-translationally modified, with more than 30 phosphorylations that are supported by at least two independent lines of evidence. The phosphorylation of two particular residues, serines 52 and 178 of mouse TTP (serines 60 and 186 of the human orthologue), has profound effects on the expression, function and localisation of TTP. Here, we discuss the control of TTP biology via its phosphorylation and dephosphorylation, with a particular focus on recent advances and on questions that remain unanswered.
Collapse
|
42
|
Ross EA, Naylor AJ, O'Neil JD, Crowley T, Ridley ML, Crowe J, Smallie T, Tang TJ, Turner JD, Norling LV, Dominguez S, Perlman H, Verrills NM, Kollias G, Vitek MP, Filer A, Buckley CD, Dean JL, Clark AR. Treatment of inflammatory arthritis via targeting of tristetraprolin, a master regulator of pro-inflammatory gene expression. Ann Rheum Dis 2017; 76:612-619. [PMID: 27597652 PMCID: PMC5446007 DOI: 10.1136/annrheumdis-2016-209424] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Tristetraprolin (TTP), a negative regulator of many pro-inflammatory genes, is strongly expressed in rheumatoid synovial cells. The mitogen-activated protein kinase (MAPK) p38 pathway mediates the inactivation of TTP via phosphorylation of two serine residues. We wished to test the hypothesis that these phosphorylations contribute to the development of inflammatory arthritis, and that, conversely, joint inflammation may be inhibited by promoting the dephosphorylation and activation of TTP. METHODS The expression of TTP and its relationship with MAPK p38 activity were examined in non-inflamed and rheumatoid arthritis (RA) synovial tissue. Experimental arthritis was induced in a genetically modified mouse strain, in which endogenous TTP cannot be phosphorylated and inactivated. In vitro and in vivo experiments were performed to test anti-inflammatory effects of compounds that activate the protein phosphatase 2A (PP2A) and promote dephosphorylation of TTP. RESULTS TTP expression was significantly higher in RA than non-inflamed synovium, detected in macrophages, vascular endothelial cells and some fibroblasts and co-localised with MAPK p38 activation. Substitution of TTP phosphorylation sites conferred dramatic protection against inflammatory arthritis in mice. Two distinct PP2A agonists also reduced inflammation and prevented bone erosion. In vitro anti-inflammatory effects of PP2A agonism were mediated by TTP activation. CONCLUSIONS The phosphorylation state of TTP is a critical determinant of inflammatory responses, and a tractable target for novel anti-inflammatory treatments.
Collapse
Affiliation(s)
- E A Ross
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - A J Naylor
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - J D O'Neil
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - T Crowley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - M L Ridley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - J Crowe
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - T Smallie
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - T J Tang
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - J D Turner
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - L V Norling
- William Harvey Research Institute, QMUL, London, UK
| | - S Dominguez
- Division of Rheumatology, Northwestern University, Chicago, Illinois, USA
| | - H Perlman
- Division of Rheumatology, Northwestern University, Chicago, Illinois, USA
| | - N M Verrills
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - G Kollias
- Division of Immunology, Biomedical Sciences Research Center ‘Alexander Fleming’, Vari, Greece
| | - M P Vitek
- Cognosci Inc., Research Triangle Park, North Carolina, USA
| | - A Filer
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - C D Buckley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - J L Dean
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - A R Clark
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
43
|
Zielińska KA, Van Moortel L, Opdenakker G, De Bosscher K, Van den Steen PE. Endothelial Response to Glucocorticoids in Inflammatory Diseases. Front Immunol 2016; 7:592. [PMID: 28018358 PMCID: PMC5155119 DOI: 10.3389/fimmu.2016.00592] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/29/2016] [Indexed: 12/16/2022] Open
Abstract
The endothelium plays a crucial role in inflammation. A balanced control of inflammation requires the action of glucocorticoids (GCs), steroidal hormones with potent cell-specific anti-inflammatory properties. Besides the classic anti-inflammatory effects of GCs on leukocytes, recent studies confirm that endothelial cells also represent an important target for GCs. GCs regulate different aspects of endothelial physiology including expression of adhesion molecules, production of pro-inflammatory cytokines and chemokines, and maintenance of endothelial barrier integrity. However, the regulation of endothelial GC sensitivity remains incompletely understood. In this review, we specifically examine the endothelial response to GCs in various inflammatory diseases ranging from multiple sclerosis, stroke, sepsis, and vasculitis to atherosclerosis. Shedding more light on the cross talk between GCs and endothelium will help to improve existing therapeutic strategies and develop new therapies better tailored to the needs of patients.
Collapse
Affiliation(s)
- Karolina A. Zielińska
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Laura Van Moortel
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-UGent, VIB Medical Biotechnology Center, Ghent, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-UGent, VIB Medical Biotechnology Center, Ghent, Belgium
| | | |
Collapse
|
44
|
Chen T, Moore TM, Ebbert MTW, McVey NL, Madsen SR, Hallowell DM, Harris AM, Char RE, Mackay RP, Hancock CR, Hansen JM, Kauwe JS, Thomson DM. Liver kinase B1 inhibits the expression of inflammation-related genes postcontraction in skeletal muscle. J Appl Physiol (1985) 2016; 120:876-88. [PMID: 26796753 DOI: 10.1152/japplphysiol.00727.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/20/2016] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle-specific liver kinase B1 (LKB1) knockout mice (skmLKB1-KO) exhibit elevated mitogen-activated protein kinase (MAPK) signaling after treadmill running. MAPK activation is also associated with inflammation-related signaling in skeletal muscle. Since exercise can induce muscle damage, and inflammation is a response triggered by damaged tissue, we therefore hypothesized that LKB1 plays an important role in dampening the inflammatory response to muscle contraction, and that this may be due in part to increased susceptibility to muscle damage with contractions in LKB1-deficient muscle. Here we studied the inflammatory response and muscle damage with in situ muscle contraction or downhill running. After in situ muscle contractions, the phosphorylation of both NF-κB and STAT3 was increased more in skmLKB1-KO vs. wild-type (WT) muscles. Analysis of gene expression via microarray and RT-PCR shows that expression of many inflammation-related genes increased after contraction only in skmLKB1-KO muscles. This was associated with mild skeletal muscle fiber membrane damage in skmLKB1-KO muscles. Gene markers of oxidative stress were also elevated in skmLKB1-KO muscles after contraction. Using the downhill running model, we observed significantly more muscle damage after running in skmLKB1-KO mice, and this was associated with greater phosphorylation of both Jnk and STAT3 and increased expression of SOCS3 and Fos. In conclusion, we have shown that the lack of LKB1 in skeletal muscle leads to an increased inflammatory state in skeletal muscle that is exacerbated by muscle contraction. Increased susceptibility of the muscle to damage may underlie part of this response.
Collapse
Affiliation(s)
- Ting Chen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Timothy M Moore
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Mark T W Ebbert
- Department of Biology, Brigham Young University, Provo, Utah
| | - Natalie L McVey
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Steven R Madsen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - David M Hallowell
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Alexander M Harris
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Robin E Char
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Ryan P Mackay
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Chad R Hancock
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, Utah; and
| | - Jason M Hansen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - John S Kauwe
- Department of Biology, Brigham Young University, Provo, Utah
| | - David M Thomson
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah;
| |
Collapse
|
45
|
Smallie T, Ross EA, Ammit AJ, Cunliffe HE, Tang T, Rosner DR, Ridley ML, Buckley CD, Saklatvala J, Dean JL, Clark AR. Dual-Specificity Phosphatase 1 and Tristetraprolin Cooperate To Regulate Macrophage Responses to Lipopolysaccharide. THE JOURNAL OF IMMUNOLOGY 2015; 195:277-88. [PMID: 26019272 PMCID: PMC4472943 DOI: 10.4049/jimmunol.1402830] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/27/2015] [Indexed: 01/01/2023]
Abstract
Dual-specificity phosphatase (DUSP) 1 dephosphorylates and inactivates members of the MAPK superfamily, in particular, JNKs, p38α, and p38β MAPKs. It functions as an essential negative regulator of innate immune responses, hence disruption of the Dusp1 gene renders mice extremely sensitive to a wide variety of experimental inflammatory challenges. The principal mechanisms behind the overexpression of inflammatory mediators by Dusp1(-/-) cells are not known. In this study, we use a genetic approach to identify an important mechanism of action of DUSP1, involving the modulation of the activity of the mRNA-destabilizing protein tristetraprolin. This mechanism is key to the control of essential early mediators of inflammation, TNF, CXCL1, and CXCL2, as well as the anti-inflammatory cytokine IL-10. The same mechanism also contributes to the regulation of a large number of transcripts induced by treatment of macrophages with LPS. These findings demonstrate that modulation of the phosphorylation status of tristetraprolin is an important physiological mechanism by which innate immune responses can be controlled.
Collapse
Affiliation(s)
- Tim Smallie
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Ewan A Ross
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Alaina J Ammit
- Faculty of Pharmacy, The University of Sydney, New South Wales 2006, Australia; and
| | - Helen E Cunliffe
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Tina Tang
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Dalya R Rosner
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Michael L Ridley
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Christopher D Buckley
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jeremy Saklatvala
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Jonathan L Dean
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Andrew R Clark
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
46
|
Yu JY, Zhang B, Peng L, Wu CH, Cao H, Zhong JF, Hoffman J, Huang SH. Repositioning of Memantine as a Potential Novel Therapeutic Agent against Meningitic E. coli-Induced Pathogenicities through Disease-Associated Alpha7 Cholinergic Pathway and RNA Sequencing-Based Transcriptome Analysis of Host Inflammatory Responses. PLoS One 2015; 10:e0121911. [PMID: 25993608 PMCID: PMC4437645 DOI: 10.1371/journal.pone.0121911] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/07/2015] [Indexed: 01/09/2023] Open
Abstract
Neonatal sepsis and meningitis (NSM) remains a leading cause worldwide of mortality and morbidity in newborn infants despite the availability of antibiotics over the last several decades. E. coli is the most common gram-negative pathogen causing NSM. Our previous studies show that α7 nicotinic receptor (α7 nAChR), an essential regulator of inflammation, plays a detrimental role in the host defense against NSM. Despite notable successes, there still exists an unmet need for new effective therapeutic approaches to treat this disease. Using the in vitro/in vivo models of the blood-brain barrier (BBB) and RNA-seq, we undertook a drug repositioning study to identify unknown antimicrobial activities for known drugs. We have demonstrated for the first time that memantine (MEM), a FDA-approved drug for treatment of Alzheimer’s disease, could very efficiently block E. coli-caused bacteremia and meningitis in a mouse model of NSM in a manner dependent on α7 nAChR. MEM was able to synergistically enhance the antibacterial activity of ampicillin in HBMEC infected with E. coli K1 (E44) and in neonatal mice with E44-caused bacteremia and meningitis. Differential gene expression analysis of RNA-Seq data from mouse BMEC infected with E. coli K1 showed that several E44-increased inflammatory factors, including IL33, IL18rap, MMP10 and Irs1, were significantly reduced by MEM compared to the infected cells without drug treatment. MEM could also significantly up-regulate anti-inflammatory factors, including Tnfaip3, CISH, Ptgds and Zfp36. Most interestingly, these factors may positively and negatively contribute to regulation of NF-κB, which is a hallmark feature of bacterial meningitis. Furthermore, we have demonstrated that circulating BMEC (cBMEC) are the potential novel biomarkers for NSM. MEM could significantly reduce E44-increased blood level of cBMEC in mice. Taken together, our data suggest that memantine can efficiently block host inflammatory responses to bacterial infection through modulation of both inflammatory and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Jing-Yi Yu
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Bao Zhang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Liang Peng
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America; Department of Clinic Laboratory, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Chun-Hua Wu
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China
| | - John F Zhong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States of America; Department of Perio, Diagnostic Sciences & Biomedical Sciences, School of Dentistry, University of Southern California, Los Angeles, CA, 93003, United States of America; Department of Pediatrics, School of Medicine, University of Southern California, Los Angeles, CA, 93003, United States of America
| | - Jill Hoffman
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Sheng-He Huang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| |
Collapse
|
47
|
Karalok HM, Aydin E, Saglam O, Torun A, Guzeloglu-Kayisli O, Lalioti MD, Kristiansson H, Duke CMP, Choe G, Flannery C, Kallen CB, Seli E. mRNA-binding protein TIA-1 reduces cytokine expression in human endometrial stromal cells and is down-regulated in ectopic endometrium. J Clin Endocrinol Metab 2014; 99:E2610-9. [PMID: 25140393 PMCID: PMC4255110 DOI: 10.1210/jc.2013-3488] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 08/13/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cytokines and growth factors play important roles in endometrial function and the pathogenesis of endometriosis. mRNAs encoding cytokines and growth factors undergo rapid turnover; primarily mediated by adenosine- and uridine-rich elements (AREs) located in their 3'-untranslated regions. T-cell intracellular antigen (TIA-1), an mRNA-binding protein, binds to AREs in target transcripts, leading to decreased gene expression. OBJECTIVE The purpose of this article was to determine whether TIA-1 plays a role in the regulation of endometrial cytokine and growth factor expression during the normal menstrual cycle and whether TIA-1 expression is altered in women with endometriosis. METHODS Eutopic endometrial tissue obtained from women without endometriosis (n = 30) and eutopic and ectopic endometrial tissues from women with endometriosis (n = 17) were immunostained for TIA-1. Staining intensities were evaluated by histological scores (HSCOREs). The regulation of endometrial TIA-1 expression by immune factors and steroid hormones was studied by treating primary cultured human endometrial stromal cells (HESCs) with vehicle, lipopolysaccharide, TNF-α, IL-6, estradiol, or progesterone, followed by protein blot analyses. HESCs were engineered to over- or underexpress TIA-1 to test whether TIA-1 regulates IL-6 or TNF-α expression in these cells. RESULTS We found that TIA-1 is expressed in endometrial stromal and glandular cells throughout the menstrual cycle and that this expression is significantly higher in the perimenstrual phase. In women with endometriosis, TIA-1 expression in eutopic and ectopic endometrium was reduced compared with TIA-1 expression in eutopic endometrium of unaffected control women. Lipopolysaccharide and TNF-α increased TIA-1 expression in HESCs in vitro, whereas IL-6 or steroid hormones had no effect. In HESCs, down-regulation of TIA-1 resulted in elevated IL-6 and TNF-α expression, whereas TIA-1 overexpression resulted in decreased IL-6 and TNF-α expression. CONCLUSIONS Endometrial TIA-1 is regulated throughout the menstrual cycle, TIA-1 modulates the expression of immune factors in endometrial cells, and downregulation of TIA-1 may contribute to the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Hakan Mete Karalok
- Department of Obstetrics, Gynecology and Reproductive Sciences (H.M.K., E.A., A.T., O.G.-K., M.D.L., H.K., C.M.P.D., G.C., C.F., E.S.) and Department of Pathology (O.S.), Yale School of Medicine, New Haven, Connecticut 06520-8063; and Department of Obstetrics and Gynecology (C.B.K.), Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Xin H, Deng K, Fu M. Post-transcriptional gene regulation by RNA-binding proteins in vascular endothelial dysfunction. SCIENCE CHINA. LIFE SCIENCES 2014; 57:836-44. [PMID: 25104457 PMCID: PMC7089175 DOI: 10.1007/s11427-014-4703-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/01/2014] [Indexed: 12/11/2022]
Abstract
Endothelial cell dysfunction is a term which implies the dysregulation of normal endothelial cell functions, including impairment of the barrier functions, control of vascular tone, disturbance of proliferative and migratory capacity of endothelial cells, as well as control of leukocyte trafficking. Endothelial dysfunction is an early step in vascular inflammatory diseases such as atherosclerosis, diabetic vascular complications, sepsis-induced or severe virus infection-induced organ injuries. The expressions of inflammatory cytokines and vascular adhesion molecules induced by various stimuli, such as modified lipids, smoking, advanced glycation end products and bacteria toxin, significantly contribute to the development of endothelial dysfunction. The transcriptional regulation of inflammatory cytokines and vascular adhesion molecules has been well-studied. However, the regulation of those gene expressions at post-transcriptional level is emerging. RNA-binding proteins have emerged as critical regulators of gene expression acting predominantly at the post-transcriptional level in microRNA-dependent or independent manners. This review summarizes the latest insights into the roles of RNA-binding proteins in controlling vascular endothelial cell functions and their contribution to the pathogenesis of vascular inflammatory diseases.
Collapse
Affiliation(s)
- HongBo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| | - KeYu Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| | - MinGui Fu
- Institute of Translational Medicine, Nanchang University, Nanchang, 330031 China
| |
Collapse
|
49
|
Bollmann F, Wu Z, Oelze M, Siuda D, Xia N, Henke J, Daiber A, Li H, Stumpo DJ, Blackshear PJ, Kleinert H, Pautz A. Endothelial dysfunction in tristetraprolin-deficient mice is not caused by enhanced tumor necrosis factor-α expression. J Biol Chem 2014; 289:15653-65. [PMID: 24727475 PMCID: PMC4140920 DOI: 10.1074/jbc.m114.566984] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 04/09/2014] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular events are important co-morbidities in patients with chronic inflammatory diseases like rheumatoid arthritis. Tristetraprolin (TTP) regulates pro-inflammatory processes through mRNA destabilization and therefore TTP-deficient mice (TTP(-/-) mice) develop a chronic inflammation resembling human rheumatoid arthritis. We used this mouse model to evaluate molecular signaling pathways contributing to the enhanced atherosclerotic risk in chronic inflammatory diseases. In the aorta of TTP(-/-) mice we observed elevated mRNA expression of known TTP targets like tumor necrosis factor-α (TNF-α) and macrophage inflammatory protein-1α, as well as of other pro-atherosclerotic mediators, like Calgranulin A, Cathepsin S, and Osteopontin. Independent of cholesterol levels TTP(-/-) mice showed a significant reduction of acetylcholine-induced, nitric oxide-mediated vasorelaxation. The endothelial dysfunction in TTP(-/-) mice was associated with increased levels of reactive oxygen and nitrogen species (RONS), indicating an enhanced nitric oxide inactivation by RONS in the TTP(-/-) animals. The altered RONS generation correlates with increased expression of NADPH oxidase 2 (Nox2) resulting from enhanced Nox2 mRNA stability. Although TNF-α is believed to be a central mediator of inflammation-driven atherosclerosis, genetic inactivation of TNF-α neither improved endothelial function nor normalized Nox2 expression or RONS production in TTP(-/-) animals. Systemic inflammation caused by TTP deficiency leads to endothelial dysfunction. This process is independent of cholesterol and not mediated by TNF-α solely. Thus, other mediators, which need to be identified, contribute to enhanced cardiovascular risk in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Franziska Bollmann
- From the Department of Pharmacology, Center for Thrombosis and Hemostasis, and
| | | | - Matthias Oelze
- 2nd Medical Clinic, Molecular Cardiology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany and
| | - Daniel Siuda
- From the Department of Pharmacology, Center for Thrombosis and Hemostasis, and
| | - Ning Xia
- From the Department of Pharmacology
| | | | - Andreas Daiber
- 2nd Medical Clinic, Molecular Cardiology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany and
| | - Huige Li
- From the Department of Pharmacology
| | - Deborah J Stumpo
- the Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Perry J Blackshear
- the Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | | | | |
Collapse
|
50
|
Hahn RT, Hoppstädter J, Hirschfelder K, Hachenthal N, Diesel B, Kessler SM, Huwer H, Kiemer AK. Downregulation of the glucocorticoid-induced leucine zipper (GILZ) promotes vascular inflammation. Atherosclerosis 2014; 234:391-400. [PMID: 24747114 DOI: 10.1016/j.atherosclerosis.2014.03.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 02/28/2014] [Accepted: 03/23/2014] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Glucocorticoid-induced leucine zipper (GILZ) represents an anti-inflammatory mediator, whose downregulation has been described in various inflammatory processes. Aim of our study was to decipher the regulation of GILZ in vascular inflammation. APPROACH AND RESULTS Degenerated aortocoronary saphenous vein bypass grafts (n = 15), which exhibited inflammatory cell activation as determined by enhanced monocyte chemoattractrant protein 1 (MCP-1, CCL2) and Toll-like receptor 2 (TLR2) expression, showed significantly diminished GILZ protein and mRNA levels compared to healthy veins (n = 23). GILZ was also downregulated in human umbilical vein endothelial cells (HUVEC) and macrophages upon treatment with the inflammatory cytokine TNF-α in a tristetraprolin (ZFP36, TTP)- and p38 MAPK-dependent manner. To assess the functional implications of decreased GILZ expression, we determined NF-κB activation after GILZ knockdown by siRNA and found that NF-κB activity and inflammatory gene expression were significantly enhanced. Importantly, ZFP36 is induced in TNF-α-activated HUVEC as well as in degenerated vein bypasses. When atheroprotective laminar shear stress was employed, GILZ levels in HUVEC increased on mRNA and protein level. Laminar flow also counteracted TNF-α-induced ZFP36 expression and GILZ downregulation. MAP kinase phosphatase 1 (MKP-1, DUSP1), a negative regulator of ZFP36 expression, was distinctly upregulated under laminar shear stress conditions and downregulated in degenerated vein bypasses. CONCLUSION Our data show a diminished expression of the anti-inflammatory mediator GILZ in the inflamed vasculature and indicate that GILZ downregulation requires the mRNA binding protein ZFP36. We suggest that reduced GILZ levels play a role in cardiovascular disease.
Collapse
Affiliation(s)
- Rebecca T Hahn
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbruecken, Germany
| | - Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbruecken, Germany
| | - Kerstin Hirschfelder
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbruecken, Germany
| | - Nina Hachenthal
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbruecken, Germany
| | - Britta Diesel
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbruecken, Germany
| | - Sonja M Kessler
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbruecken, Germany
| | - Hanno Huwer
- Department of Cardiothoracic Surgery, Völklingen Heart Centre, Völklingen, Germany
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbruecken, Germany.
| |
Collapse
|