1
|
Tierney JW, Francisco RP, Yu F, Ma J, Cheung-Flynn J, Keech MC, D'Arcy R, Shah VM, Kittel AR, Chang DJ, McCune JT, Bezold MG, Aligwekwe AN, Cook RS, Beckman JA, Brophy CM, Duvall CL. Intravascular delivery of an MK2 inhibitory peptide to prevent restenosis after angioplasty. Biomaterials 2025; 313:122767. [PMID: 39216327 DOI: 10.1016/j.biomaterials.2024.122767] [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: 03/07/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Peripheral artery disease is commonly treated with balloon angioplasty, a procedure involving minimally invasive, transluminal insertion of a catheter to the site of stenosis, where a balloon is inflated to open the blockage, restoring blood flow. However, peripheral angioplasty has a high rate of restenosis, limiting long-term patency. Therefore, angioplasty is sometimes paired with delivery of cytotoxic drugs like paclitaxel to reduce neointimal tissue formation. We pursue intravascular drug delivery strategies that target the underlying cause of restenosis - intimal hyperplasia resulting from stress-induced vascular smooth muscle cell switching from the healthy contractile into a pathological synthetic phenotype. We have established MAPKAP kinase 2 (MK2) as a driver of this phenotype switch and seek to establish convective and contact transfer (coated balloon) methods for MK2 inhibitory peptide delivery to sites of angioplasty. Using a flow loop bioreactor, we showed MK2 inhibition in ex vivo arteries suppresses smooth muscle cell phenotype switching while preserving vessel contractility. A rat carotid artery balloon injury model demonstrated inhibition of intimal hyperplasia following MK2i coated balloon treatment in vivo. These studies establish both convective and drug coated balloon strategies as promising approaches for intravascular delivery of MK2 inhibitory formulations to improve efficacy of balloon angioplasty.
Collapse
Affiliation(s)
- J William Tierney
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - R Paolo Francisco
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Megan C Keech
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Richard D'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Chemical Engineering, School of Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, USA
| | - Veeraj M Shah
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Anna R Kittel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Devin J Chang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joshua T McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Mariah G Bezold
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Adrian N Aligwekwe
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; North Carolina State University, Raleigh, NC, 27695, USA
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Joshua A Beckman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Colleen M Brophy
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Veterans Affairs Medical Center, VA Tennessee Valley Healthcare System, Nashville, TN, 37212, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
2
|
Chun-peng ZHANG, Tian CAO, Xue YANG. Pharmacological mechanisms of Taohe Chengqi decoction in diabetic cardiovascular complications: A systematic review, network pharmacology and molecular docking. Heliyon 2024; 10:e33308. [PMID: 39044965 PMCID: PMC11263673 DOI: 10.1016/j.heliyon.2024.e33308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Background Diabetic cardiovascular complications are the leading cause of diabetes-related deaths. These complications place an enormous and growing burden on global health systems and economies. The objective of this study was to conduct a systematic review on the therapeutic mechanisms of Taohe Chengqi Decoction (THCQD) in the treatment of diabetic cardiovascular complications. To predict the potential mechanisms of action of THCQD on diabetic cardiovascular complications using network pharmacology, and to validate these predictions through molecular docking analysis. Methods To collect relevant animal experiments, we searched a total of 6 databases. Eligibility for the study was determined based on inclusion and exclusion criteria. Data extraction was then performed on the literature. Methodological quality of animal studies was assessed using SYRCLE criteria. Based on network pharmacology, intersecting genes for THCQD and diabetic cardiovascular complications were obtained using Venny, PPI analysis and topology analysis of intersecting genes were performed; GO and KEGG were used for enrichment analysis and prediction of new targets of action. Molecular docking techniques were employed to model the interactions between drug components and target genes, thereby validating the results of network pharmacology predictions. Results A total of 16 studies were finally identified that fit the direction of this review. Included 6 studies of the myocardium, 1 study of the aortic arch, 5 studies of the femoral artery, 4 studies of the thoracic aorta. THCQD exhibited anti-inflammatory, anti-fibrotic and anti-atherosclerotic effects on cardiovascular complications in diabetic rats. Network pharmacology results showed that C0363 (Resveratrol), C0041 (Emodin), and C1114 (Baicalein) were the key components in the treatment of diabetic cardiovascular complications by THCQD. PPI results showed that INS, AKT1, TNF, ALB, IL6, IL1B as the genes that interact with the top 6. KEGG enrichment analysis identified the AGE-RAGE signaling pathway in diabetic complications as the most prominent pathway enriched by THCQD for diabetic cardiovascular complications genes. The results of molecular docking showed that the key active components demonstrated favorable interactions with their corresponding target genes. Conclusion In conclusion, the results of both basic and web-based pharmacological studies support the beneficial effects of the natural herbal formulation THCQD on diabetic cardiovascular complications. This decoction has anti-inflammatory and antifibrotic properties and is effective in ameliorating diabetic cardiovascular disease. The network pharmacology results further support these ideas and identify the AGE-RAGE signaling pathway in diabetic complications as possibly the most relevant pathway for THCQD in the treatment of diabetic cardiovascular complications. The extent of the therapeutic potential of all-natural herbal components in the treatment of diabetic cardiovascular disease merits further investigation.
Collapse
Affiliation(s)
- ZHANG Chun-peng
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - CAO Tian
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - YANG Xue
- Department of Traditional Chinese Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| |
Collapse
|
3
|
Chen W, Zhong Y, Yuan Y, Zhu M, Hu W, Liu N, Xing D. New insights into the suppression of inflammation and lipid accumulation by JAZF1. Genes Dis 2023; 10:2457-2469. [PMID: 37554201 PMCID: PMC10404878 DOI: 10.1016/j.gendis.2022.10.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/27/2022] [Accepted: 10/25/2022] [Indexed: 12/03/2022] Open
Abstract
Atherosclerosis is one of the leading causes of disease and death worldwide. The identification of new therapeutic targets and agents is critical. JAZF1 is expressed in many tissues and is found at particularly high levels in adipose tissue (AT). JAZF1 suppresses inflammation (including IL-1β, IL-4, IL-6, IL-8, IL-10, TNFα, IFN-γ, IAR-20, COL3A1, laminin, and MCP-1) by reducing NF-κB pathway activation and AT immune cell infiltration. JAZF1 reduces lipid accumulation by regulating the liver X receptor response element (LXRE) of the SREBP-1c promoter, the cAMP-response element (CRE) of HMGCR, and the TR4 axis. LXRE and CRE sites are present in many cytokine and lipid metabolism gene promoters, which suggests that JAZF1 regulates these genes through these sites. NF-κB is the center of the JAZF1-mediated inhibition of the inflammatory response. JAZF1 suppresses NF-κB expression by suppressing TAK1 expression. Interestingly, TAK1 inhibition also decreases lipid accumulation. A dual-targeting strategy of NF-κB and TAK1 could inhibit both inflammation and lipid accumulation. Dual-target compounds (including prodrugs) 1-5 exhibit nanomolar inhibition by targeting NF-κB and TAK1, EGFR, or COX-2. However, the NF-κB suppressing activity of these compounds is relatively low (IC50 > 300 nM). Compounds 6-14 suppress NF-κB expression with IC50 values ranging from 1.8 nM to 38.6 nM. HS-276 is a highly selective, orally bioavailable TAK1 inhibitor. Combined structural modifications of compounds using a prodrug strategy may enhance NF-κB inhibition. This review focused on the role and mechanism of JAZF1 in inflammation and lipid accumulation for the identification of new anti-atherosclerotic targets.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Yingjie Zhong
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Yang Yuan
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Meng Zhu
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Wenchao Hu
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
- Department of Endocrinology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong 266035, China
| | - Ning Liu
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Dongming Xing
- Cancer Institute, Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
4
|
Choi JY, Jin X, Kim H, Koh S, Cho HJ, Kim BG. High Mobility Group Box 1 as an Autocrine Chemoattractant for Oligodendrocyte Lineage Cells in White Matter Stroke. Stroke 2023; 54:575-586. [PMID: 36490365 DOI: 10.1161/strokeaha.122.041414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The migration of oligodendrocyte precursor cells (OPC) is a key process of remyelination, which is essential for the treatment of white matter stroke. This study aimed to investigate the role of HMGB1 (high mobility group box 1), a damage-associated molecular pattern released from dying oligodendrocytes, as an autocrine chemoattractant that promotes OPC migration. METHODS The migratory capacity of primary cultured OPCs was measured using the Boyden chamber assay. The downstream pathway of HMGB1-mediated OPC migration was specified by siRNA-induced knockdown or pharmacological blockade of TLR2 (toll-like receptor 2), RAGE (receptor for advanced glycation end product), Src, ERK1/2 (extracellular signal-regulated kinase1/2), and FAK (focal adhesion kinase). Conditioned media were collected from oxygen-glucose deprivation-treated oligodendrocytes, and the impact on OPC migration was assessed. Lesion size and number of intralesional Olig2(+) cells were analyzed in an in vivo model of white matter stroke with N5-(1-iminoethyl)-L-ornithine (L-NIO). RESULTS HMGB1 treatment promoted OPC migration. HMGB1 antagonism reversed such effects to untreated levels. Among the candidates for the downstream signal of HMGB1-mediated migration, the knockdown of TLR2 rather than that of RAGE attenuated the migration-promoting effect of HMGB1. Further specification of the HMGB1-TLR2 axis revealed that the phosphorylation of ERK1/2 and its downstream molecule FAK, rather than of Src, was decreased in TLR2-knockdown OPCs, and pharmacological inhibition of ERK1/2 and FAK led to decreased OPC migration. Oxygen-glucose deprivation-conditioned media promoted OPC migration, suggesting the autocrine chemoattractant function of HMGB1. In vivo, TLR2(-/-)-mice showed lesser intralesional Olig2(+) cells compared to wild-type controls in response to L-NIO induced ischemic injury regardless of HMGB1 administration. CONCLUSIONS HMGB1, through the TLR2-ERK1/2-FAK axis, functions as an autocrine chemoattractant to promote OPC migration, which is an initial and indispensable step in remyelination. Thus, a novel treatment strategy for white matter stroke based on the HMGB1-TLR2 axis in the oligodendrocyte lineage could be feasible.
Collapse
Affiliation(s)
- Jun Young Choi
- Department of Brain science, Ajou University School of Medicine, Republic of Korea (J.Y.C., X.J., H.K., S.K., H.J.C., B.G.K.).,Department of Neurology, Ajou University School of Medicine, Republic of Korea (J.Y.C., S.K., B.G.K.)
| | - Xuelian Jin
- Department of Brain science, Ajou University School of Medicine, Republic of Korea (J.Y.C., X.J., H.K., S.K., H.J.C., B.G.K.).,Neuroscience graduate program, Ajou University Graduate School of Medicine, Republic of Korea (X.J., H.K., S.K.).,Department of Nephrology, Suqian First Hospital, Jiangsu, China (X.J.)
| | - Hanki Kim
- Department of Brain science, Ajou University School of Medicine, Republic of Korea (J.Y.C., X.J., H.K., S.K., H.J.C., B.G.K.).,Neuroscience graduate program, Ajou University Graduate School of Medicine, Republic of Korea (X.J., H.K., S.K.)
| | - Seungyon Koh
- Department of Brain science, Ajou University School of Medicine, Republic of Korea (J.Y.C., X.J., H.K., S.K., H.J.C., B.G.K.).,Department of Neurology, Ajou University School of Medicine, Republic of Korea (J.Y.C., S.K., B.G.K.).,Neuroscience graduate program, Ajou University Graduate School of Medicine, Republic of Korea (X.J., H.K., S.K.)
| | - Hyo Jin Cho
- Department of Brain science, Ajou University School of Medicine, Republic of Korea (J.Y.C., X.J., H.K., S.K., H.J.C., B.G.K.)
| | - Byung Gon Kim
- Department of Brain science, Ajou University School of Medicine, Republic of Korea (J.Y.C., X.J., H.K., S.K., H.J.C., B.G.K.).,Department of Neurology, Ajou University School of Medicine, Republic of Korea (J.Y.C., S.K., B.G.K.)
| |
Collapse
|
5
|
López-Acosta O, Ruiz-Ramírez A, Barrios-Maya MÁ, Alarcon-Aguilar J, Alarcon-Enos J, Céspedes Acuña CL, El-Hafidi M. Lipotoxicity, glucotoxicity and some strategies to protect vascular smooth muscle cell against proliferative phenotype in metabolic syndrome. Food Chem Toxicol 2023; 172:113546. [PMID: 36513245 DOI: 10.1016/j.fct.2022.113546] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MetS) is a risk factor for the development of cardiovascular disease (CVD) and atherosclerosis through a mechanism that involves vascular smooth muscle cell (VSMC) proliferation, lipotoxicity and glucotoxicity. Several molecules found to be increased in MetS, including free fatty acids, fatty acid binding protein 4, leptin, resistin, oxidized lipoprotein particles, and advanced glycation end products, influence VSMC proliferation. Most of these molecules act through their receptors on VSMCs by activating several signaling pathways associated with ROS generation in various cellular compartments. ROS from NADPH-oxidase and mitochondria have been found to promote VSMC proliferation and cell cycle progression. In addition, most of the natural or synthetic substances described in this review, including pharmaceuticals with hypoglycemic and hypolipidemic properties, attenuate VSMC proliferation by their simultaneous modulation of cell signaling and their scavenging property due to the presence of a phenolic ring in their structure. This review discusses recent data in the literature on the role that several MetS-related molecules and ROS play in the change from contractile to proliferative phenotype of VSMCs. Hence the importance of proposing an appropriate strategy to prevent uncontrolled VSMC proliferation using antioxidants, hypoglycemic and hypolipidemic agents.
Collapse
Affiliation(s)
- Ocarol López-Acosta
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Angélica Ruiz-Ramírez
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Miguel-Ángel Barrios-Maya
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Javier Alarcon-Aguilar
- Laboratorio de Farmacología, Depto. de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Iztapalapa, Iztapalapa, Mexico
| | - Julio Alarcon-Enos
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Av. Andres Bello 720, Chillan, Chile
| | - Carlos L Céspedes Acuña
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Av. Andres Bello 720, Chillan, Chile.
| | - Mohammed El-Hafidi
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico.
| |
Collapse
|
6
|
Xi X, Li H, Chen S, Lv T, Ma T, Jiang R, Zhang P, Wong WH, Zhang X. Unfolding the genotype-to-phenotype black box of cardiovascular diseases through cross-scale modeling. iScience 2022; 25:104790. [PMID: 35992073 PMCID: PMC9386115 DOI: 10.1016/j.isci.2022.104790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/26/2022] [Accepted: 07/14/2022] [Indexed: 12/01/2022] Open
Abstract
Complex traits such as cardiovascular diseases (CVD) are the results of complicated processes jointly affected by genetic and environmental factors. Genome-wide association studies (GWAS) identified genetic variants associated with diseases but usually did not reveal the underlying mechanisms. There could be many intermediate steps at epigenetic, transcriptomic, and cellular scales inside the black box of genotype-phenotype associations. In this article, we present a machine-learning-based cross-scale framework GRPath to decipher putative causal paths (pcPaths) from genetic variants to disease phenotypes by integrating multiple omics data. Applying GRPath on CVD, we identified 646 and 549 pcPaths linking putative causal regions, variants, and gene expressions in specific cell types for two types of heart failure, respectively. The findings suggest new understandings of coronary heart disease. Our work promoted the modeling of tissue- and cell type-specific cross-scale regulation to uncover mechanisms behind disease-associated variants, and provided new findings on the molecular mechanisms of CVD. We defined one type of cross-scale genotype-to-phenotype regulation path We designed a framework GRPath to uncover putative regulation paths for diseases GRPath helped uncover molecular mechanisms for two major types of heart failure
Collapse
Affiliation(s)
- Xi Xi
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Haochen Li
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shengquan Chen
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Tingting Lv
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Tianxing Ma
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Rui Jiang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
| | - Ping Zhang
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Wing Hung Wong
- Departments of Statistics and Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing 100084, China
- School of Medicine, Tsinghua University, Beijing 100084, China
- Corresponding author
| |
Collapse
|
7
|
Kotlyarov S, Kotlyarova A. Involvement of Fatty Acids and Their Metabolites in the Development of Inflammation in Atherosclerosis. Int J Mol Sci 2022; 23:ijms23031308. [PMID: 35163232 PMCID: PMC8835729 DOI: 10.3390/ijms23031308] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Despite all the advances of modern medicine, atherosclerosis continues to be one of the most important medical and social problems. Atherosclerosis is the cause of several cardiovascular diseases, which are associated with high rates of disability and mortality. The development of atherosclerosis is associated with the accumulation of lipids in the arterial intima and the disruption of mechanisms that maintain the balance between the development and resolution of inflammation. Fatty acids are involved in many mechanisms of inflammation development and maintenance. Endothelial cells demonstrate multiple cross-linkages between lipid metabolism and innate immunity. In addition, these processes are linked to hemodynamics and the function of other cells in the vascular wall, highlighting the central role of the endothelium in vascular biology.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
- Correspondence:
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
8
|
Klimentova EA, Suchkov IA, Egorov AA, Kalinin RE. Apoptosis and Cell Proliferation Markers in Inflammatory-Fibroproliferative Diseases of the Vessel Wall (Review). Sovrem Tekhnologii Med 2021; 12:119-126. [PMID: 34795999 PMCID: PMC8596273 DOI: 10.17691/stm2020.12.4.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Indexed: 12/11/2022] Open
Abstract
Apoptosis is the main feature of inflammatory-fibroproliferative disorders of the vessel wall. Studies in animal models have shown that smooth muscle cells (SMCs) cultured from endarterectomy specimens from the affected area proliferate more slowly and display higher apoptotic indices than SMCs derived from the normal vessel wall. Apoptotic cells were found in the destabilized atherosclerotic plaques, as well as in the samples with restenosis of the reconstruction area. Injury to the vessel wall causes two waves of apoptosis. The first wave is the rapid apoptosis in the media that occurs within a few hours after injury and leads to a marked reduction in the number of vascular wall cells. The second wave of apoptosis occurs much later (from several days to weeks) and is limited by the SMCs within the developing neointima. Up to 14% of the neointimal SMCs undergo apoptosis 20 days after balloon angioplasty. Ligation of the external carotid artery in a rabbit model led to a marked decrease in blood flow in the common carotid artery, which correlated with the increased apoptosis of endothelial cells and SMCs. Angioplasty-induced death of SMCs is regulated by a redox-sensitive signaling pathway, and topical administration of antioxidants can minimize vascular cell loss. On the whole, studies show that apoptosis is prevalent in vascular lesions, controlling the viability of both inflammatory and vascular cells, determining the cellular composition of the vessel wall. The main markers of apoptosis (Fas, Fas ligand, p53, Bcl-2, Bax) and cell proliferation (toll receptor) have been considered in the current review.
Collapse
Affiliation(s)
- E A Klimentova
- Department of Cardiovascular, X-ray Endovascular, Operative Surgery, and Topographic Anatomy; Ryazan State Medical University, 9 Vysokovoltnaya St., Ryazan, 390026, Russia
| | - I A Suchkov
- Professor, Department of Cardiovascular, X-ray Endovascular, Operative Surgery, and Topographic Anatomy; Ryazan State Medical University, 9 Vysokovoltnaya St., Ryazan, 390026, Russia
| | - A A Egorov
- Doctoral Student, Department of Cardiovascular, X-ray Endovascular, Operative Surgery, and Topographic Anatomy; Ryazan State Medical University, 9 Vysokovoltnaya St., Ryazan, 390026, Russia
| | - R E Kalinin
- Professor, Head of the Department of Cardiovascular, X-ray Endovascular, Operative Surgery, and Topographic Anatomy Ryazan State Medical University, 9 Vysokovoltnaya St., Ryazan, 390026, Russia
| |
Collapse
|
9
|
Lee GL, Liao TL, Wu JY, Wu KK, Kuo CC. Restoration of 5-methoxytryptophan protects against atherosclerotic chondrogenesis and calcification in ApoE -/- mice fed high fat diet. J Biomed Sci 2021; 28:74. [PMID: 34749728 PMCID: PMC8573875 DOI: 10.1186/s12929-021-00771-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/28/2021] [Indexed: 11/18/2022] Open
Abstract
Background Toll-like receptor-2 (TLR2) promotes vascular smooth muscle cell (VSMC) transdifferentiation to chondrocytes and calcification in a p38 MAPK-dependent manner. Vascular 5-methoxytryptophan (5-MTP) is a newly identified factor with anti-inflammatory actions. As 5-MTP targets p38 MAPK for its actions, we postulated that 5-MTP protects against vascular chondrogenesis and calcification. Methods High-fat diet-induced advanced atherosclerosis in mice were performed to investigate the effect of 5-MTP on atherosclerotic lesions and calcification. VSMCs were used to determine the role of 5-MTP in VSMC chondrogenic differentiation and calcification. Alizarin red S and Alcian blue staining were used to measure VSMC calcification and chondrogenic differentiation, respectively. Results 5-MTP was detected in aortic tissues of ApoE−/− mice fed control chow. It was reduced in ApoE−/− mice fed high-fat diet (HFD), but was restored in ApoE−/−Tlr2−/− mice, suggesting that HFD reduces vascular 5-MTP production via TLR2. Intraperitoneal injection of 5-MTP or its analog into ApoE−/− mice fed HFD reduced aortic atherosclerotic lesions and calcification which was accompanied by reduction of chondrogenesis and calcium deposition. Pam3CSK4 (Pam3), ligand of TLR2, induced SMC phenotypic switch to chondrocytes. Pretreatment with 5-MTP preserved SMC contractile proteins and blocked Pam3-induced chondrocyte differentiation and calcification. 5-MTP inhibited HFD-induced p38 MAPK activation in vivo and Pam3-induced p38 MAPK activation in SMCs. 5-MTP suppressed HFD-induced CREB activation in aortic tissues and Pam3-induced CREB and NF-κB activation in SMCs. Conclusions These findings suggest that 5-MTP is a vascular arsenal against atherosclerosis and calcification by inhibiting TLR2–mediated SMC phenotypic switch to chondrocytes and the consequent calcification. 5-MTP exerts these effects by blocking p38 MAPK activation and inhibiting CREB and NF-κB transactivation activity. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-021-00771-1.
Collapse
Affiliation(s)
- Guan-Lin Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Tsai-Lien Liao
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Jing-Yiing Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Kenneth K Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan. .,College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan. .,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.
| |
Collapse
|
10
|
Tierney JW, Evans BC, Cheung-Flynn J, Wang B, Colazo JM, Polcz ME, Cook RS, Brophy CM, Duvall CL. Therapeutic MK2 inhibition blocks pathological vascular smooth muscle cell phenotype switch. JCI Insight 2021; 6:e142339. [PMID: 34622803 PMCID: PMC8525639 DOI: 10.1172/jci.insight.142339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Vascular procedures, such as stenting, angioplasty, and bypass grafting, often fail due to intimal hyperplasia (IH), wherein contractile vascular smooth muscle cells (VSMCs) dedifferentiate to synthetic VSMCs, which are highly proliferative, migratory, and fibrotic. Previous studies suggest MAPK-activated protein kinase 2 (MK2) inhibition may limit VSMC proliferation and IH, although the molecular mechanism underlying the observation remains unclear. We demonstrated here that MK2 inhibition blocked the molecular program of contractile to synthetic dedifferentiation and mitigated IH development. Molecular markers of the VSMC contractile phenotype were sustained over time in culture in rat primary VSMCs treated with potent, long-lasting MK2 inhibitory peptide nanopolyplexes (MK2i-NPs), a result supported in human saphenous vein specimens cultured ex vivo. RNA-Seq of MK2i-NP-treated primary human VSMCs revealed programmatic switching toward a contractile VSMC gene expression profile, increasing expression of antiinflammatory and contractile-associated genes while lowering expression of proinflammatory, promigratory, and synthetic phenotype-associated genes. Finally, these results were confirmed using an in vivo rabbit vein graft model where brief, intraoperative treatment with MK2i-NPs decreased IH and synthetic phenotype markers while preserving contractile proteins. These results support further development of MK2i-NPs as a therapy for blocking VSMC phenotype switch and IH associated with cardiovascular procedures.
Collapse
Affiliation(s)
- J. William Tierney
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Brian C. Evans
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of General Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bo Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Monica E. Polcz
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Department of General Surgery and
| | - Rebecca S. Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Colleen M. Brophy
- Division of Vascular Surgery, Department of General Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
11
|
Guo Z, Zhang Y, Liu C, Youn JY, Cai H. Toll-Like Receptor 2 (TLR2) Knockout Abrogates Diabetic and Obese Phenotypes While Restoring Endothelial Function via Inhibition of NOX1. Diabetes 2021; 70:2107-2119. [PMID: 34127487 PMCID: PMC8576422 DOI: 10.2337/db20-0591] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/08/2021] [Indexed: 11/13/2022]
Abstract
We have previously demonstrated a novel role of bone morphogenic protein 4 (BMP4) in inducing NOX1-dependent endothelial nitric oxide synthase (eNOS) uncoupling, endothelial dysfunction, and inflammatory activation in type 2 diabetes mellitus (T2DM). However, how BMP4 activates NOX1 and whether targeting the new mechanistic pathway revealed is effective in preserving endothelial function in T2DM remains unclear. In this study, we observed that BMP4 induced a marked, time-dependent increase in physiological binding between TLR2 and NOX1 in aortic endothelial cells as well as increased binding of TLR2 to NOXO1. In TLR2 knockout (Tlr2 -/-) mice fed high-fat diet, body weight gain was significantly less compared with wild-type (WT) mice both in males and females. The high-fat diet-induced increases in fasting blood glucose levels, as well as in circulating insulin and leptin levels, were absent in Tlr2 -/- mice. High-fat feeding induced increases in overall fat mass, and in fat mass of different pockets were abrogated in Tlr2 -/- mice. Whereas energy intake was similar in high-fat-fed WT and Tlr2 -/- mice, TLR2 deficiency resulted in higher energy expenditure attributable to improved physical activity, which was accompanied by restored skeletal muscle mitochondrial function. In addition, TLR2 deficiency recoupled eNOS, reduced total superoxide production, improved H4B and NO bioavailabilities in aortas, and restored endothelium-dependent vasorelaxation. Collectively, our data strongly indicate that TLR2 plays important roles in the development of metabolic features of T2DM and its related endothelial/vascular dysfunction. Therefore, targeting TLR2 may represent a novel therapeutic strategy for T2DM, obesity, and cardiovascular complications via specific inhibition of NOX1.
Collapse
Affiliation(s)
- Zhen Guo
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Chang Liu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
12
|
Qi B, Chen JH, Tao L, Zhu CM, Wang Y, Deng GX, Miao L. Integrated Weighted Gene Co-expression Network Analysis Identified That TLR2 and CD40 Are Related to Coronary Artery Disease. Front Genet 2021; 11:613744. [PMID: 33574831 PMCID: PMC7870792 DOI: 10.3389/fgene.2020.613744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/15/2020] [Indexed: 01/14/2023] Open
Abstract
The current research attempted to identify possible hub genes and pathways of coronary artery disease (CAD) and to detect the possible mechanisms. Array data from GSE90074 were downloaded from the Gene Expression Omnibus (GEO) database. Integrated weighted gene co-expression network analysis (WGCNA) was performed to analyze the gene module and clinical characteristics. Gene Ontology annotation (GO), Disease Ontology (DO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed by clusterProfiler and the DOSE package in R. A protein-protein interaction (PPI) network was established using Cytoscape software, and significant modules were analyzed using Molecular Complex Detection (MCODE) to identify hub genes. Then, further functional validation of hub genes in other microarrays and population samples was performed, and survival analysis was performed to investigate the prognosis. A total of 660 genes were located in three modules and associated with CAD. GO functions identified 484 biological processes, 39 cellular components, and 22 molecular functions with an adjusted P < 0.05. In total, 38 pathways were enriched in KEGG pathway analysis, and 147 DO items were identified with an adjusted P < 0.05 (false discovery rate, FDR set at < 0.05). There was a total of four modules with a score > 10 after PPI network analysis using the MCODE app, and two hub genes (TLR2 and CD14) were identified. Then, we validated the information from the GSE60993 dataset using the GSE59867 dataset and population samples, and we found that these two genes were associated with plaque vulnerability. These two genes varied at different time points after myocardial infarction, and both of them had the lowest prognosis of heart failure when they were expressed at low levels. We performed an integrated WGCNA and validated that TLR2 and CD14 were closely associated with the severity of coronary artery disease, plaque instability and the prognosis of heart failure after myocardial infarction.
Collapse
Affiliation(s)
- Bin Qi
- Departments of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Jian-Hong Chen
- Departments of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Lin Tao
- Departments of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Chuan-Meng Zhu
- Departments of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Yong Wang
- Departments of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| | - Guo-Xiong Deng
- Departments of Cardiology, The First People's Hospital of Nanning, Nanning, China
| | - Liu Miao
- Departments of Cardiology, Liuzhou People's Hospital, Liuzhou, China
| |
Collapse
|
13
|
Truong R, Thankam FG, Agrawal DK. Immunological mechanisms underlying sterile inflammation in the pathogenesis of atherosclerosis: potential sites for intervention. Expert Rev Clin Immunol 2020; 17:37-50. [PMID: 33280442 DOI: 10.1080/1744666x.2020.1860757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Innate and adaptive immunity play a critical role in the underlying pathological mechanisms of atherosclerosis and potential target sites of sterile inflammation open opportunities to develop novel therapeutics. In response to oxidized LDL in the intimal layer, T cell subsets are recruited and activated at the site of atheroma to upregulate pro-atherogenic cytokines which exacerbate plaque formation instability.Areas covered: A systematic search of PubMed and the Web of Science was performed between January 2001- September 2020 and relevant articles in sterile inflammation and atherosclerosis were critically reviewed. The original information was collected on the interconnection between danger associated molecular patterns (DAMPs) as the mediators of sterile inflammation and the receptor complex of CD36-TLR4-TLR6 that primes and activates inflammasomes in the pathophysiology of atherosclerosis. Mediators of sterile inflammation are identified to target therapeutic strategies in the management of atherosclerosis.Expert opinion: Sterile inflammation via NLRP3 inflammasome is perpetuated by the activation of IL-1β and IL-18 and induction of pyroptosis resulting in the release of additional inflammatory cytokines and DAMPs. Challenges with current inhibitors of the NLRP3 inflammasome lie in the specificity, stability, and efficacy in targeting the NLRP3 inflammasome constituents without ameliorating upstream or downstream responses necessary for survival.
Collapse
Affiliation(s)
- Roland Truong
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
14
|
Dailing EA, Kilchrist KV, Tierney JW, Fletcher RB, Evans BC, Duvall CL. Modifying Cell Membranes with Anionic Polymer Amphiphiles Potentiates Intracellular Delivery of Cationic Peptides. ACS APPLIED MATERIALS & INTERFACES 2020; 12:50222-50235. [PMID: 33124813 PMCID: PMC9082340 DOI: 10.1021/acsami.0c13304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Rapid, facile, and noncovalent cell membrane modification with alkyl-grafted anionic polymers was sought as an approach to enhance intracellular delivery and bioactivity of cationic peptides. We synthesized a library of acrylic acid-based copolymers containing varying amounts of an amine-reactive pentafluorophenyl acrylate monomer followed by postpolymerization modification with a series of alkyl amines to afford precise control over the length and density of aliphatic alkyl side chains. This synthetic strategy enabled systematic investigation of the effect of the polymer structure on membrane binding, potentiation of peptide cell uptake, pH-dependent disruption of lipid bilayers for endosome escape, and intracellular bioavailability. A subset of these polymers exhibited pKa of ∼6.8, which facilitated stable membrane association at physiological pH and rapid, pH-dependent endosomal disruption upon endocytosis as quantified in Galectin-8-YFP reporter cells. Cationic cell penetrating peptide (CPP) uptake was enhanced up to 15-fold in vascular smooth muscle cells in vitro when peptide treatment was preceded by a 30-min pretreatment with lead candidate polymers. We also designed and implemented a new and highly sensitive assay for measuring the intracellular bioavailability of CPPs based on the NanoLuciferase (NanoLuc) technology previously developed for measuring intracellular protein-protein interactions. Using this split luciferase class of assay, polymer pretreatment enhanced intracellular delivery of the CPP-modified HiBiT peptide up to 30-fold relative to CPP-HiBiT without polymer pretreatment (p < 0.05). The overall structural analyses show that polymers containing 50:50 or 70:30 molar ratios of carboxyl groups to alkyl side chains of 6-8 carbons maximized peptide uptake, pH-dependent membrane disruption, and intracellular bioavailability and that this potentiation effect was maximized by pairing with CPPs with high cationic charge density. These results demonstrate a rapid, mild method for polymer modification of cell surfaces to potentiate intracellular delivery, endosome escape, and bioactivity of cationic peptides.
Collapse
Affiliation(s)
- Eric A Dailing
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - Kameron V Kilchrist
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - J William Tierney
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - R Brock Fletcher
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - Brian C Evans
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, PMB 351634, Nashville, Tennessee 37235, United States
| |
Collapse
|
15
|
Qaradakhi T, Gadanec LK, McSweeney KR, Abraham JR, Apostolopoulos V, Zulli A. The Anti-Inflammatory Effect of Taurine on Cardiovascular Disease. Nutrients 2020; 12:E2847. [PMID: 32957558 PMCID: PMC7551180 DOI: 10.3390/nu12092847] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/02/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022] Open
Abstract
Taurine is a non-protein amino acid that is expressed in the majority of animal tissues. With its unique sulfonic acid makeup, taurine influences cellular functions, including osmoregulation, antioxidation, ion movement modulation, and conjugation of bile acids. Taurine exerts anti-inflammatory effects that improve diabetes and has shown benefits to the cardiovascular system, possibly by inhibition of the renin angiotensin system. The beneficial effects of taurine are reviewed.
Collapse
Affiliation(s)
- Tawar Qaradakhi
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia; (L.K.G.); (K.R.M.); (J.R.A.); (V.A.); (A.Z.)
| | | | | | | | | | | |
Collapse
|
16
|
Jaén RI, Val-Blasco A, Prieto P, Gil-Fernández M, Smani T, López-Sendón JL, Delgado C, Boscá L, Fernández-Velasco M. Innate Immune Receptors, Key Actors in Cardiovascular Diseases. JACC Basic Transl Sci 2020; 5:735-749. [PMID: 32760860 PMCID: PMC7393405 DOI: 10.1016/j.jacbts.2020.03.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the industrialized world. Most CVDs are associated with increased inflammation that arises mainly from innate immune system activation related to cardiac damage. Sustained activation of the innate immune system frequently results in maladaptive inflammatory responses that promote cardiovascular dysfunction and remodeling. Much research has focused on determining whether some mediators of the innate immune system are potential targets for CVD therapy. The innate immune system has specific receptors-termed pattern recognition receptors (PRRs)-that not only recognize pathogen-associated molecular patterns, but also sense danger-associated molecular signals. Activation of PRRs triggers the inflammatory response in different physiological systems, including the cardiovascular system. The classic PRRs, toll-like receptors (TLRs), and the more recently discovered nucleotide-binding oligomerization domain-like receptors (NLRs), have been recently proposed as key partners in the progression of several CVDs (e.g., atherosclerosis and heart failure). The present review discusses the key findings related to the involvement of TLRs and NLRs in the progression of several vascular and cardiac diseases, with a focus on whether some NLR subtypes (nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing receptor 3 and nucleotide-binding oligomerization domain-containing protein 1) can be candidates for the development of new therapeutic strategies for several CVDs.
Collapse
Key Words
- AMI, acute myocardial infarction
- CARD, caspase activation and recruitment domain
- CVD, cardiovascular disease
- Ca2+, calcium ion
- DAMPs, danger-associated molecular patterns
- DAP, D-glutamyl-meso-diaminopimelic acid
- ER, endoplasmic reticulum
- HF, heart failure
- I/R, ischemia/reperfusion
- IL, interleukin
- MAPK, mitogen-activated protein kinase
- NF-κB, nuclear factor κ-light-chain-enhancer of activated B cells
- NLR, nucleotide-binding oligomerization domain-like receptors
- NLRP, nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing receptor
- NLRP3
- NOD, Nucleotide-binding oligomerization domain-containing protein
- NOD1
- PAMP, pathogen-associated molecular pattern
- ROS, reactive oxygen species
- SR, sarcoplasmic reticulum
- TLR, toll-like receptor
- cardiovascular disease
- innate immune system
- nucleotide-binding oligomerization domain-like receptors
- toll-like receptors
Collapse
Affiliation(s)
- Rafael I. Jaén
- Biomedical Research Institute “Alberto Sols” CSIC-UAM, Madrid, Spain
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | - Almudena Val-Blasco
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Patricia Prieto
- Biomedical Research Institute “Alberto Sols” CSIC-UAM, Madrid, Spain
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Pharmacology, Pharmacognosy and Botany department, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Dr. Patricia Prieto, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain. @IIBmCSICUAM
| | - Marta Gil-Fernández
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Tarik Smani
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain
| | - José Luis López-Sendón
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Servicio de Cardiología, Hospital Universitario La Paz, Madrid, Spain
| | - Carmen Delgado
- Biomedical Research Institute “Alberto Sols” CSIC-UAM, Madrid, Spain
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | - Lisardo Boscá
- Biomedical Research Institute “Alberto Sols” CSIC-UAM, Madrid, Spain
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | - María Fernández-Velasco
- CIBER Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Address for correspondence: Dr. María Fernández-Velasco, Instituto de Investigación Hospital la Paz, IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain. @IdipazScience@CIBER_CV@Mfvlorenzo
| |
Collapse
|
17
|
Zhou Y, Little PJ, Downey L, Afroz R, Wu Y, Ta HT, Xu S, Kamato D. The Role of Toll-like Receptors in Atherothrombotic Cardiovascular Disease. ACS Pharmacol Transl Sci 2020; 3:457-471. [PMID: 32566912 PMCID: PMC7296543 DOI: 10.1021/acsptsci.9b00100] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are dominant components of the innate immune system. Activated by both pathogen-associated molecular patterns and damage-associated molecular patterns, TLRs underpin the pathology of numerous inflammation related diseases that include not only immune diseases, but also cardiovascular disease (CVD), diabetes, obesity, and cancers. Growing evidence has demonstrated that TLRs are involved in multiple cardiovascular pathophysiologies, such as atherosclerosis and hypertension. Specifically, a trial called the Canakinumab Anti-inflammatory Thrombosis Outcomes Study showed the use of an antibody that neutralizes interleukin-1β, reduces the recurrence of cardiovascular events, demonstrating inflammation as a therapeutic target and also the research value of targeting the TLR system in CVD. In this review, we provide an update of the interplay between TLR signaling, inflammatory mediators, and atherothrombosis, with an aim to identify new therapeutic targets for atherothrombotic CVD.
Collapse
Affiliation(s)
- Ying Zhou
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Peter J. Little
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| | - Liam Downey
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Rizwana Afroz
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Yuao Wu
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Hang T. Ta
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Suowen Xu
- Aab
Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | - Danielle Kamato
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| |
Collapse
|
18
|
Miao G, Zhao X, Wang B, Zhang L, Wang G, Zheng N, Liu J, Xu Z, Zhang L. TLR2/CXCR4 coassociation facilitatesChlamydia pneumoniaeinfection-induced atherosclerosis. Am J Physiol Heart Circ Physiol 2020; 318:H1420-H1435. [DOI: 10.1152/ajpheart.00011.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Toll-like receptor 2 (TLR2) and C-X-C motif chemokine receptor 4 (CXCR4) have both been shown to be involved in atherosclerosis. We demonstrate for the first time the presence of TLR2/CXCR4 coassociation during C. pneumoniae infection-induced atherosclerosis. Amazingly, blocking of both TLR2 and CXCR4 significantly retards and even almost reverses this infection-induced atherosclerosis. Our work reveals new mechanisms about C. pneumoniae infection-induced atherosclerosis and identifies potential new therapeutic targets for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Guolin Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xi Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Beibei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Guangyan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ningbo Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jingya Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhelong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
19
|
Liu W, Eczko JC, Otto M, Bajorat R, Vollmar B, Roesner JP, Wagner NM. Toll-like receptor 2-deficiency on bone marrow-derived cells augments vascular healing of murine arterial lesions. Life Sci 2019; 242:117189. [PMID: 31891724 DOI: 10.1016/j.lfs.2019.117189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 11/30/2022]
Abstract
AIMS Neointimal hyperplasia contributes to arterial restenosis after percutaneous transluminal coronary angioplasty or vascular surgery. Neointimal thickening after arterial injury is determined by inflammatory processes. We investigated the role of the innate immune receptor toll-like receptor 2 (TLR2) in neointima formation after arterial injury in mice. MATERIALS AND METHODS Carotid artery injury was induced by 10% ferric chloride in C57Bl/6J wild type (WT), TLR2 deficient (B6.129-Tlr2tm1Kir/J, TLR2-/-) and WT mice treated with a TLR2 blocking antibody. 21 days after injury, carotid arteries were assessed histomorphometrically and for smooth muscle cell (SMC) content. To identify the contribution of circulating cells in mediating the effects of TLR2-deficiency, arterial injury was induced in WT/TLR2-/--chimeric mice and the paracrine modulation of bone marrow-derived cells from WT and TLR2-/- on SMC migration compared in vitro. KEY FINDINGS TLR2-/- mice and WT mice treated with TLR2 blocking antibodies exhibited reduced neointimal thickening (23.7 ± 4.2 and 6.5 ± 3.0 vs. 43.1 ± 5.9 μm, P < 0.05 and P < 0.01), neointimal area (5491 ± 1152 and 315 ± 76.7 vs. 13,756 ± 2627 μm2, P < 0.05 and P < 0.01) and less luminal stenosis compared to WT mice (8.5 ± 1.6 and 5.0 ± 1.3 vs. 22.4 ± 2.2%, both P < 0.001n = 4-8 mice/group). The phenotypes of TLR2-/- vs. WT mice were completely reverted in WT/TLR2-/- bone marrow chimeric mice (5.9 ± 1.5 μm neointimal thickness, 874.2 ± 290.2 μm2 neointima area and 2.7 ± 0.6% luminal stenoses in WT mice transplanted with TLR2-/- bone marrow vs. 23.6 ± 5.1 μm, 3555 ± 511 μm2 and 12.0 ± 1.3% in WT mice receiving WT bone marrow, all P < 0.05, n = 6/group). Neointimal lesions of WT and WT mice transplanted with TLR2-/- bone marrow chimeric mice showed increased numbers of SMC (10.8 ± 1.4 and 12.6 ± 1.4 vs. 3.8 ± 0.9 in TLR2-/- and 3.5 ± 1.1 cells in WT mice transplanted with TLR2-/- bone marrow, all P < 0.05, n = 6). WT bone marrow cells stimulated SMC migration more than TLR2-deficient bone marrow cells (1.7 ± 0.05 vs. 1.3 ± 0.06-fold, P < 0.05, n = 7) and this effect was aggravated by TLR2 stimulation and diminished by TLR2 blockade (1.1 ± 0.03-fold after stimulation with TLR2 agonists and 0.8 ± 0.02-fold after TLR2 blockade vs. control treated cells defined as 1.0, P < 0.05, n = 7). SIGNIFICANCE TLR2-deficiency on hematopoietic but not vessel wall resident cells augments vascular healing after arterial injury. Pharmacological blockade of TLR2 may thus be a promising therapeutic option to improve vessel patency after iatrogenic arterial injury.
Collapse
Affiliation(s)
- W Liu
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - J-C Eczko
- Department of Anesthesia and Intensive Care, University Medical Center Rostock, Rostock, Germany
| | - M Otto
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - R Bajorat
- Department of Anesthesia and Intensive Care, University Medical Center Rostock, Rostock, Germany
| | - B Vollmar
- Institute for Experimental Surgery, University Medical Center Rostock, Rostock, Germany
| | - J-P Roesner
- Department of Anesthesia and Intensive Care, University Medical Center Rostock, Rostock, Germany
| | - N-M Wagner
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany.
| |
Collapse
|
20
|
Hu L, Huang Z, Ishii H, Wu H, Suzuki S, Inoue A, Kim W, Jiang H, Li X, Zhu E, Piao L, Zhao G, Lei Y, Okumura K, Shi GP, Murohara T, Kuzuya M, Cheng XW. PLF-1 (Proliferin-1) Modulates Smooth Muscle Cell Proliferation and Development of Experimental Intimal Hyperplasia. J Am Heart Assoc 2019; 8:e005886. [PMID: 31838975 PMCID: PMC6951060 DOI: 10.1161/jaha.117.005886] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Although apoptosis and cell proliferation have been extensively investigated in atherosclerosis and restenosis postinjury, the communication between these 2 cellular events has not been evaluated. Here, we report an inextricable communicative link between apoptosis and smooth muscle cell proliferation in the promotion of vascular remodeling postinjury. Methods and Results Cathepsin K-mediated caspase-8 maturation is a key initial step for oxidative stress-induced smooth muscle cell apoptosis. Apoptotic cells generate a potential growth-stimulating signal to facilitate cellular mass changes in response to injury. One downstream mediator that cathepsin K regulates is PLF-1 (proliferin-1), which can potently stimulate growth of surviving neighboring smooth muscle cells through activation of PI3K/Akt/p38MAPK (phosphatidylinositol 3-kinase/protein kinase B/p38 mitogen-activated protein kinase)-dependent and -independent mTOR (mammalian target of rapamycin) signaling cascades. We observed that cathepsin K deficiency substantially mitigated neointimal hyperplasia by reduction of Toll-like receptor-2/caspase-8-mediated PLF-1 expression. Interestingly, PLF-1 blocking, with its neutralizing antibody, suppressed neointima formation and remodeling in response to injury in wild-type mice. Contrarily, administration of recombinant mouse PLF-1 accelerated injury-induced vascular actions. Conclusions This is the first study detailing PLF-1 as a communicator between apoptosis and proliferation during injury-related vascular remodeling and neointimal hyperplasia. These data suggested that apoptosis-driven expression of PLF-1 is thus a novel target for treatment of apoptosis-based hyperproliferative disorders.
Collapse
Affiliation(s)
- Lina Hu
- Department of Public Health Guilin Medical College Guilin Guangxi China.,Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Zhe Huang
- Department of Neurology Occupational and Environmental Health Kitakyushu Hukuoka Japan
| | - Hideki Ishii
- Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Hongxian Wu
- Department of Cardiology Shanghai General Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Susumu Suzuki
- Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Aiko Inoue
- Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,Institute of Innovation for Future Society Nagoya University Nagoya Japan
| | - Weon Kim
- Division of Cardiology Department of Internal Medicine Kyung Hee University Seoul South Korea
| | - Haiying Jiang
- Department of Physiology and Pathophysiology Yanbian University School of Medicine Yanji Jinlin China
| | - Xiang Li
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
| | - Enbo Zhu
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
| | - Limei Piao
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan
| | - Guangxian Zhao
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
| | - Yanna Lei
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China
| | - Kenji Okumura
- Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Guo-Ping Shi
- Department of Cardiovascular Medicine Brigham and Women's Hospital and Harvard Medical School Boston MA
| | - Toyoaki Murohara
- Department of Cardiology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Masafumi Kuzuya
- Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,Institute of Innovation for Future Society Nagoya University Nagoya Japan
| | - Xian Wu Cheng
- Department of Cardiology/Hypertension and Heart Center Yanbian University Hospital Yanji Jilin China.,Department of Community & Geriatrics Nagoya University Graduate School of Medicine Nagoya Japan.,Division of Cardiology Department of Internal Medicine Kyung Hee University Seoul South Korea.,Institute of Innovation for Future Society Nagoya University Nagoya Japan
| |
Collapse
|
21
|
Vanadium Derivative Exposure Promotes Functional Alterations of VSMCs and Consequent Atherosclerosis via ROS/p38/NF-κB-Mediated IL-6 Production. Int J Mol Sci 2019; 20:ijms20246115. [PMID: 31817202 PMCID: PMC6940940 DOI: 10.3390/ijms20246115] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 12/22/2022] Open
Abstract
Vanadium is a transition metal widely distributed in the Earth’s crust, and is a major contaminant in fossil fuels. Its pathological effect and regulation in atherosclerosis remain unclear. We found that intranasal administration of the vanadium derivative NaVO3 significantly increased plasma and urinary vanadium levels and induced arterial lipid accumulation and atherosclerotic lesions in apolipoprotein E-deficient knockout mice (ApoE−/−) murine aorta compared to those in vehicle-exposed mice. This was accompanied by an increase in plasma reactive oxygen species (ROS) and interleukin 6 (IL-6) levels and a decrease in the vascular smooth muscle cell (VSMC) differentiation marker protein SM22α in the atherosclerotic lesions. Furthermore, exposure to NaVO3 or VOSO4 induced cytosolic ROS generation and IL-6 production in VSMCs and promoted VSMC synthetic differentiation, migration, and proliferation. The anti-oxidant N-acetylcysteine (NAC) not only suppresses IL-6 production and VSMC pathological responses including migration and proliferation but also prevents atherosclerosis in ApoE−/− mice. Inhibition experiments with NAC and pharmacological inhibitors demonstrated that NaVO3-induced IL-6 production is signaled by ROS-triggered p38-mediated NF-κB-dependent pathways. Neutralizing anti-IL-6 antibodies impaired NaVO3-mediated VSMC migration and proliferation. We concluded that NaVO3 exposure activates the ROS-triggering p38 signaling to selectively induce NF-κB-mediated IL-6 production. These signaling pathways induce VSMC synthetic differentiation, migration, and proliferation, leading to lipid accumulation and atherosclerosis.
Collapse
|
22
|
Lee GL, Yeh CC, Wu JY, Lin HC, Wang YF, Kuo YY, Hsieh YT, Hsu YJ, Kuo CC. TLR2 Promotes Vascular Smooth Muscle Cell Chondrogenic Differentiation and Consequent Calcification via the Concerted Actions of Osteoprotegerin Suppression and IL-6–Mediated RANKL Induction. Arterioscler Thromb Vasc Biol 2019; 39:432-445. [DOI: 10.1161/atvbaha.118.311874] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Objective—
Vascular smooth muscle cell (VSMC) transformation to an osteochondrogenic phenotype is an initial step toward arterial calcification, which is highly correlated with cardiovascular disease–related morbidity and mortality. TLR2 (Toll-like receptor 2) plays a pathogenic role in the development of vascular diseases, but its regulation in calcification of arteries and VSMCs remains unclear. We postulate that TLR2-mediated inflammation participates in mediating atherosclerotic arterial calcification and VSMC calcification.
Approach and Results—
We found that
ApoE
−/−
Tlr2
−/−
genotype in mice suppressed high-fat diet–induced atherosclerotic plaques formation during initiation but progressively lost its preventative capacity, compared with
ApoE
−/−
mice. However, TLR2 deficiency prohibited high-fat diet–induced advanced atherosclerotic calcification, chondrogenic metaplasia, and OPG (osteoprotegerin) downregulation in the calcified lesions. Incubation of VSMCs in a calcifying medium revealed that TLR2 agonists significantly increased VSMC calcification and chondrogenic differentiation. Furthermore, TLR2 deficiency suppressed TLR2 agonist–mediated VSMC chondrogenic differentiation and consequent calcification, which were triggered via the concerted actions of IL (interleukin)-6–mediated RANKL (receptor activator of nuclear factor κB ligand) induction and OPG suppression. Inhibition experiments with pharmacological inhibitors demonstrated that IL-6–mediated RANKL induction is signaled by p38 and ERK1/2 (extracellular signal-regulated kinase 1/2) pathways, whereas the OPG is suppressed via NF-κB (nuclear factor κB) dependent signaling mediated by ERK1/2.
Conclusions—
We concluded that on ligand binding, TLR2 activates p38 and ERK1/2 signaling to selectively modulate the upregulation of IL-6–mediated RANKL and downregulation of OPG. These signaling pathways act in concert to induce chondrogenic transdifferentiation of VSMCs, which in turn leads to vascular calcification during the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Guan-Lin Lee
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Maioli, Taiwan (G.-L.L., C.-C.Y., J.-Y.W., H.-C.L., Y.-F.W., Y.-Y.K., Y.-T.H., C.-C.K.)
| | - Chang-Ching Yeh
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Maioli, Taiwan (G.-L.L., C.-C.Y., J.-Y.W., H.-C.L., Y.-F.W., Y.-Y.K., Y.-T.H., C.-C.K.)
- Graduate Institutes of Life Sciences, National Defense Medical Center, Taipei, Taiwan (C.-C.Y., Y.-J.H., C.-C.K.)
| | - Jing-Yiing Wu
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Maioli, Taiwan (G.-L.L., C.-C.Y., J.-Y.W., H.-C.L., Y.-F.W., Y.-Y.K., Y.-T.H., C.-C.K.)
| | - Hui-Chen Lin
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Maioli, Taiwan (G.-L.L., C.-C.Y., J.-Y.W., H.-C.L., Y.-F.W., Y.-Y.K., Y.-T.H., C.-C.K.)
| | - Yi-Fu Wang
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Maioli, Taiwan (G.-L.L., C.-C.Y., J.-Y.W., H.-C.L., Y.-F.W., Y.-Y.K., Y.-T.H., C.-C.K.)
| | - Ya-Yi Kuo
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Maioli, Taiwan (G.-L.L., C.-C.Y., J.-Y.W., H.-C.L., Y.-F.W., Y.-Y.K., Y.-T.H., C.-C.K.)
| | - Yi-Ting Hsieh
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Maioli, Taiwan (G.-L.L., C.-C.Y., J.-Y.W., H.-C.L., Y.-F.W., Y.-Y.K., Y.-T.H., C.-C.K.)
| | - Yu-Juei Hsu
- Division of Nephrology and Department of Medicine, Tri-Service General Hospital, Taipei, Taiwan (Y.-J.H)
- Graduate Institutes of Life Sciences, National Defense Medical Center, Taipei, Taiwan (C.-C.Y., Y.-J.H., C.-C.K.)
| | - Cheng-Chin Kuo
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Maioli, Taiwan (G.-L.L., C.-C.Y., J.-Y.W., H.-C.L., Y.-F.W., Y.-Y.K., Y.-T.H., C.-C.K.)
- Graduate Institutes of Life Sciences, National Defense Medical Center, Taipei, Taiwan (C.-C.Y., Y.-J.H., C.-C.K.)
- Metabolomic Research Center and Graduate Institute of Basic Medical Science China Medical University Hospital, Taichung, Taiwan (C.-C.K.)
| |
Collapse
|
23
|
C1q and TNF related protein 1 regulates expression of inflammatory genes in vascular smooth muscle cells. Genes Genomics 2018; 41:397-406. [PMID: 30474828 DOI: 10.1007/s13258-018-0770-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/19/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND C1q and TNF related protein 1 (C1QTNF1) is known to be associated with coronary artery diseases. However, the molecular function of C1QTNF1 on the vascular smooth muscles remains to be investigated. OBJECTIVE This study was therefore undertaken to investigate the effect of C1QTNF1 on gene expression of human smooth muscle cells and to reveal potential molecular mechanisms mediated by C1QTNF1. METHODS Vascular smooth muscle cells were incubated with recombinant C1QTNF1 for 16 h, followed by determining any change in mRNA expressions by Affymetrix genechip. Gene ontology (GO), KEGG pathway, and protein-protein interaction (PPI) network were analyzed in differentially expressed genes. In addition, validation of microarray data was performed using quantitative real-time PCR. RESULTS The mRNA expressions of annotated 74 genes were significantly altered after incubation with recombinant C1QTNF1; 41 genes were up-regulated and 33 down-regulated. The differentially expressed genes were enriched in biological processes and KEGG pathways associated with inflammatory responses. In the PPI network analysis, IL-6, CCL2, and ICAM1 were identified as potential key genes with relatively high degree. The cluster analysis in the PPI network identified a significant module composed of upregulated genes, such as IL-6, CCL2, NFKBIA, SOD2, and ICAM1. The quantitative real-time PCR results of potential key genes were consistent with microarray data. CONCLUSION The results in the present study provide insights on the effects of C1QTNF1 on gene expression of smooth muscle cells. We believe our findings will help to elucidate the molecular mechanisms regarding the functions of C1QTNF1 on smooth muscle cells in inflammatory diseases.
Collapse
|
24
|
RAGE and TLRs as Key Targets for Antiatherosclerotic Therapy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7675286. [PMID: 30225265 PMCID: PMC6129363 DOI: 10.1155/2018/7675286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/08/2018] [Indexed: 02/08/2023]
Abstract
Receptor for advanced glycation end-products (RAGE) and toll-like receptors (TLRs) are the key factors indicating a danger to the organism. They recognize the microbial origin pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). The primary response induced by PAMPs or DAMPs is inflammation. Excessive stimulation of the innate immune system occurs in arterial wall with the participation of effector cells. Persistent adaptive responses can also cause tissue damage and disease. However, inflammation mediated by the molecules innate responses is an important way in which the adaptive immune system protects us from infection. The specific detection of PAMPs and DAMPs by host receptors drives a cascade of signaling that converges at nuclear factor-κB (NF-κB) and interferon regulatory factors (IRFs) and induces the secretion of proinflammatory cytokines, type I interferon (IFN), and chemokines, which promote direct killing of the pathogen. Therefore, signaling of these receptors' pathways also appear to present new avenue for the modulation of inflammatory responses and to serve as potential novel therapeutic targets for antiatherosclerotic therapy.
Collapse
|
25
|
Sterpetti AV, Borrelli V, Cucina A, Ventura M. Cross talk between TGF beta and TNF alfa in regression of myointimal hyperplasia. J Surg Res 2017; 220:6-11. [PMID: 29180213 DOI: 10.1016/j.jss.2017.06.087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/05/2017] [Accepted: 06/29/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND The phenomena involved in regression of arterial myointimal hyperplasia have not been analyzed in detail. MATERIALS AND METHODS In 24 Lewis rats, a 1-cm-long venous graft, obtained from syngenic Lewis rats, was implanted in the infrarenal aorta. After 4 wk, the grafts were removed and analyzed using scanning electron microscopy and histochemistry. The grafts showed evidence of myointimal hyperplasia; 16 of these explanted grafts were reimplanted in the vein circulation of syngenic Lewis rats. These grafts were harvested 2 wk (8 animals) and 8 wk (8 animals) later, showing complete regression of myointimal hyperplasia. RESULTS Regression of experimental myointimal hyperplasia was correlated with the simultaneous and complementary action of Transforming Growth Factor beta and Tumor Necrosis Factor alfa. Inflammatory cytokines (IL1, IL2, and IL6) inhibit Tumor Necrosis Factor alfa-induced apoptosis. CONCLUSIONS Regression of myointimal hyperplasia is an active process, which implies the action of several inhibitory factors. The analysis of these phenomena can lead to new therapeutic approaches to prevent myointimal hyperplasia progression.
Collapse
Affiliation(s)
| | - Valeria Borrelli
- Istituto Pietro Valdoni, University of Rome Sapienza, Rome, Italy
| | | | - Marco Ventura
- Department of Vascular Surgery, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW The present review explores the mechanisms of superficial intimal erosion, a common cause of thrombotic complications of atherosclerosis. RECENT FINDINGS Human coronary artery atheroma that give rise to thrombosis because of erosion differ diametrically from those associated with fibrous cap rupture. Eroded lesions characteristically contain few inflammatory cells, abundant extracellular matrix, and neutrophil extracellular traps (NETs). Innate immune mechanisms such as engagement of Toll-like receptor 2 (TLR2) on cultured endothelial cells can impair their viability, attachment, and ability to recover a wound. Hyaluronan fragments may serve as endogenous TLR2 ligands. Mouse experiments demonstrate that flow disturbance in arteries with neointimas tailored to resemble features of human eroded plaques disturbs endothelial cell barrier function, impairs endothelial cell viability, recruits neutrophils, and provokes endothelial cells desquamation, NET formation, and thrombosis in a TLR2-dependent manner. SUMMARY Mechanisms of erosion have received much less attention than those that provoke plaque rupture. Intensive statin treatment changes the characteristic of plaques that render them less susceptible to rupture. Thus, erosion may contribute importantly to the current residual burden of risk. Understanding the mechanisms of erosion may inform the development and deployment of novel therapies to combat the remaining atherothrombotic risk in the statin era.
Collapse
Affiliation(s)
- Thibaut Quillard
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Grégory Franck
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Thomas Mawson
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Eduardo Folco
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Peter Libby
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
27
|
Xu X, Wang B, Ren C, Hu J, Greenberg DA, Chen T, Xie L, Jin K. Age-related Impairment of Vascular Structure and Functions. Aging Dis 2017; 8:590-610. [PMID: 28966804 PMCID: PMC5614324 DOI: 10.14336/ad.2017.0430] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/30/2017] [Indexed: 12/12/2022] Open
Abstract
Among age-related diseases, cardiovascular and cerebrovascular diseases are major causes of death. Vascular dysfunction is a key characteristic of these diseases wherein age is an independent and essential risk factor. The present work will review morphological alterations of aging vessels in-depth, which includes the discussion of age-related microvessel loss and changes to vasculature involving the capillary basement membrane, intima, media, and adventitia as well as the accompanying vascular dysfunctions arising from these alterations.
Collapse
Affiliation(s)
- Xianglai Xu
- 1Zhongshan Hospital, Fudan University, Shanghai 200032, China.,2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Brian Wang
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Changhong Ren
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.,4Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University. Beijing, China
| | - Jiangnan Hu
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | | | - Tianxiang Chen
- 6Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Liping Xie
- 3Department of Urology, the First Affiliated Hospital, Zhejiang University, Zhejiang Province, China
| | - Kunlin Jin
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| |
Collapse
|
28
|
Lacolley P, Regnault V, Segers P, Laurent S. Vascular Smooth Muscle Cells and Arterial Stiffening: Relevance in Development, Aging, and Disease. Physiol Rev 2017; 97:1555-1617. [DOI: 10.1152/physrev.00003.2017] [Citation(s) in RCA: 332] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/15/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
The cushioning function of large arteries encompasses distension during systole and recoil during diastole which transforms pulsatile flow into a steady flow in the microcirculation. Arterial stiffness, the inverse of distensibility, has been implicated in various etiologies of chronic common and monogenic cardiovascular diseases and is a major cause of morbidity and mortality globally. The first components that contribute to arterial stiffening are extracellular matrix (ECM) proteins that support the mechanical load, while the second important components are vascular smooth muscle cells (VSMCs), which not only regulate actomyosin interactions for contraction but mediate also mechanotransduction in cell-ECM homeostasis. Eventually, VSMC plasticity and signaling in both conductance and resistance arteries are highly relevant to the physiology of normal and early vascular aging. This review summarizes current concepts of central pressure and tensile pulsatile circumferential stress as key mechanical determinants of arterial wall remodeling, cell-ECM interactions depending mainly on the architecture of cytoskeletal proteins and focal adhesion, the large/small arteries cross-talk that gives rise to target organ damage, and inflammatory pathways leading to calcification or atherosclerosis. We further speculate on the contribution of cellular stiffness along the arterial tree to vascular wall stiffness. In addition, this review provides the latest advances in the identification of gene variants affecting arterial stiffening. Now that important hemodynamic and molecular mechanisms of arterial stiffness have been elucidated, and the complex interplay between ECM, cells, and sensors identified, further research should study their potential to halt or to reverse the development of arterial stiffness.
Collapse
Affiliation(s)
- Patrick Lacolley
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Véronique Regnault
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Patrick Segers
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Stéphane Laurent
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| |
Collapse
|
29
|
Li X, Lu J, Kan Q, Li X, Fan Q, Li Y, Huang R, Slipicevic A, Dong HP, Eide L, Wang J, Zhang H, Berge V, Goscinski MA, Kvalheim G, Nesland JM, Suo Z. Metabolic reprogramming is associated with flavopiridol resistance in prostate cancer DU145 cells. Sci Rep 2017; 7:5081. [PMID: 28698547 PMCID: PMC5506068 DOI: 10.1038/s41598-017-05086-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 05/24/2017] [Indexed: 01/19/2023] Open
Abstract
Flavopiridol (FP) is a pan-cyclin dependent kinase inhibitor, which shows strong efficacy in inducing cancer cell apoptosis. Although FP is potent against most cancer cells in vitro, unfortunately it proved less efficacious in clinical trials in various aggressive cancers. To date, the molecular mechanisms of the FP resistance are mostly unknown. Here, we report that a small fraction human prostate cancer DU145 cells can survive long-term FP treatment and emerge as FP-resistant cells (DU145FP). These DU145FP cells show accumulated mitochondrial lesions with stronger glycolytic features, and they proliferate in slow-cycling and behave highly migratory with strong anti-apoptotic potential. In addition, the cells are less sensitive to cisplatin and docetaxel-induced apoptotic pressure, and over-express multiple stem cell associated biomarkers. Our studies collectively uncover for the first time that FP-resistant prostate cancer cells show metabolic remodeling, and the metabolic plasticity might be required for the FP resistance-associated cancer cell stemness up-regulation.
Collapse
Affiliation(s)
- Xiaoran Li
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
- Department of Pathology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway
| | - Jie Lu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Quancheng Kan
- Department of Clinical Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xiaoli Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qiong Fan
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0316, Norway
| | - Yaqing Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruixia Huang
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
| | - Ana Slipicevic
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Hiep Phuc Dong
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Lars Eide
- Department of Medical Biochemistry, University of Oslo and Oslo University Hospital, Oslo, 0372, Norway
| | - Junbai Wang
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Hongquan Zhang
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Peking University Health Science Center, Beijing, 100191, China
| | - Viktor Berge
- Department of Urology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Mariusz Adam Goscinski
- Departments of Surgery, The Norwegian Radium Hospital, Oslo University Hospital, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0379, Norway
| | - Gunnar Kvalheim
- Department of Cell Therapy, Cancer Institute, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
| | - Jahn M Nesland
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway
- Department of Pathology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway
| | - Zhenhe Suo
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, 0379, Norway.
- Department of Pathology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway.
| |
Collapse
|
30
|
Sterpetti AV, Cucina A, Borrelli V, Ventura M. Inflammation and myointimal hyperplasia. Correlation with hemodynamic forces. Vascul Pharmacol 2017; 117:1-6. [PMID: 28687339 DOI: 10.1016/j.vph.2017.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/29/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The aim of our study was to correlate flow dynamics and the release of inflammatory cytokines Interleukin 1, 2, 6, TNF (Tumour Necrosis Factor) alfa, both in vitro and in vivo. MATERIALS AND METHODS Endothelial cells were exposed to laminar flow (6dyne/cm2) in an in vitro circulatory system and the release of Interleukin 1, 2, 6 and TNF alfa was quantified by ELISA. Interleukin 1, 2, 6 and TNF alfa release was also assessed in vein grafts implanted in the arterial circulation of Lewis rats. Arterial vein grafts were explanted at different time intervals from 3days to 12weeks after surgery. Vein grafts implanted in the arterial circulation for 4weeks, were re implanted in the venous circulation of syngenic Lewis rats, and the release of Interleukin 1, 2, 6 and TNF alfa, was assessed in an organ culture. Six vein grafts (4 occluded, 2 patent) implanted in humans as femorodistal bypass were examined for the presence of myointimal hyperplasia and perigraft inflammatory cells. RESULTS In vitro, endothelial cells exposed to laminar flow released an increased amount of Interleukin 1, 2, 6 and TNF alfa in comparison to endothelial cells not exposed to flow. In experimental vein grafts implanted in the arterial circulation there was an increased release of inflammatory cytokines associated to inflammatory changes in the adventitia. Once the vein grafts were re implanted in the venous circulation, the release of these cytokines diminished, while the inflammatory changes in the adventitia regressed. CONCLUSIONS Increased shear stress induces release of cytokines and inflammatory changes in the adventitia. These inflammatory changes can contribute to plaque progression and to un stable plaque. These findings support the use of anti-inflammatory therapy in patients prone to develop atherosclerosis and in those who had arterial reconstructive surgery.
Collapse
|
31
|
Seidl SE, Pessolano LG, Bishop CA, Best M, Rich CB, Stone PJ, Schreiber BM. Toll-like receptor 2 activation and serum amyloid A regulate smooth muscle cell extracellular matrix. PLoS One 2017; 12:e0171711. [PMID: 28257481 PMCID: PMC5336220 DOI: 10.1371/journal.pone.0171711] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/24/2017] [Indexed: 11/19/2022] Open
Abstract
Smooth muscle cells contribute to extracellular matrix remodeling during atherogenesis. De-differentiated, synthetic smooth muscle cells are involved in processes of migration, proliferation and changes in expression of extracellular matrix components, all of which contribute to loss of homeostasis accompanying atherogenesis. Elevated levels of acute phase proteins, including serum amyloid A (SAA), are associated with an increased risk for atherosclerosis. Although infection with periodontal and respiratory pathogens via activation of inflammatory cell Toll-like receptor (TLR)2 has been linked to vascular disease, little is known about smooth muscle cell TLR2 in atherosclerosis. This study addresses the role of SAA and TLR2 activation on smooth muscle cell matrix gene expression and insoluble elastin accumulation. Cultured rat aortic smooth muscle cells were treated with SAA or TLR2 agonists and the effect on expression of matrix metallopeptidase 9 (MMP9) and tropoelastin studied. SAA up-regulated MMP9 expression. Tropoelastin is an MMP9 substrate and decreased tropoelastin levels in SAA-treated cells supported the concept of extracellular matrix remodeling. Interestingly, SAA-induced down-regulation of tropoelastin was not only evident at the protein level but at the level of gene transcription as well. Contributions of proteasomes, nuclear factor κ B and CCAAT/enhancer binding protein β on regulation of MMP9 vs. tropoleastin expression were revealed. Effects on Mmp9 and Eln mRNA expression persisted with long-term SAA treatment, resulting in decreased insoluble elastin accumulation. Interestingly, the SAA effects were TLR2-dependent and TLR2 activation by bacterial ligands also induced MMP9 expression and decreased tropoelastin expression. These data reveal a novel mechanism whereby SAA and/or infection induce changes in vascular elastin consistent with atherosclerosis.
Collapse
Affiliation(s)
- Stephanie E. Seidl
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lawrence G. Pessolano
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Christopher A. Bishop
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Michael Best
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Celeste B. Rich
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Phillip J. Stone
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Barbara M. Schreiber
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
32
|
The Role of TLR2, TLR4, and TLR9 in the Pathogenesis of Atherosclerosis. Int J Inflam 2016; 2016:1532832. [PMID: 27795867 PMCID: PMC5067326 DOI: 10.1155/2016/1532832] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/05/2016] [Accepted: 09/15/2016] [Indexed: 02/07/2023] Open
Abstract
Toll-like receptors (TLRs) are key players in the pathogenesis of inflammatory conditions including coronary arterial disease (CAD). They are expressed by a variety of immune cells where they recognize pathogen-associated molecular patterns (PAMPs). TLRs recruit adaptor molecules, including myeloid differentiation primary response protein (MYD88) and TIRF-related adaptor protein (TRAM), to mediate activation of MAPKs and NF-kappa B pathways. They are associated with the development of CAD through various mechanisms. TLR4 is expressed in lipid-rich and atherosclerotic plaques. In TLR2−/− and TLR4−/− mice, atherosclerosis-associated inflammation was diminished. Moreover, TLR2 and TLR4 may induce expression of Wnt5a in advanced staged atheromatous plaque leading to activation of the inflammatory processes. TLR9 is activated by CpG motifs in nucleic acids and have been implicated in macrophage activation and the uptake of oxLDL from the circulation. Furthermore, TLR9 also stimulates interferon-α (INF-α) secretion and increases cytotoxic activity of CD4+ T-cells towards coronary artery tunica media smooth muscle cells. This review outlines the pathophysiological role of TLR2, TLR4, and TLR9 in atherosclerosis, focusing on evidence from animal models of the disease.
Collapse
|
33
|
Wise ES, Brophy CM. The Case for Endothelial Preservation via Pressure-Regulated Distension in the Preparation of Autologous Saphenous Vein Conduits in Cardiac and Peripheral Bypass Operations. Front Surg 2016; 3:54. [PMID: 27713879 PMCID: PMC5031700 DOI: 10.3389/fsurg.2016.00054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/12/2016] [Indexed: 12/04/2022] Open
Affiliation(s)
- Eric S Wise
- Department of Surgery, University of Maryland Medical Center , Baltimore, MD , USA
| | - Colleen M Brophy
- VA Tennessee Valley Healthcare System, Vanderbilt University, Nashville, TN, USA; Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
34
|
Goulopoulou S, McCarthy CG, Webb RC. Toll-like Receptors in the Vascular System: Sensing the Dangers Within. Pharmacol Rev 2016; 68:142-67. [PMID: 26721702 DOI: 10.1124/pr.114.010090] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Toll-like receptors (TLRs) are components of the innate immune system that respond to exogenous infectious ligands (pathogen-associated molecular patterns, PAMPs) and endogenous molecules that are released during host tissue injury/death (damage-associated molecular patterns, DAMPs). Interaction of TLRs with their ligands leads to activation of downstream signaling pathways that induce an immune response by producing inflammatory cytokines, type I interferons (IFN), and other inflammatory mediators. TLR activation affects vascular function and remodeling, and these molecular events prime antigen-specific adaptive immune responses. Despite the presence of TLRs in vascular cells, the exact mechanisms whereby TLR signaling affects the function of vascular tissues are largely unknown. Cardiovascular diseases are considered chronic inflammatory conditions, and accumulating data show that TLRs and the innate immune system play a determinant role in the initiation and development of cardiovascular diseases. This evidence unfolds a possibility that targeting TLRs and the innate immune system may be a novel therapeutic goal for these conditions. TLR inhibitors and agonists are already in clinical trials for inflammatory conditions such as asthma, cancer, and autoimmune diseases, but their study in the context of cardiovascular diseases is in its infancy. In this article, we review the current knowledge of TLR signaling in the cardiovascular system with an emphasis on atherosclerosis, hypertension, and cerebrovascular injury. Furthermore, we address the therapeutic potential of TLR as pharmacological targets in cardiovascular disease and consider intriguing research questions for future study.
Collapse
Affiliation(s)
- Styliani Goulopoulou
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - R Clinton Webb
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| |
Collapse
|
35
|
Lee GL, Wu JY, Tsai CS, Lin CY, Tsai YT, Lin CS, Wang YF, Yet SF, Hsu YJ, Kuo CC. TLR4-Activated MAPK-IL-6 Axis Regulates Vascular Smooth Muscle Cell Function. Int J Mol Sci 2016; 17:ijms17091394. [PMID: 27563891 PMCID: PMC5037674 DOI: 10.3390/ijms17091394] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/11/2016] [Accepted: 08/17/2016] [Indexed: 11/29/2022] Open
Abstract
Migration of vascular smooth muscle cells (VSMCs) into the intima is considered to be a vital event in the pathophysiology of atherosclerosis. Despite substantial evidence supporting the pathogenic role of Toll-like receptor 4 (TLR4) in the progression of atherogenesis, its function in the regulation of VSMC migration remains unclear. The goal of the present study was to elucidate the mechanism by which TLR4 regulates VSMC migration. Inhibitor experiments revealed that TLR4-induced IL-6 secretion and VSMC migration were mediated via the concerted actions of MyD88 and TRIF on the activation of p38 MAPK and ERK1/2 signaling. Neutralizing anti-IL-6 antibodies abrogated TLR4-driven VSMC migration and F-actin polymerization. Blockade of p38 MAPK or ERK1/2 signaling cascade inhibited TLR4 agonist-mediated activation of cAMP response element binding protein (CREB). Moreover, siRNA-mediated suppression of CREB production repressed TLR4-induced IL-6 production and VSMC migration. Rac-1 inhibitor suppressed TLR4-driven VSMC migration but not IL-6 production. Importantly, the serum level of IL-6 and TLR4 endogenous ligand HMGB1 was significantly higher in patients with coronary artery diseases (CAD) than in healthy subjects. Serum HMGB1 level was positively correlated with serum IL-6 level in CAD patients. The expression of both HMGB1 and IL-6 was clearly detected in the atherosclerotic tissue of the CAD patients. Additionally, there was a positive association between p-CREB and HMGB1 in mouse atherosclerotic tissue. Based on our findings, we concluded that, upon ligand binding, TLR4 activates p38 MAPK and ERK1/2 signaling through MyD88 and TRIF in VSMCs. These signaling pathways subsequently coordinate an additive augmentation of CREB-driven IL-6 production, which in turn triggers Rac-1-mediated actin cytoskeleton to promote VSMC migration.
Collapse
Affiliation(s)
- Guan-Lin Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
- Graduate Institutes of Life Sciences, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Jing-Yiing Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Yi-Ting Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Yi-Fu Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
- Department of Biochemistry, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan.
- Graduate Institutes of Life Sciences, National Defense Medical Center, Neihu, Taipei 11490, Taiwan.
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
36
|
Guo X, Yu L, Chen M, Wu T, Peng X, Guo R, Zhang B. miR-145 mediated the role of aspirin in resisting VSMCs proliferation and anti-inflammation through CD40. J Transl Med 2016; 14:211. [PMID: 27412561 PMCID: PMC4944465 DOI: 10.1186/s12967-016-0961-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/27/2016] [Indexed: 11/17/2022] Open
Abstract
Background Aspirin (ASA) is the most widely used medicine to prevent cardiovascular diseases; however, the mechanisms by which ASA exerts its anti-proliferative effect remain not fully understood. This study was designed to investigate whether miR-145 is involved in the regulation of vascular smooth muscle cells’ (VSMCs) proliferation and to determine the anti-inflammatory effects of ASA via its regulation of CD40 to provide a new theoretical basis for the pharmacological effect of aspirin. Methods The TNF-α induced proliferation model of VSMCs was divided into different groups with or without aspirin. Cell proliferation was detected by EdU; Real-time PCR was used to detect the mRNA expression of miR-145, CD40, and Calponin, a VSMCs differentiation marker gene. Western blot was used to detect the protein expression of CD40; ELISA was used to determine the concentrations of the inflammatory cytokine IL-6 in cell supernatants. Results The proliferation of VSMCs was stimulated by TNF-α and accompanied by decreased levels of Calponin. TNF-α also decreased the levels of miR-145 and increased the levels of CD40 and IL-6. Pretreatment with 20 μg/mL of aspirin in VSMCs could partially block the above-mentioned effects induced by TNF-α. The protective effects of ASA in VSMCs were reversed by a pretreatment with a miR-145 inhibitor. We also found that the expression of miR-145 in peripheral blood mononuclear cells in ischemic stroke patients was significantly increased after a 10-day treatment with aspirin. Conclusion miR-145 is involved in the anti-proliferation and anti-inflammation effects of aspirin on VSMCs by inhibiting the expression of CD40.
Collapse
Affiliation(s)
- Xin Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Lijin Yu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Min Chen
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Tian Wu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xiangdong Peng
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Ren Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
37
|
Wang YF, Hsu YJ, Wu HF, Lee GL, Yang YS, Wu JY, Yet SF, Wu KK, Kuo CC. Endothelium-Derived 5-Methoxytryptophan Is a Circulating Anti-Inflammatory Molecule That Blocks Systemic Inflammation. Circ Res 2016; 119:222-36. [DOI: 10.1161/circresaha.116.308559] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/05/2016] [Indexed: 01/10/2023]
Abstract
Rationale:
Systemic inflammation has emerged as a key pathophysiological process that induces multiorgan injury and causes serious human diseases. Endothelium is critical in maintaining cellular and inflammatory homeostasis, controlling systemic inflammation, and progression of inflammatory diseases. We postulated that endothelium produces and releases endogenous soluble factors to modulate inflammatory responses and protect against systemic inflammation.
Objective:
To identify endothelial cell–released soluble factors that protect against endothelial barrier dysfunction and systemic inflammation.
Methods and Results:
We found that conditioned medium of endothelial cells inhibited cyclooxgenase-2 and interleukin-6 expression in macrophages stimulated with lipopolysaccharide. Analysis of conditioned medium extracts by liquid chromatography–mass spectrometry showed the presence of 5-methoxytryptophan (5-MTP), but not other related tryptophan metabolites. Furthermore, endothelial cell–derived 5-MTP suppressed lipopolysaccharide-induced inflammatory responses and signaling in macrophages and endotoxemic lung tissues. Lipopolysaccharide suppressed 5-MTP level in endothelial cell-conditioned medium and reduced serum 5-MTP level in the murine sepsis model. Intraperitoneal injection of 5-MTP restored serum 5-MTP accompanied by the inhibition of lipopolysaccharide-induced endothelial leakage and suppression of lipopolysaccharide- or cecal ligation and puncture–mediated proinflammatory mediators overexpression. 5-MTP administration rescued lungs from lipopolysaccharide-induced damages and prevented sepsis-related mortality. Importantly, compared with healthy subjects, serum 5-MTP level in septic patients was decreased by 65%, indicating an important clinical relevance.
Conclusions:
We conclude that 5-MTP belongs to a novel class of endothelium-derived protective molecules that defend against endothelial barrier dysfunction and excessive systemic inflammatory responses.
Collapse
Affiliation(s)
- Yi-Fu Wang
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| | - Yu-Juei Hsu
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| | - Hsu-Feng Wu
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| | - Guan-Lin Lee
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| | - Ya-Sung Yang
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| | - Jing-Yiing Wu
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| | - Shaw-Fang Yet
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| | - Kenneth K. Wu
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| | - Cheng-Chin Kuo
- From the Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan (Y.-F.W., H.-F.W., G.-L.L., J.-Y.W., S.-F.Y., K.K.W., C.-C.K.); Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan (Y.-F.W.); Division of Nephrology (Y.-J.H.), Division of Infectious Diseases and Tropical Medicine (Y.-S.Y.), Department of Medicine, Tri-Service General Hospital (Y.-J.H., Y.-S.Y.), and Graduate Institutes of Life Sciences and Biochemistry (Y.-J.H., G.-L
| |
Collapse
|
38
|
Butcher MJ, Waseem TC, Galkina EV. Smooth Muscle Cell-Derived Interleukin-17C Plays an Atherogenic Role via the Recruitment of Proinflammatory Interleukin-17A+ T Cells to the Aorta. Arterioscler Thromb Vasc Biol 2016; 36:1496-506. [PMID: 27365405 DOI: 10.1161/atvbaha.116.307892] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 06/10/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Atherosclerosis is characterized by frequent communication between infiltrating leukocytes and vascular cells, through chemokine and cytokine networks. Interleukin-17C (IL-17C) is detectable within atherosclerotic lesions; however, the potential involvement of this cytokine has not been examined. Thus, we sought to investigate the role of IL-17C in atherosclerosis. APPROACH AND RESULTS The expression of IL-17 cytokines was profiled within aortas of apolipoprotein E double knockout (Apoe(-/-)) mice, and Il17c expression was elevated. Flow cytometry experiments revealed a major population of aortic IL-17C-producing smooth muscle cells. Next, we generated Il17c(-/-)Apoe(-/-) mice and demonstrated that atherosclerotic lesion and collagen content was diminished within Western diet-fed Il17c(-/-)Apoe(-/-) aortas and aortic roots in comparison to Apoe(-/-) controls. Smooth muscle cells and fibroblasts were mainly responsible for the reduced Col1A1 expression in the aorta of Il17c(-/-)Apoe(-/-) mice. Importantly, IL-17C-treated Apoe(-/-) aortas showed upregulated Col1A1 expression ex vivo. Il17c(-/-)Apoe(-/-) mice displayed a proportional reduction in aortic macrophages, neutrophils, T cells, T helper 1 cells, and T regulatory cells, without corresponding changes in the peripheral immune composition. Examination of aortic IL-17A(+) T-cell receptor γδ T cells and Th17 cells demonstrated a stark reduction in the percentage and number of these subsets within Il17c(-/-)Apoe(-/-) versus Apoe(-/-) mice. Explanted 12-week Western diet-fed Apoe(-/-) aortas treated with IL-17C resulted in the induction of multiple vascular chemokines and cytokines. Th17 cells demonstrated attenuated migration toward supernatants from cultures of Il17c(-/-)Apoe(-/-) smooth muscle cells, and short-term homing experiments revealed diminished recruitment of Th17 cells to the aorta of Il17c(-/-)Apoe(-/-) recipients. CONCLUSIONS Smooth muscle cell-derived IL-17C plays a proatherogenic role by supporting the recruitment of Th17 cells to atherosclerotic lesions.
Collapse
Affiliation(s)
- Matthew J Butcher
- From the Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk
| | - Tayab C Waseem
- From the Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk
| | - Elena V Galkina
- From the Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk.
| |
Collapse
|
39
|
Wang YF, Lee GL, Huang YH, Kuo CC. sn-1,2-diacylglycerols protect against lethal endotoxemia by controlling systemic inflammation. Immunobiology 2016; 221:1309-18. [PMID: 27357266 DOI: 10.1016/j.imbio.2016.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/06/2016] [Accepted: 05/23/2016] [Indexed: 01/01/2023]
Abstract
Systemic inflammation has emerged as a key pathophysiological process that induces multiple organ injuries and causes serious human diseases. Despite substantial evidence supporting the role of diacylglycerols (DAG) in modulating chronic inflammation and chronic diseases, the potential mechanisms of its involvement in TLRs-mediated inflammation are still unclear. Here, we show that sn-1,2-diacylglycerols modulate LPS/TLR4-mediated inflammation in vitro and in vivo. ELISA and western blotting experiments indicated that sn-1,2-diacylglycerols suppress LPS-induced responses, including IL-6 and TNF-α production, and COX-2 expression in mouse RAW264.7 macrophages and human endothelial cells, in a dose-dependent manner. Using LPS-induced murine model of systemic inflammation, we show that sn-1,2-diacylglycerols block the cytokine storm, the expression of inflammatory mediators, and LPS-induced septic lung damage and mortality. sn-1,2-diacylglycerols reduce systemic inflammation by inhibiting LPS-induced p38 MAPK- and PI3K/AKT-mediated NF-κB activation in macrophages. These results suggest that exogenous DAG probably acts by blocking p38 MAPK or PI3K/AKT signal transduction, thereby down-regulating NF-κB activation and NF-κB-mediated transcription of genes encoding cytokines and pro-inflammatory oxidative enzymes. Our findings demonstrate that exogenous sn-1,2-diacylglycerol protects mice from LPS-induced lethal endotoxemia by suppressing TLR4-driven inflammatory responses, suggesting that 1,2-diacylglycerols may be used as dietary health supplements for the prevention or therapy of systemic inflammatory diseases.
Collapse
Affiliation(s)
- Yi-Fu Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan; Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Guan-Lin Lee
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan; Graduate Institutes of Life Sciences and Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Hua Huang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan; Graduate Institutes of Life Sciences and Biochemistry, National Defense Medical Center, Taipei, Taiwan; Metabolomic Research Center, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.
| |
Collapse
|
40
|
Lee GL, Wu JY, Yeh CC, Kuo CC. TLR4 induces CREB-mediated IL-6 production via upregulation of F-spondin to promote vascular smooth muscle cell migration. Biochem Biophys Res Commun 2016; 473:1205-1210. [PMID: 27091427 DOI: 10.1016/j.bbrc.2016.04.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 04/09/2016] [Indexed: 10/21/2022]
Abstract
Toll-like receptor 4 (TLR4) is important in promoting inflammation and vascular smooth muscle cell (VSMC) migration, both of which contribute to atherosclerosis development and progression. But the mechanism underlying the regulation of TLR4 in VSMC migration remains unclear. Stimulation of VSMCs with LPS increased the cellular level of F-spondin which is associated with the regulation of proinflammatory cytokine production. The LPS-induced F-spondin expression depended on TLR4-mediated PI3K/Akt pathway. Suppression of F-spondin level by siRNA inhibited not only F-spondin expression but also LPS-induced phosphorylation of cAMP response element binding protein (CREB) and IL-6 expression, VSMC migration and proliferation as well as MMP9 expression. Moreover, suppression of CREB level by siRNA inhibited TLR4-induced IL-6 production and VSMC migration. Inhibition of F-spondin siRNA on LPS-induced migration was restored by addition of exogenous recombinant mouse IL-6. We conclude that upon ligand binding, TLR4 activates PI3K/Akt signaling to induce F-spondin expression, subsequently control CREB-mediated IL-6 production to promote VSMC migration. These findings provide vital insights into the essential role of F-spondin in VSMC function and will be valuable for developing new therapeutic strategies against atherosclerosis.
Collapse
Affiliation(s)
- Guan-Lin Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan; Graduate Institutes of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Jing-Yiing Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Chang-Ching Yeh
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan; Graduate Institutes of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan; Graduate Institutes of Life Sciences, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.
| |
Collapse
|
41
|
The Comparison of Adipose Stem Cell and Placental Stem Cell in Secretion Characteristics and in Facial Antiaging. Stem Cells Int 2016; 2016:7315830. [PMID: 27057176 PMCID: PMC4761676 DOI: 10.1155/2016/7315830] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 12/15/2015] [Accepted: 01/10/2016] [Indexed: 12/29/2022] Open
Abstract
Background. Mesenchymal stem cells are the most commonly used seed cells in biomedical research and tissue engineering. Their secretory proteins have also been proven to play an important role in tissue healing. Methods. We isolated adipose stem cells and placental stem cells and performed analysis examining characteristics. The secretory proteins were extracted from conditioned medium and analyzed by MALDI-TOF/TOF. The antiaging effect of conditioned mediums was evaluated by the results of facial skin application. Results. Adipose stem cells and placental stem cells were found to be very similar in their surface markers and multipotency. The specific proteins secreted from adipose stem cells were more adept at cell adhesion, migration, wound healing, and tissue remodeling, while the proteins secreted by placental stem cells were more adept at angiogenesis, cell proliferation, differentiation, cell survival, immunomodulation, and collagen degradation. While these two types of conditioned medium could improve the facial index, the improvement of Melanin index after injection of the adipose stem cell conditioned medium was much more significant. Conclusion. The results suggest that the secreted proteins are ideal cell-free substances for regeneration medicine, especially in the antiaging field.
Collapse
|
42
|
Shiny A, Regin B, Mohan V, Balasubramanyam M. Coordinated augmentation of NFAT and NOD signaling mediates proliferative VSMC phenotype switch under hyperinsulinemia. Atherosclerosis 2016; 246:257-66. [PMID: 26814423 DOI: 10.1016/j.atherosclerosis.2016.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/20/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
Abstract
AIM Although hyperglycemia has been demonstrated to play a significant role in the vascular disease associated with type 2 diabetes, the mechanisms underlying hyperinsulinemia mediated vascular dysfunction are not well understood. We have analyzed whether hyperinsulinemia could activate NFAT (Nuclear factor of activated T cells) signaling and thereby influence vascular smooth muscle cell (VSMC) migration and proliferation, a major event in the progression of atherosclerosis. METHODS AND RESULTS Human aortic VSMCs upon chronic insulin treatment exhibited increased expression of NFATc1 both at the mRNA and protein levels. The mechanistic role of NFAT in VSMC migration and proliferation was examined using 11R-VIVIT, a cell permeable NFAT specific inhibitor, where it reduced the insulin effect on VSMC, which was further substantiated by over expression or silencing of NFATc1gene (p < 0.05). This study also report for the first time the role of NFAT in NOD (Nucleotide oligomerization domain) mediated innate immune signaling and its significance in insulin effect on VSMCs. mRNA expression of NOD was up regulated when cells were treated with insulin or ligands whereas pretreatment with 11R-VIVIT reversed this effect (p < 0.05). Our study uphold the clinical significance as we observed an increased mRNA expression of NFATc1 in monocytes isolated from patients with type 2 diabetes which correlated positively with insulin resistance and glycemic load (p < 0.05). DISCUSSION This study suggests that targeted NFAT inhibition can be an effective strategy to coordinately quench insulin induced proliferative and inflammatory responses along with innate immunity alterations in vascular smooth muscle cells, which underlie atherosclerosis.
Collapse
Affiliation(s)
- Abhijit Shiny
- Department of Cell and Molecular Biology, Madras Diabetes Research Foundation, Dr. Mohan's Diabetes Specialities Centre Gopalapuram, Chennai, India.
| | - Bhaskaran Regin
- Department of Cell and Molecular Biology, Madras Diabetes Research Foundation, Dr. Mohan's Diabetes Specialities Centre Gopalapuram, Chennai, India
| | - Viswanathan Mohan
- Department of Cell and Molecular Biology, Madras Diabetes Research Foundation, Dr. Mohan's Diabetes Specialities Centre Gopalapuram, Chennai, India
| | - Muthuswamy Balasubramanyam
- Department of Cell and Molecular Biology, Madras Diabetes Research Foundation, Dr. Mohan's Diabetes Specialities Centre Gopalapuram, Chennai, India
| |
Collapse
|
43
|
Wu JY, Kuo CC. ADP-Ribosylation Factor 3 Mediates Cytidine-Phosphate-Guanosine Oligodeoxynucleotide-Induced Responses by Regulating Toll-Like Receptor 9 Trafficking. J Innate Immun 2015; 7:623-36. [PMID: 26067373 DOI: 10.1159/000430785] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 04/17/2015] [Indexed: 11/19/2022] Open
Abstract
Toll-like receptor 9 (TLR9) trafficking from the endoplasmic reticulum (ER) into endolysosomes is critical for eliciting cytidine-phosphate-guanosine (CpG) DNA-mediated immune responses. ADP-ribosylation factor 3 (ARF3) is a member of the Ras superfamily, which is crucial for a wide variety of cellular events including protein trafficking. In this study, we found that the inhibition of ARF3 by dominant mutants and siRNA impaired CpG oligodeoxynucleotide (ODN)-mediated responses whereas cells expressing the constitutively active ARF3 mutant enhanced CpG ODN-induced NF-x03BA;B activation and cytokine production. Further experiments with MyD88-overexpressing fibroblast cells transfected with a dominant-negative mutant and a constitutively active mutant of ARF3 demonstrated that ARF3 regulated CpG ODN-mediated signaling upstream of MyD88. Additional studies have shown that ARF3 inhibition impairs TLR9 trafficking from the ER into endolysosomes, thereby inhibiting the functional cleavage of TLR9, although it has no significant effect on CpG ODN uptake. Furthermore, activated ARF3 is associated with Unc93B1 and TLR9, suggesting that ARF3 conducts TLR9 trafficking by forming the TLR9-Unc93B1-ARF3 complex. Overall, our findings demonstrate that a novel ARF3 axis pathway mediates CpG ODN-induced responses by regulating TLR9 trafficking.
Collapse
Affiliation(s)
- Jing-Yiing Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, ROC
| | | |
Collapse
|
44
|
Evans BC, Hocking KM, Osgood MJ, Voskresensky I, Dmowska J, Kilchrist KV, Brophy CM, Duvall CL. MK2 inhibitory peptide delivered in nanopolyplexes prevents vascular graft intimal hyperplasia. Sci Transl Med 2015; 7:291ra95. [PMID: 26062847 PMCID: PMC5371354 DOI: 10.1126/scitranslmed.aaa4549] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Autologous vein grafts are commonly used for coronary and peripheral artery bypass but have a high incidence of intimal hyperplasia (IH) and failure. We present a nanopolyplex (NP) approach that efficiently delivers a mitogen-activated protein kinase (MAPK)-activated protein (MAPKAP) kinase 2 inhibitory peptide (MK2i) to graft tissue to improve long-term patency by inhibiting pathways that initiate IH. In vitro testing in human vascular smooth muscle cells revealed that formulation into MK2i-NPs increased cell internalization, endosomal escape, and intracellular half-life of MK2i. This efficient delivery mechanism enabled MK2i-NPs to sustain potent inhibition of inflammatory cytokine production and migration in vascular cells. In intact human saphenous vein, MK2i-NPs blocked inflammatory and migratory signaling, as confirmed by reduced phosphorylation of the posttranscriptional gene regulator heterogeneous nuclear ribonucleoprotein A0, the transcription factor cAMP (adenosine 3',5'-monophosphate) element-binding protein, and the chaperone heat shock protein 27. The molecular effects of MK2i-NPs caused functional inhibition of IH in human saphenous vein cultured ex vivo. In a rabbit vein transplant model, a 30-min intraoperative graft treatment with MK2i-NPs significantly reduced in vivo IH 28 days posttransplant compared with untreated or free MK2i-treated grafts. The decrease in IH in MK2i-NP-treated grafts in the rabbit model also corresponded with decreased cellular proliferation and maintenance of the vascular wall smooth muscle cells in a more contractile phenotype. These data indicate that nanoformulated MK2 inhibitors are a promising strategy for preventing graft failure.
Collapse
Affiliation(s)
- Brian C Evans
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kyle M Hocking
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Michael J Osgood
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Igor Voskresensky
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Julia Dmowska
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kameron V Kilchrist
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Colleen M Brophy
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA. Veterans Affairs Medical Center, VA Tennessee Valley Healthcare System, Nashville TN 37212, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
45
|
Abstract
SIGNIFICANCE Pattern-recognition receptors (PRRs) are a family of receptors that are used to detect pathogen-associated molecular patterns or damage-associated molecular patterns, which initiate immune responses to resolve infections and repair damaged tissues. Abnormalities in PRR activation will unavoidably lead to excessive inflammation. RECENT ADVANCES Although multiple pathophysiological processes are involved in cardiovascular disease, recent studies have highlighted the importance of innate PRRs, in particular, Toll-like receptors and nucleotide-binding oligomerization domain-like receptors, in mediating inflammatory responses and cardiovascular function. CRITICAL ISSUES The functional roles and regulatory mechanisms of PRRs in cardiovascular diseases are still largely unknown. In particular, controversies exist on the certainty of these detrimental or beneficial effects of some PRRs in different diseased states or different experimental animal models. FUTURE DIRECTIONS Considering that the molecular mechanisms for individual PRR to regulate cellular function are complex and multiple PRRs are activated simultaneously or synergistically, a better understanding of the function of individual PRRs and the interplay of PRRs will provide unexpected opportunities to develop new therapies for cardiovascular disease by modulation of an innate immune system.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Pharmacology, Shandong University School of Medicine , Jinan, China
| | | |
Collapse
|
46
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf) 2015; 214:33-50. [PMID: 25677529 DOI: 10.1111/apha.12466] [Citation(s) in RCA: 292] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/02/2015] [Accepted: 02/09/2015] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) exhibit phenotypic and functional plasticity in order to respond to vascular injury. In case of the vessel damage, VSMCs are able to switch from the quiescent 'contractile' phenotype to the 'proinflammatory' phenotype. This change is accompanied by decrease in expression of smooth muscle (SM)-specific markers responsible for SM contraction and production of proinflammatory mediators that modulate induction of proliferation and chemotaxis. Indeed, activated VSMCs could efficiently proliferate and migrate contributing to the vascular wall repair. However, in chronic inflammation that occurs in atherosclerosis, arterial VSMCs become aberrantly regulated and this leads to increased VSMC dedifferentiation and extracellular matrix formation in plaque areas. Proatherosclerotic switch in VSMC phenotype is a complex and multistep mechanism that may be induced by a variety of proinflammatory stimuli and hemodynamic alterations. Disturbances in hemodynamic forces could initiate the proinflammatory switch in VSMC phenotype even in pre-clinical stages of atherosclerosis. Proinflammatory signals play a crucial role in further dedifferentiation of VSMCs in affected vessels and propagation of pathological vascular remodelling.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Research Center for Children's Health; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
| | - A. N. Orekhov
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Department of Biophysics; Biological Faculty; Moscow State University; Moscow Russia
| | - Y. V. Bobryshev
- Institute for Atherosclerosis; Skolkovo Innovative Center; Moscow Russia
- Faculty of Medicine; School of Medical Sciences; University of New South Wales; Kensington Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
47
|
Quillard T, Araújo HA, Franck G, Shvartz E, Sukhova G, Libby P. TLR2 and neutrophils potentiate endothelial stress, apoptosis and detachment: implications for superficial erosion. Eur Heart J 2015; 36:1394-404. [PMID: 25755115 DOI: 10.1093/eurheartj/ehv044] [Citation(s) in RCA: 258] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/01/2015] [Indexed: 12/12/2022] Open
Abstract
AIMS Superficial erosion of atheromata causes many acute coronary syndromes, but arises from unknown mechanisms. This study tested the hypothesis that Toll-like receptor-2 (TLR2) activation contributes to endothelial apoptosis and denudation and thus contributes to the pathogenesis of superficial erosion. METHODS AND RESULTS Toll-like receptor-2 and neutrophils localized at sites of superficially eroded human plaques. In vitro, TLR2 ligands (including hyaluronan, a matrix macromolecule abundant in eroded lesions) induced endothelial stress, characterized by reactive oxygen species production, endoplasmic reticulum (ER) stress, and apoptosis. Co-incubation of neutrophils with endothelial cells (ECs) potentiated these effects and induced EC apoptosis and detachment. We then categorized human atherosclerotic plaques (n = 56) based on morphologic features associated with superficial erosion, 'stable' fibrotic, or 'vulnerable' lesions. Morphometric analyses of the human atheromata localized neutrophils and neutrophil extracellular traps (NETs) near clusters of apoptotic ECs in smooth muscle cell (SMC)-rich plaques. The number of luminal apoptotic ECs correlated with neutrophil accumulation, amount of NETs, and TLR2 staining in SMC-rich plaques, but not in 'vulnerable' atheromata. CONCLUSION These in vitro observations and analyses of human plaques indicate that TLR2 stimulation followed by neutrophil participation may render smooth muscle cell-rich plaques susceptible to superficial erosion and thrombotic complications by inducing ER stress, apoptosis, and favouring detachment of EC.
Collapse
Affiliation(s)
- Thibaut Quillard
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA INSERM, UMR957, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, Nantes 44035, France
| | - Haniel Alves Araújo
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gregory Franck
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eugenia Shvartz
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Galina Sukhova
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Toll-like receptor 2 mediates vascular contraction and activates RhoA signaling in vascular smooth muscle cells from STZ-induced type 1 diabetic rats. Pflugers Arch 2015; 467:2361-74. [PMID: 25600901 DOI: 10.1007/s00424-015-1688-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 01/11/2023]
Abstract
Increased vascular smooth muscle cell (VSMC) contraction is an early and critical contributor to the pathogenesis of vascular dysfunction in diabetes; however, knowledge regarding the underlying mechanisms is scarce. Toll-like receptor 2 (TLR2), a well-known component of the innate immunity, is expressed in VSMC and recently has been identified to be systemically activated in diabetes. Whether TLR2 is locally activated in the diabetic blood vessels and have effect on contraction is not known. In the current study, we examined the role of TLR2 in increased vascular contraction in diabetes. Utilizing rat model of type 1 diabetes (induced by streptozotocin (STZ)), we demonstrated that aortas from STZ-diabetic rats exhibit increased expression of TLR2 and its adaptor protein, myeloid differentiation primary response 88 (MyD88), as well as enhanced protein-protein interaction between TLR2 and MyD88, suggesting a TLR2 signaling activation. Blockade of TLR2 in intact aortas using anti-TLR2 antibody attenuated increased vascular contraction in STZ-diabetic rat as assessed by wire myograph. Activation of TLR2 by specific ligand in primary aortic VSMC cultures triggered activation of RhoA which was exacerbated in cells from STZ-diabetic rats than control rats. Activation of RhoA was accompanied by phosphorylation and therefore activation of its downstream targets myosin phosphatase target subunit I and myosin light chain (markers of VSMC contraction). Taken together, these results provide evidence for the role of TLR2 in increased contraction in diabetic blood vessels that involves RhoA signaling. Thus, targeting vascular TLR2 offers a promising drug target to treat vascular dysfunction in diabetes.
Collapse
|
49
|
Affiliation(s)
- Valerie Z Wall
- From the Departments of Pathology (V.Z.W., K.E.B.) and Medicine, Division of Metabolism, Endocrinology and Nutrition (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle
| | - Karin E Bornfeldt
- From the Departments of Pathology (V.Z.W., K.E.B.) and Medicine, Division of Metabolism, Endocrinology and Nutrition (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle.
| |
Collapse
|
50
|
Lim S, Park S. Role of vascular smooth muscle cell in the inflammation of atherosclerosis. BMB Rep 2014; 47:1-7. [PMID: 24388105 PMCID: PMC4163848 DOI: 10.5483/bmbrep.2014.47.1.285] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Indexed: 01/13/2023] Open
Abstract
Atherosclerosis is a pathologic process occurring within the artery, in which many cell types, including T cell, macrophages, endothelial cells, and smooth muscle cells, interact, and cause chronic inflammation, in response to various inner- or outer-cellular stimuli. Atherosclerosis is characterized by a complex interaction of inflammation, lipid deposition, vascular smooth muscle cell proliferation, endothelial dysfunction, and extracellular matrix remodeling, which will result in the formation of an intimal plaque. Although the regulation and function of vascular smooth muscle cells are important in the progression of atherosclerosis, the roles of smooth muscle cells in regulating vascular inflammation are rarely focused upon, compared to those of endothelial cells or inflammatory cells. Therefore, in this review, we will discuss here how smooth muscle cells contribute or regulate the inflammatory reaction in the progression of atherosclerosis, especially in the context of the activation of various membrane receptors, and how they may regulate vascular inflammation.
Collapse
Affiliation(s)
| | - Sungha Park
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine; Cardiovascular Research Institute, Yonsei University College of Medicine; Division of Cardiology, Yonsei University College of Medicine, Seoul 120-752, Korea
| |
Collapse
|