1
|
Grootaert MOJ, Bennett MR. Vascular smooth muscle cells in atherosclerosis: time for a re-assessment. Cardiovasc Res 2021; 117:2326-2339. [PMID: 33576407 PMCID: PMC8479803 DOI: 10.1093/cvr/cvab046] [Citation(s) in RCA: 244] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are key participants in both early and late-stage atherosclerosis. VSMCs invade the early atherosclerotic lesion from the media, expanding lesions, but also forming a protective fibrous cap rich in extracellular matrix to cover the 'necrotic' core. Hence, VSMCs have been viewed as plaque-stabilizing, and decreased VSMC plaque content-often measured by expression of contractile markers-associated with increased plaque vulnerability. However, the emergence of lineage-tracing and transcriptomic studies has demonstrated that VSMCs comprise a much larger proportion of atherosclerotic plaques than originally thought, demonstrate multiple different phenotypes in vivo, and have roles that might be detrimental. VSMCs down-regulate contractile markers during atherosclerosis whilst adopting alternative phenotypes, including macrophage-like, foam cell-like, osteochondrogenic-like, myofibroblast-like, and mesenchymal stem cell-like. VSMC phenotypic switching can be studied in tissue culture, but also now in the media, fibrous cap and deep-core region, and markedly affects plaque formation and markers of stability. In this review, we describe the different VSMC plaque phenotypes and their presumed cellular and paracrine functions, the regulatory mechanisms that control VSMC plasticity, and their impact on atherogenesis and plaque stability.
Collapse
Affiliation(s)
- Mandy O J Grootaert
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, ACCI, Addenbrookes Hospital, CB2 0QQ Cambridge, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, ACCI, Addenbrookes Hospital, CB2 0QQ Cambridge, UK
| |
Collapse
|
2
|
Checkouri E, Blanchard V, Meilhac O. Macrophages in Atherosclerosis, First or Second Row Players? Biomedicines 2021; 9:biomedicines9091214. [PMID: 34572399 PMCID: PMC8465019 DOI: 10.3390/biomedicines9091214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages represent a cell type that has been widely described in the context of atherosclerosis since the earliest studies in the 17th century. Their role has long been considered to be preponderant in the onset and aggravation of atherosclerosis, in particular by participating in the establishment of a chronic inflammatory state by the release of pro-inflammatory cytokines and by uncontrolled engorgement of lipids resulting in the formation of foam cells and later of the necrotic core. However, recent evidence from mouse models using an elegant technique of tracing vascular smooth muscle cells (VSMCs) during plaque development revealed that resident VSMCs display impressive plastic properties in response to an arterial injury, allowing them to switch into different cell types within the plaque, including mesenchymal-like cells, macrophage-like cells and osteochondrogenic-like cells. In this review, we oppose the arguments in favor or against the influence of macrophages versus VSMCs in all stages of atherosclerosis including pre-atherosclerosis, formation of lipid-rich foam cells, development of the necrotic core and the fibrous cap as well as calcification and rupture of the plaque. We also analyze the relevance of animal models for the investigation of the pathophysiological mechanisms of atherosclerosis in humans, and discuss potential therapeutic strategies targeting either VSMCs or macrophage to prevent the development of cardiovascular events. Overall, although major findings have been made from animal models, efforts are still needed to better understand and therefore prevent the development of atherosclerotic plaques in humans.
Collapse
Affiliation(s)
- Eloïse Checkouri
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- Habemus Papam, Food Industry, 97470 Saint-Benoit, France
| | - Valentin Blanchard
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- Departments of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Olivier Meilhac
- INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Sainte-Clotilde, France; (E.C.); (V.B.)
- CHU de La Réunion, INSERM, CIC1410, 97500 Saint-Pierre, France
- Correspondence: ; Tel.: +33-262-93-8811
| |
Collapse
|
3
|
Lipid accumulation and novel insight into vascular smooth muscle cells in atherosclerosis. J Mol Med (Berl) 2021; 99:1511-1526. [PMID: 34345929 DOI: 10.1007/s00109-021-02109-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 06/03/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
Atherosclerosis is a chronic and progressive process. It is the most important pathological basis of cardiovascular disease and stroke. Vascular smooth muscle cells (VSMCs) are an essential cell type in atherosclerosis. Previous studies have revealed that VSMCs undergo phenotypic transformation in atherosclerosis to participate in the retention of atherogenic lipoproteins as well as the formation of the fibrous cap and the underlying necrotic core in plaques. The emergence of lineage-tracing studies indicates that the function and number of VSMCs in plaques have been greatly underestimated. In addition, recent studies have revealed that VSMCs make up at least 50% of the foam cell population in human and mouse atherosclerotic lesions. Therefore, understanding the formation of lipid-loaded VSMCs and their regulatory mechanisms is critical to elucidate the pathogenesis of atherosclerosis and to explore potential therapeutic targets. Moreover, combination of many complementary technologies such as lineage tracing, single-cell RNA sequencing (scRNA-seq), flow cytometry, and mass cytometry (CyTOF) with immunostaining has been performed to further understand the complex VSMC function. Correct identification of detrimental and beneficial processes may reveal successful therapeutic treatments targeting VSMCs and their derivatives during atherosclerosis. The purpose of this review is to summarize the process of lipid-loaded VSMC formation in atherosclerosis and to describe novel insight into VSMCs gained by using multiple advanced methods.
Collapse
|
4
|
Valin A, Del Rey MJ, Municio C, Usategui A, Romero M, Fernández-Felipe J, Cañete JD, Blanco FJ, Ruano Y, Criado G, Pablos JL. IL6/sIL6R regulates TNFα-inflammatory response in synovial fibroblasts through modulation of transcriptional and post-transcriptional mechanisms. BMC Mol Cell Biol 2020; 21:74. [PMID: 33126846 PMCID: PMC7596982 DOI: 10.1186/s12860-020-00317-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/14/2020] [Indexed: 01/01/2023] Open
Abstract
Introduction The clinical efficacy of specific interleukin-6 inhibitors has confirmed the central role of IL6 in rheumatoid arthritis (RA). However the local role of IL6, in particular in synovial fibroblasts (SF) as a direct cellular target to IL6/sIL6R signal is not well characterized. The purpose of the study was to characterize the crosstalk between TNFα and IL6/sIL6R signaling to the effector pro-inflammatory response of SF. Methods SF lines were stimulated with either TNFα, IL6/sIL6R, or both together, for the time and dose indicated for each experiment, and where indicated, cells were treated with inhibitors actinomycin D, adalimumab, ruxolitinib and cycloheximide. mRNA expression of cytokines, chemokines and matrix metalloproteases (MMPs) were analyzed by quantitative RT-PCR. Level of IL8/CXCL8 and CCL8 in culture supernatants was measured by ELISA. Mononuclear and polymorphonuclear cells migration assays were assessed by transwell using conditioned medium from SF cultures. Statistical analyses were performed as indicated in the corresponding figure legends and a p-value < 0.05 was considered statistically significant. Results The stimulation of SF with IL6/sIL6R and TNFα, cooperatively promotes the expression of mono- and lymphocytic chemokines such as IL6, CCL8 and CCL2, as well as matrix degrading enzymes such as MMP1, while inhibiting the induction of central neutrophil chemokines such as IL8/CXCL8. These changes in the pattern of chemokines expression resulted in reduced polymorphonuclear (PMN) and increased mononuclear cells (MNC) chemoattraction by SF. Mechanistic analyses of the temporal expression of genes demonstrated that the cooperative regulation mediated by these two factors is mostly induced through de novo transcriptional mechanisms activated by IL6/sIL6R. Furthermore, we also demonstrate that TNFα and IL6/sIL6R cooperation is partially mediated by the expression of secondary factors signaling through JAK/STAT pathways. Conclusions These results point out to a highly orchestrated response to IL6 in TNFα-induced SF and provide additional insights into the role of IL6/sIL6R in the context of RA, highlighting the contribution of IL6/sIL6R to the interplay of SF with other inflammatory cells. Supplementary information Supplementary information accompanies this paper at 10.1186/s12860-020-00317-7.
Collapse
Affiliation(s)
- Alvaro Valin
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain. .,Present Address: Springer Healthcare Iberica SL, Madrid, Spain.
| | - Manuel J Del Rey
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Cristina Municio
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Alicia Usategui
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Marina Romero
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Jesús Fernández-Felipe
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Juan D Cañete
- Unitat d'Artritis, Servei de Reumatologia, Hospital Clínic de Barcelona and Institut d'Investigacions Biomèdiques August Pí i Sunyer, Barcelona, Spain
| | - Francisco J Blanco
- Laboratorio de Investigación Osteoarticular y del Envejecimiento, Instituto de Investigación Biomédica de A Coruña, INIBIC, A Coruña, Spain
| | - Yolanda Ruano
- Servicio de Anatomía Patológica, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Gabriel Criado
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - José L Pablos
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain. .,Servicio de Reumatología, Hospital 12 de Octubre, Universidad Complutense de Madrid, 28041, Madrid, Spain.
| |
Collapse
|
5
|
Zhang L, She ZG, Li H, Zhang XJ. Non-alcoholic fatty liver disease: a metabolic burden promoting atherosclerosis. Clin Sci (Lond) 2020; 134:1775-1799. [PMID: 32677680 DOI: 10.1042/cs20200446] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/06/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the fastest growing chronic liver disease, with a prevalence of up to 25% worldwide. Individuals with NAFLD have a high risk of disease progression to cirrhosis, hepatocellular carcinoma (HCC), and liver failure. With the exception of intrahepatic burden, cardiovascular disease (CVD) and especially atherosclerosis (AS) are common complications of NAFLD. Furthermore, CVD is a major cause of death in NAFLD patients. Additionally, AS is a metabolic disorder highly associated with NAFLD, and individual NAFLD pathologies can greatly increase the risk of AS. It is increasingly clear that AS-associated endothelial cell damage, inflammatory cell activation, and smooth muscle cell proliferation are extensively impacted by NAFLD-induced systematic dyslipidemia, inflammation, oxidative stress, the production of hepatokines, and coagulations. In clinical trials, drug candidates for NAFLD management have displayed promising effects for the treatment of AS. In this review, we summarize the key molecular events and cellular factors contributing to the metabolic burden induced by NAFLD on AS, and discuss therapeutic strategies for the improvement of AS in individuals with NAFLD.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
- Basic Medical School, Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Luojia Mount Wuchang, Wuhan 430072, China
| |
Collapse
|
6
|
Shatoor AS, Al Humayed S, Alkhateeb MA, Shatoor KA, Aldera H, Alassiri M, Shati AA. Crataegus Aronia protects and reverses vascular inflammation in a high fat diet rat model by an antioxidant mechanism and modulating serum levels of oxidized low-density lipoprotein. PHARMACEUTICAL BIOLOGY 2019; 57:38-48. [PMID: 30702358 PMCID: PMC6366417 DOI: 10.1080/13880209.2018.1564930] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
CONTEXT Crataegus aronia (Willd.) Bosc (Rosaceae) (syn. Azarolus L) is traditionally used to treat cardiovascular disorders. OBJECTIVES To investigate C. aronia protection against a high-fat diet (HFD)-induced vascular inflammation in rats. MATERIALS AND METHODS Wistar Male rats (180-220 g) were divided (n = 10/group) as control fed a standard diet (STD), STD + C. aronia (200 mg/kg, orally), HFD, HFD + C. aronia and HFD post-treated with C. aronia. Simvastatin (20 mg/kg) was co- or post-administered as a positive control drug. HFD was given for 8 weeks, and all other treatments were administered for 4 weeks. RESULTS Most significantly, co-administration of C. aronia to HFD-fed rats reduced the thickness of aorta tunica media (90 ± 5 vs. 160 ± 11.3 µm) and adventitia (54.3 ± 3.8 vs. 93.6 ± 9.4 µm). It also lowered protein levels of TNF-α (0.51 ± 0.15 and 0.15 ± 0.16 vs. 0.1 ± 0.09%) and IL-6 (0.52 ± 0.19 vs. 1.0 ± 0.2%) in their aorta or serum (5.9 ± 0.91 vs. 12.98 ± 1.3 ng/mL and 78.1 ± 6.7 vs. 439 ± 78 pg/mL, respectively). It also lowered all serum lipids and increased aorta levels of GSH levels (70.4 ± 4.0 vs. 40.7 µM) and activity of SOD (5.7 ± 0.7 vs. 2.9 ± 0.6 U/mg) and decreased serum levels of ox-LDL-c (566.7 ± 46 vs. 1817 ± 147 ng/mL). Such effects were more profound than all other treatments. CONCLUSIONS C. aronia inhibits the HFD-induced vascular inflammation and its use in clinical trials is recommended.
Collapse
Affiliation(s)
- Abdullah S. Shatoor
- Department of Medicine, Cardiology Section, College of Medicine, King Khalid University (KKU), Abha, Saudi Arabia
- CONTACT Abdullah S. Shatoor Department of Medicine, Cardiology Section, College of Medicine, King Khalid University, Abha64121, Saudi Arabia
| | - Suliman Al Humayed
- Department of Medicine, Cardiology Section, College of Medicine, King Khalid University (KKU), Abha, Saudi Arabia
| | - Mahmoud A. Alkhateeb
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Khalid A. Shatoor
- An intern, College of Medicine, King Khalid University (KKU), Abha, Saudi Arabia
| | - Hussain Aldera
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- King Abdullah International Medical Research center (KAIMRC), Riyadh, Saudi Arabia
| | - Mohammed Alassiri
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- King Abdullah International Medical Research center (KAIMRC), Riyadh, Saudi Arabia
| | - Ali A. Shati
- Department of Biology College of Science, College of Medicine, King Khalid University (KKU), Abha, Saudi Arabia
| |
Collapse
|
7
|
Upregulated LOX-1 Receptor: Key Player of the Pathogenesis of Atherosclerosis. Curr Atheroscler Rep 2019; 21:38. [DOI: 10.1007/s11883-019-0801-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
8
|
Choi B, Shin MK, Kim EY, Park JE, Lee H, Kim SW, Song JK, Chang EJ. Elevated Neuropeptide Y in Endothelial Dysfunction Promotes Macrophage Infiltration and Smooth Muscle Foam Cell Formation. Front Immunol 2019; 10:1701. [PMID: 31379881 PMCID: PMC6657015 DOI: 10.3389/fimmu.2019.01701] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 07/08/2019] [Indexed: 12/19/2022] Open
Abstract
Endothelial dysfunction has been linked to vascular inflammation and foam cell formation but the underlying mechanisms still remain unclear. We sought to define the factors inducing inflammation and smooth muscle foam cell formation under endothelial dysfunction using endothelial nitric oxide synthase (eNOS)-deficient mice. Vascular smooth muscle cells (VSMCs) from eNOS-deficient mice displayed increased expression of macrophage-related genes and elevated lipid uptake. Neuropeptide Y (NPY) was upregulated in the aorta from the eNOS-deficient mice and promoted macrophage chemotaxis toward VSMCs while enhancing the activity of matrix metalloproteinase-3. Notably, NPY induced lipid uptake in VSMCs, facilitating smooth muscle foam cell formation, in association with enhanced expression of genes related to modified low-density lipoprotein uptake and macrophages. NPY was augmented by inflammatory pentraxin 3 (PTX3) in VSMCs. PTX3 enhanced macrophage migratory capacity through the NPY/neuropeptide Y receptor axis and this effect was attenuated by pharmacological inhibition with a receptor-specific antagonist. These observations suggest that endothelial dysfunction leads to the elevation of NPY that amplifies vascular inflammation by increasing inflammatory cell chemotaxis and triggers smooth muscle foam cell formation.
Collapse
Affiliation(s)
- Bongkun Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Min-Kyung Shin
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Eun-Young Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji-Eun Park
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Halim Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seong Who Kim
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae-Kwan Song
- Division of Cardiology, Asan Medical Center, Research Institute for Valvular Heart Disease University of Ulsan College of Medicine, Seoul, South Korea
| | - Eun-Ju Chang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Smooth muscle cells (SMCs) are the major cell type in human atherosclerosis-prone arteries and take up excess lipids, thereby contributing to luminal occlusion. Here we provide a focused review on pathways by which smooth muscle cells (SMCs) can become foam cells in atherosclerosis. RECENT FINDINGS A synthesis of recent and older investigations provides key mechanistic insights into SMC foam cell formation. LDL and other apoB-containing lipoproteins are modified by a diverse array of oxidative, enzymatic, and nonenzymatic processes present in the arterial intima. These modifications of LDL all promote the aggregation of LDL (agLDL), a key finding from analysis of arterial lesion particles. Scavenger receptor and phagocytic capacity of SMCs can vary greatly, perhaps related to differences in SMC phenotype or in-vitro cell culture environments, and can be increased with exposure to cytokines, growth factors, and cholesterol. Macrophages promote the formation of SMC foam cells in direct or indirect co-culture models. SUMMARY SMCs contribute significantly to the foam cell population in atherosclerosis. Further investigation and identification of key mechanisms of SMC foam cell formation will help drive new therapeutics to reduce cardiovascular disease.
Collapse
MESH Headings
- Animals
- Apolipoproteins B/genetics
- Apolipoproteins B/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cell Differentiation
- Coculture Techniques
- Cytokines/pharmacology
- Foam Cells/drug effects
- Foam Cells/metabolism
- Foam Cells/pathology
- Gene Expression
- Humans
- Intercellular Signaling Peptides and Proteins/pharmacology
- Lipoproteins, LDL/genetics
- Lipoproteins, LDL/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phagocytosis
- Protein Aggregates/drug effects
- Receptors, Scavenger/genetics
- Receptors, Scavenger/metabolism
- Tunica Intima/drug effects
- Tunica Intima/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Collin S Pryma
- Department of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute at St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
10
|
Enzyme-modified non-oxidized LDL (ELDL) induces human coronary artery smooth muscle cell transformation to a migratory and osteoblast-like phenotype. Sci Rep 2018; 8:11954. [PMID: 30097618 PMCID: PMC6086911 DOI: 10.1038/s41598-018-30073-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/11/2018] [Indexed: 12/29/2022] Open
Abstract
Enzyme modified non-oxidative LDL (ELDL) is effectively taken up by vascular smooth muscle cells (SMC) and mediates transition into foam cells and produces phenotypic changes in SMC function. Our data show that incubation of human coronary artery SMC (HCASMC) with low concentration of ELDL (10 μg/ml) results in significantly enhanced foam cell formation compared to oxidized LDL (200 μg/ml; p < 0.01) or native LDL (200 μg/ml; p < 0.01). Bioinformatic network analysis identified activation of p38 MAPK, NFkB, ERK as top canonical pathways relevant for biological processes linked to cell migration and osteoblastic differentiation in ELDL-treated cells. Functional studies confirmed increased migration of HCASMC upon stimulation with ELDL (10 μg/ml) or Angiopoietin like protein 4, (ANGPTL4, 5 μg/ml), and gain in osteoblastic gene profile with significant increase in mRNA levels for DMP-1, ALPL, RUNX2, OPN/SPP1, osterix/SP7, BMP and reduction in mRNA for MGP and ENPP1. Enhanced calcification of HCASMC by ELDL was demonstrated by Alizarin Red staining. In summary, ELDL is highly potent in inducing foam cells in HCASMC and mediates a phenotypic switch with enhanced migration and osteoblastic gene profile. These results point to the potential of ELDL to induce migratory and osteoblastic effects in human smooth muscle cells with potential implications for migration and calcification of SMCs in human atherosclerosis.
Collapse
|
11
|
Goikuria H, Vandenbroeck K, Alloza I. Inflammation in human carotid atheroma plaques. Cytokine Growth Factor Rev 2018; 39:62-70. [PMID: 29396056 DOI: 10.1016/j.cytogfr.2018.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 12/31/2022]
Abstract
Inflammation in carotid atherosclerotic plaque is linked to plaque rupture and cerebrovascular accidents. The balance between pro- and anti-inflammatory mediators governs development of the plaque, and may mediate enhancement of lesion broadening or, on the contrary, delay progression. In addition to macrophages and endothelial cells, smooth muscle cells (SMCs), which are the dominant cell subset in advanced plaques, are crucial players in carotid atherosclerosis development given their ability to differentiate into distinct phenotypes in reponse to specific signals received from the environment of the lesion. Carotid atheroma SMCs actively contribute to the inflammation in the lesion because of their acquired capacity to produce inflammatory mediators. We review the successive stages of carotid atheroma plaque formation via fatty streak early-stage toward more advanced rupture-prone lesions and document involvement of cytokines and chemokines and their cellular sources and targets in plaque progression and rupture.
Collapse
Affiliation(s)
- Haize Goikuria
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain
| | - Koen Vandenbroeck
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Iraide Alloza
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain.
| |
Collapse
|
12
|
Hosaka K, Rojas K, Fazal HZ, Schneider MB, Shores J, Federico V, McCord M, Lin L, Hoh B. Monocyte Chemotactic Protein-1-Interleukin-6-Osteopontin Pathway of Intra-Aneurysmal Tissue Healing. Stroke 2017; 48:1052-1060. [PMID: 28292871 DOI: 10.1161/strokeaha.116.015590] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND PURPOSE We have previously demonstrated that the local delivery of monocyte chemotactic protein-1 (MCP-1) via an MCP-1-releasing poly(lactic-co-glycolic acid)-coated coil promotes intra-aneurysmal tissue healing. In this study, we demonstrate that interleukin-6 (IL-6) and osteopontin are downstream mediators in the MCP-1-mediated aneurysm-healing pathway. METHODS Murine carotid aneurysms were created in C57BL/6 mice. Drug-releasing coils (MCP-1, IL-6, and osteopontin) and control poly(lactic-co-glycolic acid) coils were created and then implanted into the aneurysms to evaluate their intra-aneurismal-healing capacity. To investigate the downstream mediators for aneurysm healing, blocking antibodies for IL-6 receptor and osteopontin were given to the mice implanted with the MCP-1-releasing coils. A histological analysis of both murine and human aneurysms was utilized to cross-validate the data. RESULTS We observed increased expression of IL-6 in MCP-1-coil-treated aneurysms and not in control-poly(lactic-co-glycolic acid)-only-treated aneurysms. MCP-1-mediated intra-aneurysmal healing is inhibited in mice given blocking antibody to IL-6 receptor. MCP-1-mediated intra-aneurysmal healing is also inhibited by blocking antibody to osteopontin. The role of IL-6 in intra-aneurysmal healing is in recruiting of endothelial cells and fibroblasts. Local delivery of osteopontin to murine carotid aneurysms via osteopontin-releasing coil significantly promotes intra-aneurysmal healing, but IL-6-releasing coil does not, suggesting that IL-6 cannot promote aneurysm healing independent of MCP-1. In the MCP-1-mediated aneurysm healing, osteopontin expression is dependent on IL-6; inhibition of IL-6 receptor significantly inhibits osteopontin expression in MCP-1-mediated aneurysm healing. CONCLUSIONS Our findings suggest that IL-6 and osteopontin are key downstream mediators of MCP-1-mediated intra-aneurysmal healing.
Collapse
Affiliation(s)
- Koji Hosaka
- From the Department of Neurosurgery, University of Florida, Gainesville.
| | - Kelley Rojas
- From the Department of Neurosurgery, University of Florida, Gainesville
| | - Hanain Z Fazal
- From the Department of Neurosurgery, University of Florida, Gainesville
| | | | - Jorma Shores
- From the Department of Neurosurgery, University of Florida, Gainesville
| | - Vincent Federico
- From the Department of Neurosurgery, University of Florida, Gainesville
| | - Matthew McCord
- From the Department of Neurosurgery, University of Florida, Gainesville
| | - Li Lin
- From the Department of Neurosurgery, University of Florida, Gainesville
| | - Brian Hoh
- From the Department of Neurosurgery, University of Florida, Gainesville
| |
Collapse
|
13
|
Čolak E, Ignjatović S, Radosavljević A, Žorić L. The association of enzymatic and non-enzymatic antioxidant defense parameters with inflammatory markers in patients with exudative form of age-related macular degeneration. J Clin Biochem Nutr 2017; 60:100-107. [PMID: 28366988 PMCID: PMC5371514 DOI: 10.3164/jcbn.16-30] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/26/2016] [Indexed: 02/05/2023] Open
Abstract
There are evidence that oxidative stress and inflammation are involved in the pathogenesis of the age-related macular degeneration (AMD). The aim of this study was to analyze the antioxidant defense parameters and inflammatory markers in patients with exudative form of AMD (eAMD), their mutual correlations and association with the specific forms of AMD. The cross-sectional study, included 75 patients with the eAMD, 31 patients with the early form, and 87 aged-matched control subjects. Significantly lower SOD, TAS and albumin values and higher GR, CRP and IL-6 were found in the eAMD compared to the early form (p<0.05). Significant negative correlations were found between GPx and fibrinogen (r = -0.254), TAS and IL-6 (r = -0.999) and positive correlations between uric acid and CRP (r = 0.292), IL-6 and uric acid (r = 0.398) in the eAMD. A significant association of CRP (OR: 1.16, 95% CI: 1.03-1.32, p = 0.018), fibrinogen (OR: 2.21, 95% CI: 1.14-4.85, p = 0.021), TAS (OR: 7.45, 95% CI: 3.97-14.35, p = 0.0001), albumin (OR: 1.25, 95% CI: 1.11-1.41, p = 0.0001) and uric acid (OR: 1.006, 95% CI: 1.00-1.02, p = 0.003) was found with the eAMD. In conclusion it may be suggested, there is a significant impairment of antioxidant and inflammatory parameter levels in eAMD patients. In addition, significant association exists between the tested inflammatory markers and antioxidant parameters with late-eAMD.
Collapse
Affiliation(s)
- Emina Čolak
- Institute of Medical Biochemistry, Clinical Center of Serbia, School of Pharmacy, University of Belgrade, Belgrade 11000, Serbia
| | - Svetlana Ignjatović
- Institute of Medical Biochemistry, Clinical Center of Serbia, School of Pharmacy, University of Belgrade, Belgrade 11000, Serbia
| | - Aleksandra Radosavljević
- Institute of Ophthalmology, Medical Retina Department, Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Lepša Žorić
- Clinic for Eye Diseases, Clinical Center, Faculty of Medicine, Settlement Kosovska Mitrovica, Kosovska Mitrovica 38200, Serbia
| |
Collapse
|
14
|
Song D, Fang G, Greenberg H, Liu SF. Chronic intermittent hypoxia exposure-induced atherosclerosis: a brief review. Immunol Res 2016; 63:121-30. [PMID: 26407987 DOI: 10.1007/s12026-015-8703-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Obstructive sleep apnea (OSA) is highly prevalent in the USA and is recognized as an independent risk factor for atherosclerotic cardiovascular disease. Identification of atherosclerosis risk factor attributable to OSA may provide opportunity to develop preventive measures for cardiovascular risk reduction. Chronic intermittent hypoxia (CIH) is a prominent feature of OSA pathophysiology and may be a major mechanism linking OSA to arteriosclerosis. Animal studies demonstrated that CIH exposure facilitated high-cholesterol diet (HCD)-induced atherosclerosis, accelerated the progression of existing atherosclerosis, and induced atherosclerotic lesions in the absence of other atherosclerosis risk factors, demonstrating that CIH is an independent causal factor of atherosclerosis. Comparative studies revealed major differences between CIH-induced and the classic HCD-induced atherosclerosis. Systemically, CIH was a much weaker inducer of atherosclerosis. CIH and HCD differentially activated inflammatory pathways. Histologically, CIH-induced atherosclerotic plaques had no clear necrotic core, contained a large number of CD31+ endothelial cells, and had mainly elastin deposition, whereas HCD-induced plaques had typical necrotic cores and fibrous caps, contained few endothelial cells, and had mainly collagen deposition. Metabolically, CIH caused mild, but HCD caused more severe dyslipidemia. Mechanistically, CIH did not, but HCD did, cause macrophage foam cell formation. NF-κB p50 gene deletion augmented CIH-induced, but not HCD-induced atherosclerosis. These differences reflect the intrinsic differences between the two types of atherosclerosis in terms of pathological nature and underlying mechanisms and support the notion that CIH-induced atherosclerosis is a new paradigm that differs from the classic HCD-induced atherosclerosis.
Collapse
Affiliation(s)
- Dongmei Song
- Centers for Heart and Lung Research, and Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Hofstra North Shore-LIJ School of Medicine, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Guoqiang Fang
- Centers for Heart and Lung Research, and Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Hofstra North Shore-LIJ School of Medicine, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Harly Greenberg
- Centers for Heart and Lung Research, and Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Hofstra North Shore-LIJ School of Medicine, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Shu Fang Liu
- Centers for Heart and Lung Research, and Pulmonary, Critical Care and Sleep Medicine, The Feinstein Institute for Medical Research, Hofstra North Shore-LIJ School of Medicine, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
15
|
Chellan B, Reardon CA, Getz GS, Hofmann Bowman MA. Enzymatically Modified Low-Density Lipoprotein Promotes Foam Cell Formation in Smooth Muscle Cells via Macropinocytosis and Enhances Receptor-Mediated Uptake of Oxidized Low-Density Lipoprotein. Arterioscler Thromb Vasc Biol 2016; 36:1101-13. [PMID: 27079883 DOI: 10.1161/atvbaha.116.307306] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 03/31/2016] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Enzyme-modified nonoxidized low-density lipoprotein (ELDL) is present in human atherosclerotic lesions. Our objective is to understand the mechanisms of ELDL uptake and its effects on vascular smooth muscle cells (SMC). APPROACH AND RESULTS Transformation of murine aortic SMCs into foam cells in response to ELDL was analyzed. ELDL, but not acetylated or oxidized LDL, was potent in inducing SMC foam cell formation. Inhibitors of macropinocytosis (LY294002, wortmannin, amiloride) attenuated ELDL uptake. In contrast, inhibitors of receptor-mediated endocytosis (dynasore, sucrose) and inhibitor of caveolae-/lipid raft-mediated endocytosis (filipin) had no effect on ELDL uptake in SMC, suggesting that macropinocytosis is the main mechanism of ELDL uptake by SMC. Receptor for advanced glycation end products (RAGE) is not obligatory for ELDL-induced SMC foam cell formation, but primes SMC for the uptake of oxidized LDL in a RAGE-dependent manner. ELDL increased intracellular reactive oxygen species, cytosolic calcium, and expression of lectin-like oxidized LDL receptor-1 in wild-type SMC but not in RAGE(-/-) SMC. The macropinocytotic uptake of ELDL is regulated predominantly by intracellular calcium because ELDL uptake was completely inhibited by pretreatment with the calcium channel inhibitor lacidipine in wild-type and RAGE(-/-) SMC. This is in contrast to pretreatment with PI3 kinase inhibitors which completely prevented ELDL uptake in RAGE(-/-) SMC, but only partially in wild-type SMC. CONCLUSIONS ELDL is highly potent in inducing foam cells in murine SMC. ELDL endocytosis is mediated by calcium-dependent macropinocytosis. Priming SMC with ELDL enhances the uptake of oxidized LDL.
Collapse
Affiliation(s)
- Bijoy Chellan
- From the Department of Medicine (B.C., M.A.H.B.), and Department of Pathology (C.A.R., G.S.G), University of Chicago, IL.
| | - Catherine A Reardon
- From the Department of Medicine (B.C., M.A.H.B.), and Department of Pathology (C.A.R., G.S.G), University of Chicago, IL
| | - Godfrey S Getz
- From the Department of Medicine (B.C., M.A.H.B.), and Department of Pathology (C.A.R., G.S.G), University of Chicago, IL
| | - Marion A Hofmann Bowman
- From the Department of Medicine (B.C., M.A.H.B.), and Department of Pathology (C.A.R., G.S.G), University of Chicago, IL.
| |
Collapse
|
16
|
Dubland JA, Francis GA. So Much Cholesterol: the unrecognized importance of smooth muscle cells in atherosclerotic foam cell formation. Curr Opin Lipidol 2016; 27:155-61. [PMID: 26836481 DOI: 10.1097/mol.0000000000000279] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Smooth muscle cells (SMCs) form the thickened intimal layer in atherosclerosis-prone arteries in early life, and provide the initial site for retention and uptake of atherogenic lipoproteins. Here we review current knowledge regarding the importance of SMCs in the deposition of cholesterol in atherosclerotic plaque. RECENT FINDINGS SMCs were found to comprise at least 50% of total foam cells in human coronary artery atherosclerosis, and exhibit a selective loss of expression of the cholesterol efflux promoter ATP-binding cassette transporter A1. Cholesterol loading induced a loss of SMC gene expression and an increase in macrophage and proinflammatory marker expression by cultured mouse and human arterial SMCs, with reversal of these effects upon removal of the excess cholesterol. Mice engineered to track all cells of SMC lineage indicated that, at most, SMCs make up about one-third of total cells in atherosclerotic plaque in these animals. SUMMARY SMCs appear to be the origin of the majority of foam cells in human atherosclerotic plaque. Recent studies suggest a renaissance of research on the role of SMCs in atherosclerosis is needed to make the next leap forward in the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Joshua A Dubland
- Division of Endocrinology and Metabolism, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
17
|
Palomino‐Morales R, Perales S, Torres C, Linares A, Alejandre MJ. Cholesterol loading in vivo and in vitro alters extracellular matrix proteins production in smooth muscle cells. EUR J LIPID SCI TECH 2015. [DOI: 10.1002/ejlt.201500287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Rogelio Palomino‐Morales
- Department of Biochemistry and Molecular Biology IFaculty of SciencesCampus Universitario de Fuentenueva, University of GranadaSpain
| | - Sonia Perales
- Department of Biochemistry and Molecular Biology IFaculty of SciencesCampus Universitario de Fuentenueva, University of GranadaSpain
| | - Carolina Torres
- Department of Biochemistry and Molecular Biology IFaculty of SciencesCampus Universitario de Fuentenueva, University of GranadaSpain
| | - Ana Linares
- Department of Biochemistry and Molecular Biology IFaculty of SciencesCampus Universitario de Fuentenueva, University of GranadaSpain
| | - Maria Jose Alejandre
- Department of Biochemistry and Molecular Biology IFaculty of SciencesCampus Universitario de Fuentenueva, University of GranadaSpain
| |
Collapse
|
18
|
Vengrenyuk Y, Nishi H, Long X, Ouimet M, Savji N, Martinez FO, Cassella CP, Moore KJ, Ramsey SA, Miano JM, Fisher EA. Cholesterol loading reprograms the microRNA-143/145-myocardin axis to convert aortic smooth muscle cells to a dysfunctional macrophage-like phenotype. Arterioscler Thromb Vasc Biol 2015; 35:535-46. [PMID: 25573853 PMCID: PMC4344402 DOI: 10.1161/atvbaha.114.304029] [Citation(s) in RCA: 272] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We previously showed that cholesterol loading in vitro converts mouse aortic vascular smooth muscle cells (VSMC) from a contractile state to one resembling macrophages. In human and mouse atherosclerotic plaques, it has become appreciated that ≈40% of cells classified as macrophages by histological markers may be of VSMC origin. Therefore, we sought to gain insight into the molecular regulation of this clinically relevant process. APPROACH AND RESULTS VSMC of mouse (or human) origin were incubated with cyclodextrin-cholesterol complexes for 72 hours, at which time the expression at the protein and mRNA levels of contractile-related proteins was reduced and of macrophage markers increased. Concurrent was downregulation of miR-143/145, which positively regulate the master VSMC differentiation transcription factor myocardin. Mechanisms were further probed in mouse VSMC. Maintaining the expression of myocardin or miR-143/145 prevented and reversed phenotypic changes caused by cholesterol loading. Reversal was also seen when cholesterol efflux was stimulated after loading. Notably, despite expression of macrophage markers, bioinformatic analyses showed that cholesterol-loaded cells remained closer to the VSMC state, consistent with impairment in classical macrophage functions of phagocytosis and efferocytosis. In apoE-deficient atherosclerotic plaques, cells positive for VSMC and macrophage markers were found lining the cholesterol-rich necrotic core. CONCLUSIONS Cholesterol loading of VSMC converts them to a macrophage-appearing state by downregulating the miR-143/145-myocardin axis. Although these cells would be classified by immunohistochemistry as macrophages in human and mouse plaques, their transcriptome and functional properties imply that their contributions to atherogenesis would not be those of classical macrophages.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Binding Sites
- Cell Lineage
- Cell Transdifferentiation
- Cholesterol/metabolism
- Cholesterol, HDL/metabolism
- Coculture Techniques
- Disease Models, Animal
- Foam Cells/metabolism
- Foam Cells/pathology
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Humans
- Jurkat Cells
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Necrosis
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Oligonucleotide Array Sequence Analysis
- Phagocytosis
- Phenotype
- Plaque, Atherosclerotic
- Signal Transduction
- Sterol Regulatory Element Binding Protein 2/metabolism
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transfection
Collapse
Affiliation(s)
- Yuliya Vengrenyuk
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Hitoo Nishi
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Xiaochun Long
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Mireille Ouimet
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Nazir Savji
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Fernando O Martinez
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Courtney P Cassella
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Kathryn J Moore
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Stephen A Ramsey
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Joseph M Miano
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Edward A Fisher
- From the Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (Y.V., H.N., M.O., N.S., C.P.C., K.J.M., E.A.F.); Center for Cardiovascular Sciences, Albany Medical College, NY (X.L.); Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom (F.O.M.); Department of Biomedical Sciences, Oregon State University, Corvallis (S.A.R.); and Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.).
| |
Collapse
|
19
|
Atherosclerosis regulation via media lipid-driven VSMC cholesterol efflux switch. Med Hypotheses 2015; 84:141-4. [DOI: 10.1016/j.mehy.2014.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 11/27/2014] [Accepted: 12/04/2014] [Indexed: 02/02/2023]
|
20
|
WANG HUAN, LIU YAN, ZHU LING, WANG WENJING, WAN ZHAOFEI, CHEN FANGYUAN, WU YAN, ZHOU JUAN, YUAN ZUYI. 17β-estradiol promotes cholesterol efflux from vascular smooth muscle cells through a liver X receptor α-dependent pathway. Int J Mol Med 2014; 33:550-8. [DOI: 10.3892/ijmm.2014.1619] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 12/23/2013] [Indexed: 11/05/2022] Open
|
21
|
Garrido-Urbani S, Meguenani M, Montecucco F, Imhof BA. Immunological aspects of atherosclerosis. Semin Immunopathol 2014; 36:73-91. [PMID: 24212253 DOI: 10.1007/s00281-013-0402-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 10/15/2013] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is a complex chronic inflammatory and metabolic disease that involves the collaboration of several cellular components of the immune system and results in thickening of the arterial wall. Atherosclerosis is also the primary cause of coronary artery and cerebrovascular diseases. A multitude of immune cell subsets, soluble molecules such as chemokines and cytokines, and circulating lipids play pivotal roles in atherosclerosis development. In this review, we highlight the role of the immune system in the course of atherosclerotic disease development and discuss the mechanisms involved.
Collapse
Affiliation(s)
- S Garrido-Urbani
- Department of Pathology and Immunology, CMU, University of Geneva, Geneva, Switzerland,
| | | | | | | |
Collapse
|
22
|
Lipoproteínas modificadas como marcadores de riesgo cardiovascular en la diabetes mellitus. ACTA ACUST UNITED AC 2013; 60:518-28. [DOI: 10.1016/j.endonu.2012.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/17/2012] [Accepted: 12/19/2012] [Indexed: 11/17/2022]
|
23
|
Rivera J, Walduck AK, Thomas SR, Glaros EN, Hooker EU, Guida E, Sobey CG, Drummond GR. Accumulation of serum lipids by vascular smooth muscle cells involves a macropinocytosis-like uptake pathway and is associated with the downregulation of the ATP-binding cassette transporter A1. Naunyn Schmiedebergs Arch Pharmacol 2013; 386:1081-93. [PMID: 23989929 DOI: 10.1007/s00210-013-0909-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/12/2013] [Indexed: 01/01/2023]
Abstract
Vascular smooth muscle cells (VSMC) are present in arterial intima before atherosclerotic plaques develop and are likely to be exposed to unmodified serum lipids as they enter the vessel wall. We examined the effects of sera from mice on the morphology and function of mouse VSMC. Incubation of a mouse VSMC line (MOVAS) with sera from normocholesterolemic (C57BL/6J) or hypercholesterolemic (APOE(-/-)) mice caused concentration-dependent increases in lipid accumulation as measured by AdipoRed, with the extent of lipid uptake significantly greater with the latter sera type. Inhibition of c-Jun N-terminal kinases (SP600125), Src kinases (AG1879), and clathrin-dependent endocytosis (monodansylcadaverine) to disrupt scavenger receptor-mediated uptake of lipids had no effect on serum-induced lipid accumulation by VSMC. By contrast, inhibition of macropinocytosis with antagonists of PI-3 kinase (LY294002) and actin (cytochalasin D) markedly reduced lipid accumulation. Serum exposure reduced the expression of the ATP-binding cassette transporter A1, consistent with impaired cholesterol efflux, but had no effect on the expression of markers of VSMC differentiation. Moreover, the expression of several inflammation and foam cell markers was unchanged (CCL2, CCL5, and CD68) by mouse sera. The accumulation of unmodified serum lipids by VSMC involves a macropinocytosis-like uptake pathway and is associated with the downregulation of the ATP-binding cassette transporter. We speculate that VSMC may play an atheroprotective role in arterial intima by acting as a "sink" for unmodified lipids.
Collapse
Affiliation(s)
- Jennifer Rivera
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Building 13E, Wellington Road, Clayton, VIC, 3800, Australia
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Hung PH, Tsai HB, Lin CH, Hung KY. Abdominal obesity is associated with peripheral artery disease in hemodialysis patients. PLoS One 2013; 8:e67555. [PMID: 23840739 PMCID: PMC3695898 DOI: 10.1371/journal.pone.0067555] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 05/20/2013] [Indexed: 01/09/2023] Open
Abstract
Background Peripheral arterial disease (PAD) is a leading cause of morbidity in hemodialysis (HD) patients. Recent evidence suggests that abdominal obesity (AO) may play a role in PAD. However, the association between AO and PAD has not been thoroughly studied in HD patients. Methods The present cross-sectional study aimed to examine the relationship between AO and PAD in a cohort of 204 chronic HD patients. The ankle brachial index (ABI) was used as an estimate of the presence of PAD. Plasma adiponectin levels, interleukin-6 (IL-6) levels, high sensitivity C-reactive protein (hs-CRP) levels, asymmetric dimethylarginine (ADMA) levels, and lipid profiles were measured. Logistic regression was used to estimate the association between the presence of PAD and AO as well as other potential risk factors. Results The metabolic risk factors and all individual traits, including elevated ln-transformed hs-CRP, were found to be significant (P<0.05) more frequently in HD patients with AO than that in control subjects. Patients with AO had a higher prevalence of PAD than the control individuals, with a mean ABI of 0.96±0.23 and 1.08±0.16 (P<0.0001) and PAD prevalence of 26.9% and 10.8% (P = 0.003), respectively. By multivariate analysis, AO (odds ratio [OR], 4.532; 95% CI, 1.765–11.639; P = 0.002), elevated serum ln-transformed ADMA (OR, 5.535; 95% CI, 1.323–23.155; P = 0.019), and ln-transformed IL-6 (OR, 1.567; 95% CI, 1.033–2.378; P = 0.035) were independent predictors of the presence of PAD. Conclusions HD patients with AO exhibited a cluster of metabolic risk factors and lower ABI. AO, elevated serum ln-transformed ADMA, and ln-transformed IL-6 were independent predictors of the presence of PAD.
Collapse
Affiliation(s)
- Peir-Haur Hung
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-yi Christian Hospital, Chia-yi, Taiwan
| | | | | | | |
Collapse
|
25
|
Patients with unrecognized peripheral arterial disease (PAD) assessed by ankle-brachial index (ABI) present a defined profile of proinflammatory markers compared to healthy subjects. Cytokine 2012; 59:294-8. [PMID: 22595645 DOI: 10.1016/j.cyto.2012.04.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 04/19/2012] [Accepted: 04/25/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Many studies have postulated that atherosclerosis should be considered as an inflammatory disease. In addition, some studies have focused on the relationship between inflammation and peripheral arterial disease (PAD). OBJECTIVE Define the plasma levels of soluble markers, including the proinflammatory cytokine interleukin-6 (IL-6), the anti-inflammatory cytokine transforming growth factor-β1 (TGF-β1), the endothelial-specific adhesion factor (E-selectin) and two proteinases involved in extracellular matrix degradation (matrix metalloproteinases-2 and -9, MMP-2, and MMP-9) in previously unrecognized patients with peripheral artery disease (PAD) and non-PAD controls. RESULTS Significantly higher levels of IL-6, E-selectin and MMP-2/MMP-9 and significantly reduced levels of TGF-β1 were found in PAD patients (ankle-brachial index, ABI⩽0.9) compared to non-PAD control subjects (1.4>ABI>0.9). CONCLUSION The results demonstrated the subjects with unrecognized PAD (ABI⩽0.9) show a characteristic phlogistic pattern differently from healthy subjects and it strongly supports the pivotal role played by inflammatory and immunological mechanisms in the initiation and progression of the atherosclerotic process in peripheral arteries. These biomarkers could be helpful to screen the susceptibility for the diseases in peripheral arteries.
Collapse
|
26
|
Allahverdian S, Pannu PS, Francis GA. Contribution of monocyte-derived macrophages and smooth muscle cells to arterial foam cell formation. Cardiovasc Res 2012; 95:165-72. [PMID: 22345306 DOI: 10.1093/cvr/cvs094] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Smooth muscle cells (SMCs) are the main cell type in intimal thickenings and some stages of human atherosclerosis. Like monocyte-derived macrophages, SMCs accumulate excess lipids and contribute to the total intimal foam cell population. In contrast, apolipoprotein (Apo)E-deficient and LDL receptor-deficient mice develop atherosclerotic lesions that are macrophage- as opposed to SMC-rich. The lesser contribution of SMCs to lesion development in these mouse models has distracted attention away from the importance of SMC cholesterol homeostasis in the artery wall. Intimal SMCs accumulate excess amounts of cholesteryl esters when compared with medial layer SMCs, possibly explained by reduced ATP-binding cassette transporter A1 expression and ApoA-I binding to intimal-type SMCs. The aim of this review is to compare the relative contribution of monocyte-derived macrophages and SMCs to human vs. mouse atherosclerosis, and describe what is known about lipid uptake and removal mechanisms contributing to arterial macrophage and SMC foam cell formation. An increased understanding of the contribution of these cell types to lesion development will help to delineate their relative importance in atherogenesis and as potential therapeutic targets.
Collapse
Affiliation(s)
- Sima Allahverdian
- Department of Medicine, UBC James Hogg Research Centre, Providence Heart + Lung Institute at St Paul's Hospital, Room 166, Burrard Building, 1081 Burrard Street, Vancouver, BC, Canada V6Z 1Y6
| | | | | |
Collapse
|
27
|
Effect of PGC-1α on proliferation, migration, and transdifferentiation of rat vascular smooth muscle cells induced by high glucose. J Biomed Biotechnol 2012; 2012:756426. [PMID: 22461724 PMCID: PMC3303719 DOI: 10.1155/2012/756426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 11/04/2011] [Accepted: 11/08/2011] [Indexed: 11/23/2022] Open
Abstract
We assessed the role of PGC-1α (PPARγ coactivator-1 alpha) in glucose-induced proliferation, migration, and inflammatory gene expression of vascular smooth muscle cells (VSMCs). We carried out phagocytosis studies to assess the role of PGC-1α in transdifferentiation of VSMCs by flow cytometry. We found that high glucose stimulated proliferation, migration and inflammatory gene expression of VSMCs, but overexpression of PGC-1α attenuated the effects of glucose. In addition, overexpression of PGC-1α decreased mRNA and protein level of VSMCs-related genes, and induced macrophage-related gene expression, as well as phagocytosis of VSMCs. Therefore, PGC-1α inhibited glucose-induced proliferation, migration and inflammatory gene expression of VSMCs, which are key features in the pathology of atherosclerosis. More importantly, PGC-1α transdifferentiated VSMCs to a macrophage-like state. Such transdifferentiation possibly increased the portion of VSMCs-derived foam cells in the plaque and favored plaque stability.
Collapse
|
28
|
Vascular effects of glycoprotein130 ligands--part I: pathophysiological role. Vascul Pharmacol 2011; 56:34-46. [PMID: 22197898 DOI: 10.1016/j.vph.2011.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 12/02/2011] [Accepted: 12/09/2011] [Indexed: 12/25/2022]
Abstract
The vessel wall is no longer considered as only an anatomical barrier for blood cells but is recognized as an active endocrine organ. Dysfunction of the vessel wall occurs in various disease processes including atherosclerosis, hypertension, peripheral artery disease, aneurysms, and transplant and diabetic vasculopathies. Different cytokines were shown to modulate the behavior of the cells, which constitute the vessel wall such as immune cells, endothelial cells and smooth muscle cells. Glycoprotein 130 (gp130) is a common cytokine receptor that controls the activity of a group of cytokines, namely, interleukin (IL)-6, oncostatin M (OSM), IL-11, ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), cardiotrophin-1 (CT-1), cardiotrophin-like cytokine (CLC), IL-27, and neuropoietin (NP). Gp130 and associated cytokines have abundantly diverse functions. Part I of this review focuses on the pathophysiological functions of gp130 ligands. We specifically describe vascular effects of these molecules and discuss the respective underlying molecular and cellular mechanisms.
Collapse
|
29
|
Kleinedler JJ, Orchard EA, Foley JD, Rogers LK, Hebert VY, Dugas TR. A dietary approach to increase in-stent stenosis and face validity of a rat model for arterial angioplasty and stenting. Atherosclerosis 2011; 219:484-91. [DOI: 10.1016/j.atherosclerosis.2011.09.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 08/18/2011] [Accepted: 09/13/2011] [Indexed: 11/25/2022]
|
30
|
Ma S, Yang D, Li D, Tang B, Yang Y. Oleic acid induces smooth muscle foam cell formation and enhances atherosclerotic lesion development via CD36. Lipids Health Dis 2011; 10:53. [PMID: 21486455 PMCID: PMC3083368 DOI: 10.1186/1476-511x-10-53] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 04/12/2011] [Indexed: 11/14/2022] Open
Abstract
Background Elevated plasma free fatty acid (FFA) levels have been linked to the development of atherosclerosis. However, how FFA causes atherosclerosis has not been determined. Because fatty acid translocase (FAT/CD36) is responsible for the uptake of FFA, we hypothesized that the atherogenic effects of FFA may be mediated via CD36. Results We tested this hypothesis using cultured rat aortic smooth muscle cells (SMCs) treated with oleic acid (OA). We found that OA induces lipid accumulation in SMCs in a dose dependent manner. Rat aortic SMCs treated for 48 hours with OA (250 μmol/L) became foam cells based on morphological (Oil Red O staining) and biochemical (5 times increase in cellular triglyceride) criteria. Moreover, specific inhibition of CD36 by sulfo-N-succinimidyl oleate significantly attenuated OA induced lipid accumulation and foam cell formation. To confirm these results in vivo, we used ApoE-deficient mice fed with normal chow (NC), OA diet, NC plus lipolysis inhibitor acipimox or OA plus acipimox. OA-fed mice showed increased plasma FFA levels and enhanced atherosclerotic lesions in the aortic sinus compared to the NC group (both p < 0.01). This effect was partially reversed by acipimox (lesion area: OA: 3.09 ± 0.10 ×105 μm2 vs. OA plus acipimox: 2.60 ± 0.10 ×105 μm2, p < 0.05; FFA: OA: 0.91 ± 0.03 mmol/L vs. OA plus acipimox: 0.78 ± 0.03 mmol/L, p < 0.05). Conclusions These findings suggest that OA induces smooth muscle foam cell formation and enhances atherosclerotic lesions in part though CD36. Furthermore, these findings provide a novel model for the investigation of atherosclerosis.
Collapse
Affiliation(s)
- Shuangtao Ma
- Department of Cardiology, General Hospital of PLA Chengdu Military Area Command, PR China
| | | | | | | | | |
Collapse
|
31
|
Mechanisms involved in IL-6-induced muscular mechanical hyperalgesia in mice. Pain 2010; 151:345-355. [DOI: 10.1016/j.pain.2010.07.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 07/06/2010] [Accepted: 07/15/2010] [Indexed: 01/19/2023]
|
32
|
Carvalho MDT, Vendrame CMV, Ketelhuth DFJ, Yamashiro-Kanashiro EH, Goto H, Gidlund M. High-Density Lipoprotein Inhibits the Uptake of Modified Low- Density Lipoprotein and the Expression of CD36 and FcγRI. J Atheroscler Thromb 2010; 17:844-57. [DOI: 10.5551/jat.3905] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
33
|
Kedi X, Ming Y, Yongping W, Yi Y, Xiaoxiang Z. Free cholesterol overloading induced smooth muscle cells death and activated both ER- and mitochondrial-dependent death pathway. Atherosclerosis 2009; 207:123-30. [DOI: 10.1016/j.atherosclerosis.2009.04.019] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Revised: 04/08/2009] [Accepted: 04/11/2009] [Indexed: 01/22/2023]
|
34
|
Endolysosomal phospholipidosis and cytosolic lipid droplet storage and release in macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2008; 1791:524-39. [PMID: 19146988 DOI: 10.1016/j.bbalip.2008.12.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 12/08/2008] [Accepted: 12/11/2008] [Indexed: 12/12/2022]
Abstract
This review summarizes the current knowledge of endolysosomal and cytoplasmic lipid storage in macrophages induced by oxidized LDL (Ox-LDL), enzymatically degraded LDL (E-LDL) and other atherogenic lipoprotein modifications, and their relation to the adapter protein 3 (AP-3) dependent ABCA1 and ABCG1 cellular lipid efflux pathways. We compare endolysosomal lipid storage caused either through drug induced phospholipidosis, inheritable endolysosomal and cytosolic lipid storage disorders and Ox-LDL or E-LDL induced phagosomal uptake and cytosolic lipid droplet storage in macrophages. Ox-LDL is resistant to rapid endolysosomal hydrolysis and is trapped within the endolysosomal compartment generating lamellar bodies which resemble the characteristics of phospholipidosis. Various inherited lysosomal storage diseases including sphingolipidosis, glycosphingolipidosis and cholesterylester storage diseases also present a phospholipidosis phenotype. In contrast E-LDL resembling coreless unesterified cholesterol enriched LDL-particles, with a multilamellar, liposome-like structure, lead to rapid phagosomal degradation and cytosolic lipid droplet accumulation. As a consequence the uptake of E-LDL through type I and type II phagocytosis leads to increased lipid droplet formation and moderate upregulation of ABCA1 and ABCG1 while uptake of Ox-LDL leads to a rapid expansion of the lysosomal compartment and a pronounced upregulation of the ABCA1/ABCG1/AP-3 lipid efflux pathway.
Collapse
|
35
|
Loppnow H, Werdan K, Buerke M. Vascular cells contribute to atherosclerosis by cytokine- and innate-immunity-related inflammatory mechanisms. Innate Immun 2008; 14:63-87. [PMID: 18713724 DOI: 10.1177/1753425908091246] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases are the human diseases with the highest death rate and atherosclerosis is one of the major underlying causes of cardiovascular diseases. Inflammatory and innate immune mechanisms, employing monocytes, innate receptors, innate cytokines, or chemokines are suggested to be involved in atherogenesis. Among the inflammatory pathways the cytokines are central players. Plasma levels of cytokines and related proteins, such as CRP, have been investigated in cardiovascular patients, tissue mRNA expression was analyzed and correlations to vascular diseases established. Consistent with these findings the generation of cytokine-deficient animals has provided direct evidence for a role of cytokines in atherosclerosis. In vitro cell culture experiments further support the suggestion that cytokines and other innate mechanisms contribute to atherogenesis. Among the initiation pathways of atherogenesis are innate mechanisms, such as toll-like-receptors (TLRs), including the endotoxin receptor TLR4. On the other hand, innate cytokines, such as IL-1 or TNF, or even autoimmune triggers may activate the cells. Cytokines potently activate multiple functions relevant to maintain or spoil homeostasis within the vessel wall. Vascular cells, not least smooth muscle cells, can actively contribute to the inflammatory cytokine-dependent network in the blood vessel wall by: (i) production of cytokines; (ii) response to these potent cell activators; and (iii) cytokine-mediated interaction with invading cells, such as monocytes, T-cells, or mast cells. Activation of these pathways results in accumulation of cells and increased LDL- and ECM-deposition which may serve as an 'immunovascular memory' resulting in an ever-growing response to subsequent invasions. Thus, vascular cells may potently contribute to the inflammatory pathways involved in development and acceleration of atherosclerosis.
Collapse
Affiliation(s)
- Harald Loppnow
- Martin-Luther-Universität Halle-Wittenberg, Universitätsklinik und Poliklinik für Innere Medizin , Halle (Saale), Germany.
| | | | | |
Collapse
|
36
|
Bengtsson T, Karlsson H, Gunnarsson P, Skoglund C, Elison C, Leanderson P, Lindahl M. The periodontal pathogen Porphyromonas gingivalis cleaves apoB-100 and increases the expression of apoM in LDL in whole blood leading to cell proliferation. J Intern Med 2008; 263:558-71. [PMID: 18248365 DOI: 10.1111/j.1365-2796.2007.01917.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Several studies support an association between periodontal disease and atherosclerosis with a crucial role for the pathogen Porphyromonas gingivalis. This study aims at investigating the proteolytic and oxidative activity of P. gingivalis on LDL in a whole blood system using a proteomic approach and analysing the effects of P. gingivalis-modified LDL on cell proliferation. METHODS The cellular effects of P. gingivalis in human whole blood were assessed using lumi-aggregometry analysing reactive oxygen species production and aggregation. Blood was incubated for 30 min with P. gingivalis, whereafter LDL was isolated and a proteomic approach was applied to examine protein expression. LDL-oxidation was determined by analysing the formation of protein carbonyls. The effects of P. gingivalis-modified LDL on fibroblast proliferation were studied using the MTS assay. RESULTS Incubation of whole blood with P. gingivalis caused an extensive aggregation and ROS production, indicating platelet and leucocyte activation. LDL prepared from bacteria-exposed blood showed an increased protein oxidation, elevated levels of apoM and formation of two apoB-100 N-terminal fragments. Porphyromonas gingivalis-modified LDL markedly increased the growth of fibroblasts. Inhibition of gingipain R suppressed the modification of LDL by P. gingivalis. CONCLUSIONS The ability of P. gingivalis to change the protein expression and proliferative capacity of LDL may represent a crucial event in periodontitis-associated atherosclerosis.
Collapse
Affiliation(s)
- T Bengtsson
- Division of Pharmacology, Department of Medical and Health Sciences, Faculty of Health Sciences, Cardiovascular Inflammation Research Centre, Linköping University, Linköping, Sweden.
| | | | | | | | | | | | | |
Collapse
|
37
|
Girn HRS, Orsi NM, Homer-Vanniasinkam S. An overview of cytokine interactions in atherosclerosis and implications for peripheral arterial disease. Vasc Med 2008; 12:299-309. [PMID: 18048466 DOI: 10.1177/1358863x07083387] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Over the last three decades, a surge in research into the inflammatory pathophysiology of atherosclerosis has highlighted an array of cytokines and other inflammatory mediators associated with underlying inflammatory burden. The ability to identify and simultaneously measure multiple cytokines in peripheral blood highlights their potential as biomarkers of atherosclerosis. This has prompted much research in vascular medicine to identify the ;at-risk' groups for atherostenotic or atheroaneurysmal disease. This review is compiled with similar intentions and aims to discern the relevant evidence for cytokine profiling in peripheral arterial disease (PAD), where such information is lacking, while providing a holistic overview of cytokine interactions in atherosclerosis. This is pertinent given that cytokine profiles from coronary artery disease and aortic aneurysm studies cannot be directly extrapolated to PAD due to differences in inflammatory environments that exist in these conditions. Whilst plaque morphology and blood rheology play an important role in the cardiac manifestations of atherosclerosis, tissue thrombogenecity is very important in PAD. Further, cytokines act in concert rather than in isolation in a disease process, and no single cytokine in a cross-sectional model is able to serve as an absolute screening marker. Thus, it is essential to understand the regulation of cytokine production in atherosclerosis prior to evaluating the viability and merits of a multimarker approach for clinical risk stratification in PAD.
Collapse
Affiliation(s)
- H R S Girn
- Leeds Vascular Institute, Leeds General Infirmary, Leeds, UK.
| | | | | |
Collapse
|
38
|
Sonnenberg S, Shearman C, Baxter S, Morris G, Cumming D, Montgomery H, Rose-Zerilli M, Day I. Level of Ex Vivo Interleukin 6 Expression in Human Peripheral Fat Compared with Other Tissues. Eur J Vasc Endovasc Surg 2008; 35:314-9. [DOI: 10.1016/j.ejvs.2007.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Accepted: 10/01/2007] [Indexed: 01/10/2023]
|
39
|
Doran AC, Meller N, McNamara CA. Role of smooth muscle cells in the initiation and early progression of atherosclerosis. Arterioscler Thromb Vasc Biol 2008; 28:812-9. [PMID: 18276911 DOI: 10.1161/atvbaha.107.159327] [Citation(s) in RCA: 613] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The initiation of atherosclerosis results from complex interactions of circulating factors and various cell types in the vessel wall, including endothelial cells, lymphocytes, monocytes, and smooth muscle cells (SMCs). Recent reviews highlight the role of activated endothelium and inflammatory cell recruitment in the initiation of and progression of early atherosclerosis. Yet, human autopsy studies, in vitro mechanistic studies, and in vivo correlative data suggest an important role for SMCs in the initiation of atherosclerosis. SMCs are the major producers of extracellular matrix within the vessel wall and in response to atherogenic stimuli can modify the type of matrix proteins produced. In turn, the type of matrix present can affect the lipid content of the developing plaque and the proliferative index of the cells that are adherent to it. SMCs are also capable of functions typically attributed to other cell types. Like macrophages, SMCs can express a variety of receptors for lipid uptake and can form foam-like cells, thereby participating in the early accumulation of plaque lipid. Like endothelial cells, SMCs can also express a variety of adhesion molecules such as vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 to which monocytes and lymphocytes can adhere and migrate into the vessel wall. In addition, through these adhesion molecules, SMCs can also stabilize these cells against apoptosis, thus contributing to the early cellularity of the lesion. Like many cells within the developing plaque, SMCs also produce many cytokines such as PDGF, transforming growth factor-beta, IFNgamma, and MCP-1, all of which contribute to the initiation and propagation of the inflammatory response to lipid. Recent advances in SMC-specific gene modulation have enhanced our ability to determine the role of SMCs in early atherogenesis.
Collapse
Affiliation(s)
- Amanda C Doran
- Cardiovascular Division/Department of Medicine, the Cardiovascular Research Center, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
40
|
Lipid homeostasis in macrophages – Implications for atherosclerosis. REVIEWS OF PHYSIOLOGY BIOCHEMISTRY AND PHARMACOLOGY 2008; 160:93-125. [DOI: 10.1007/112_2008_802] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
41
|
Presta M, Camozzi M, Salvatori G, Rusnati M. Role of the soluble pattern recognition receptor PTX3 in vascular biology. J Cell Mol Med 2007; 11:723-38. [PMID: 17760835 PMCID: PMC3823252 DOI: 10.1111/j.1582-4934.2007.00061.x] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Pentraxins act as soluble pattern recognition receptors with a wide range of functions in various pathophysiological conditions. The long-pentraxin PTX3 shares the C-terminal pentraxin-domain with short-pentraxins C-reactive protein and serum amyloid P component and possesses an unique N-terminal domain. These structural features suggest that PTX3 may have both overlapping and distinct biological/ligand recognition properties when compared to short-pentraxins. PTX3 serves as a mechanism of amplification of inflammation and innate immunity. Indeed, vessel wall elements produce high amounts of PTX3 during inflammation and the levels of circulating PTX3 increase in several pathological conditions affecting the cardiovascular system. PTX3 exists as a free or extracellular matrix-associated molecule and it binds the complement fraction C1q. PTX3 binds also apoptotic cells and selected pathogens, playing a role in innate immunity processes. In endothelial cells and macrophages, PTX3 upregulates tissue factor expression, suggesting its action as a regulator of endothelium during thrombogenesis and ischaemic vascular disease. Finally, PTX3 binds the angiogenic fibroblast growth factor-2, thus inhibiting its biological activity. Taken together, these properties point to a role for PTX3 during vascular damage, angiogenesis, atherosclerosis, and restenosis.
Collapse
Affiliation(s)
- Marco Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy.
| | | | | | | |
Collapse
|
42
|
Demyanets S, Kaun C, Rychli K, Rega G, Pfaffenberger S, Afonyushkin T, Bochkov VN, Maurer G, Huber K, Wojta J. The inflammatory cytokine oncostatin M induces PAI-1 in human vascular smooth muscle cells in vitro via PI 3-kinase and ERK1/2-dependent pathways. Am J Physiol Heart Circ Physiol 2007; 293:H1962-8. [PMID: 17604327 DOI: 10.1152/ajpheart.01366.2006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) plays a pivotal role in the regulation of the fibrinolytic system and in the modulation of extracellular proteolysis. Increased PAI-1 was found in atherosclerotic lesions, and high PAI-1 plasma levels were associated with coronary heart disease. Smooth muscle cells (SMC) are a major source of PAI-1 within the vascular wall, and PAI-1 was implicated in SMC migration, proliferation, and apoptosis. We treated human coronary artery SMC (HCASMC) and human aortic SMC (HASMC) with the glycoprotein 130 (gp130) ligands cardiotrophin-1, interleukin-6 (IL-6), leukemia inhibitory factor (LIF), or oncostatin M (OSM). Only OSM increased PAI-1 antigen and activity production significantly in these cells up to 20-fold. OSM upregulated mRNA specific for PAI-1 up to 4.5-fold in these cells. HCASMC and HASMC express gp130, OSM receptor, IL-6 receptor, and LIF receptor. OSM induced extracellular signal-regulated kinase (ERK) 1/2 and Akt phosphorylations in HASMC. A phosphatidylinositol 3-kinase inhibitor and a mitogen-activated protein/extracellular signal-regulated kinase inhibitor reduced Akt and ERK1/2 phosphorylation, respectively, and abolished OSM-induced PAI-1 upregulation. A janus kinase/signal transducer and activator of transcription inhibitor, a p38 mitogen-activated protein kinase inhibitor, or c-Jun NH2-terminal kinase inhibitor I did not inhibit the OSM-dependent PAI-1 induction. OSM enhanced proliferation of both HCASMC and HASMC by 77 and 90%, respectively. We hypothesize that, if the effect of OSM on PAI-1 expression in smooth muscle cells is operative in vivo, it could, via modulation of fibrinolysis and extracellular proteolysis, be involved in the development of vascular pathologies such as plaque progression, destabilization and subsequent thrombus formation, and restenosis and neointima formation.
Collapse
Affiliation(s)
- Svitlana Demyanets
- Department of Internal Medicine II, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Lee KS, Park JH, Lee S, Lim HJ, Choi HE, Park HY. HB-EGF induces delayed STAT3 activation via NF-kappaB mediated IL-6 secretion in vascular smooth muscle cell. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1637-44. [PMID: 17822789 DOI: 10.1016/j.bbamcr.2007.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 06/25/2007] [Accepted: 07/09/2007] [Indexed: 01/07/2023]
Abstract
Heparin-binding EGF-like growth factor (HB-EGF) is a member of the EGF family that binds to and activates EGF receptor, and is expressed in a variety of tissues, predominantly in the lung, heart, brain and skeletal muscle. HB-EGF is known to induce vascular smooth muscle cell (VSMC) proliferation by activating PI3K-Akt and MAPK pathway. However, our preliminary data showed that Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway was also involved in HB-EGF induced VSMC proliferation. More interestingly, HB-EGF (10 ng/ml) induced a biphasic activation of STAT3 (early at 5 min and late at 60-120 min). Therefore, we tried to elucidate the underlying mechanism of this delayed STAT3 activation by HB-EGF in VSMCs. First, we examined the effect of HB-EGF on interleukin-6 (IL-6) mRNA expressions, since IL-6 have been implicated in the regulation of STAT3 activation. According to our data, HB-EGF increased transcription of IL-6, cardiotrophin-1 (CT-1), leukemia inhibitory factor (LIF) and ciliary neurotrophic factor (CNTF). The secretion of IL-6 was also increased by HB-EGF. Furthermore, these HB-EGF-mediated up-regulation of IL-6 mRNA expression and secretion were inhibited by NF-kappaB inhibitor Bay117082 (2.5 microM) treatment suggesting involvement of NF-kappaB pathway. Again, the late activation of STAT3 by HB-EGF was abolished by both Bay117082 and IL-6 neutralizing antibody (1 microg/ml) indicating IL-6 is a key molecule in the delayed activation of STAT3 by HB-EGF. In addition, IL-6 neutralizing antibody inhibited both HB-EGF conditioned media induced STAT3 activation and HB-EGF induced VSMC proliferation. In conclusion, IL-6 plays an important role in the delayed activation of STAT3 and VSMC proliferation induced by HB-EGF.
Collapse
MESH Headings
- Animals
- Antibodies/pharmacology
- Cell Proliferation/drug effects
- Culture Media, Conditioned
- Enzyme Inhibitors/pharmacology
- ErbB Receptors/metabolism
- Heparin-binding EGF-like Growth Factor
- Intercellular Signaling Peptides and Proteins/pharmacology
- Interleukin-6/genetics
- Interleukin-6/metabolism
- Janus Kinases/antagonists & inhibitors
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- NF-kappa B/metabolism
- Phosphorylation/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- STAT3 Transcription Factor/antagonists & inhibitors
- STAT3 Transcription Factor/metabolism
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Kuy-Sook Lee
- Center for Biomedical Sciences, Division of Cardiovascular Diseases, National Institute of Health, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
44
|
Chalaris A, Rabe B, Paliga K, Lange H, Laskay T, Fielding CA, Jones SA, Rose-John S, Scheller J. Apoptosis is a natural stimulus of IL6R shedding and contributes to the proinflammatory trans-signaling function of neutrophils. Blood 2007; 110:1748-55. [PMID: 17567983 DOI: 10.1182/blood-2007-01-067918] [Citation(s) in RCA: 252] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Interleukin 6 (IL6) trans-signaling has emerged as a prominent regulator of immune responses during both innate and acquired immunity. Regulation of IL6 trans-signaling is reliant upon the release of soluble IL6 receptor (sIL6R), which binds IL6 to create an agonistic IL6/sIL6R complex capable of activating cell types that would not normally respond to IL6 itself. Here we show that intrinsic and extrinsic apoptotic stimulation by DNA damage, cytokine deprivation, and Fas stimulation promotes shedding of sIL6R. Apoptosis-induced shedding of the IL6R was caspase dependent but PKC independent, with inhibition of ADAM17 preventing IL6R shedding. Such insight is relevant to the control of acute inflammation, where transition from the initial neutrophil infiltration to a more sustained population of mononuclear cells is essential for the resolution of the inflammatory process. This transitional event is governed by IL6 trans-signaling. This study demonstrates that IL6R is shed from apoptotic human neutrophils. In vivo studies in a murine inflammation model showed that neutrophil depletion resulted in reduced local sIL6R levels and a concomitant decrease in mononuclear cells, suggesting that apoptosis-induced IL6R shedding from neutrophils promotes IL6 trans-signaling and regulates the attraction of monocytic cells involved in the clearance of apoptotic neutrophils.
Collapse
Affiliation(s)
- Athena Chalaris
- Department of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rose-John S, Waetzig GH, Scheller J, Grötzinger J, Seegert D. The IL-6/sIL-6R complex as a novel target for therapeutic approaches. Expert Opin Ther Targets 2007; 11:613-24. [PMID: 17465721 DOI: 10.1517/14728222.11.5.613] [Citation(s) in RCA: 292] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IL-6 plays a pivotal role in immune responses and certain oncologic conditions. The intense investigation of its biological activity and function led to the discovery of two different IL-6-driven signalling pathways. Binding to the membrane-bound IL-6 receptor (mIL-6R, CD126) causes the recruitment of two gp130 co-receptor molecules (CD130) and the activation of intracellular signalling cascades via gp130. Although this classical pathway is mainly limited to hepatocytes, neutrophils, monocytes/macrophages and certain other leukocyte populations, which express IL-6R on their surface, an alternative mechanism has also been described. Proteolytic cleavage of the mIL-6R protein or translation from alternatively spliced mRNA leads to the generation of a soluble form of the IL-6R (sIL-6R), which is likewise able to bind to IL-6. The resulting IL-6/sIL-6R complex is also capable of binding to gp130 and inducing intracellular signalling. Through this so-called 'trans-signalling' mechanism, IL-6 is able to stimulate cells that lack an endogenous mIL-6R. High levels of IL-6 and sIL-6R have been reported in several chronic inflammatory and autoimmune diseases as well as in cancer. Preclinical animal disease models have provided strong evidence that specific blockade of IL-6-regulated signalling pathways represents a promising approach for the therapy of these diseases. An optimised variant of the recently described fusion protein sgp30Fc is now heading towards its clinical evaluation.
Collapse
Affiliation(s)
- Stefan Rose-John
- Biochemisches Institut, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| | | | | | | | | |
Collapse
|
46
|
Omoigui S. The Interleukin-6 inflammation pathway from cholesterol to aging--role of statins, bisphosphonates and plant polyphenols in aging and age-related diseases. Immun Ageing 2007; 4:1. [PMID: 17374166 PMCID: PMC1845171 DOI: 10.1186/1742-4933-4-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 03/20/2007] [Indexed: 12/25/2022]
Abstract
We describe the inflammation pathway from Cholesterol to Aging. Interleukin 6 mediated inflammation is implicated in age-related disorders including Atherosclerosis, Peripheral Vascular Disease, Coronary Artery Disease, Osteoporosis, Type 2 Diabetes, Dementia and Alzheimer's disease and some forms of Arthritis and Cancer. Statins and Bisphosphonates inhibit Interleukin 6 mediated inflammation indirectly through regulation of endogenous cholesterol synthesis and isoprenoid depletion. Polyphenolic compounds found in plants, fruits and vegetables inhibit Interleukin 6 mediated inflammation by direct inhibition of the signal transduction pathway. Therapeutic targets for the control of all the above diseases should include inhibition of Interleukin-6 mediated inflammation.
Collapse
Affiliation(s)
- Sota Omoigui
- Division of Inflammation and Pain Medicine, LA Pain Clinic, 4019 W Rosecrans Ave, Los Angeles, CA 90250, USA.
| |
Collapse
|
47
|
Abstract
Estrogens are correlated with a lower incidence of atherosclerotic vascular disease, but also provide a protective effect on neovascular disorders, such as Kaposi's sarcoma (KS). Estrogens mediate indirect antiatherosclerotic vascular effects by reducing low-density lipoprotein (LDL) levels and by influencing fibrinolysis, and they exert direct actions on vascular cells including vascular relaxation and vasodilatation, thus reducing progression of the lesion. It is increasingly appreciated that the estrogenic effects are mediated not only by the classic genomic action via the specific nuclear hormone receptors ERalpha and ERbeta, but also by distinct rapid, nongenomic actions. Vascular cells have the capacity to express different types of estrogen receptors, and we provide evidence for selective expression of estrogen receptor subtypes on different human vascular cell types. Moreover, we give an overview on the vascular effects of estrogens, selective estrogen receptor modulators (SERMs), and androgens on normal and malignant vascular cells, with particular focus on the protective estrogenic potential on the vasculature.
Collapse
Affiliation(s)
- Mariam Klouche
- Bremer Centre for Laboratory Medicine, 28205 Bremen, Germany.
| |
Collapse
|
48
|
Mitsuyama K, Sata M, Rose-John S. Interleukin-6 trans-signaling in inflammatory bowel disease. Cytokine Growth Factor Rev 2006; 17:451-61. [PMID: 17045835 DOI: 10.1016/j.cytogfr.2006.09.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The pathogenesis of inflammatory bowel disease (IBD) is complex, involving a wide range of molecules including cytokines. Recent investigations support the important role of an interleukin-6 (IL-6) signaling pathway in the development of IBD. However, the molecular mechanisms of this pathway in the intestine remain incompletely understood. The circulating and intestinal levels of IL-6 as well as soluble IL-6 receptor (sIL-6R) are increased in patients with IBD. It is remarkable that the mucosal T cells of IBD patients are extremely resistant to apoptosis and that a large fraction of these cells express membrane-bound gp130 but not IL-6R. The accumulated evidence strongly supports the hypothesis that the development and perpetuation of IBD relies on the increased formation of IL-6/sIL-6R complexes interacting with membrane-bound gp130 on T cells via trans-signaling. These studies suggest that IL-6 trans-signaling may play a role in the development of IBD; they therefore imply the possibility of a selective therapeutic strategy to target this signaling.
Collapse
Affiliation(s)
- Keiichi Mitsuyama
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Asahi-machi 67, Kurume 830-0011, Japan.
| | | | | |
Collapse
|
49
|
Lim HJ, Lee S, Lee KS, Park JH, Jang Y, Lee EJ, Park HY. PPARγ activation induces CD36 expression and stimulates foam cell like changes in rVSMCs. Prostaglandins Other Lipid Mediat 2006; 80:165-74. [PMID: 16939881 DOI: 10.1016/j.prostaglandins.2006.06.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 06/14/2006] [Accepted: 06/14/2006] [Indexed: 11/28/2022]
Abstract
The purpose of the present study was to determine the role of peroxisome proliferator-activated receptor gamma (PPARgamma) activation in smooth muscle cell (SMC) derived form cell formation. Wild and mutant type PPARgamma were delivered by adenovirus then activated with troglitazone. The result of Oil Red O staining and FACS analysis showed that PPARgamma activation induced lipid accumulation in rVSMCs. Furthermore, PPARgamma activation reduced SMC marker genes such as alpha-actin while induced adipocyte differentiation marker genes and lipid metabolism-related genes as evidenced by RT-PCR and fluorescent immunocytochemistry. All these data demonstrate that PPARgamma activation can drive foam cell like change in rVSMCs. Our results strongly suggest that PPARgamma expression induces CD36 expression and adipocyte differentiation gene activation in the process of atherosclerosis and might be one of the crucial events in SMC derived foam cell formation.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1
- ATP-Binding Cassette Transporters/genetics
- Actins/genetics
- Adiponectin/genetics
- Animals
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Cells, Cultured
- Chromans/pharmacology
- Complement Factor D/genetics
- Fatty Acid-Binding Proteins/genetics
- Foam Cells/cytology
- Foam Cells/metabolism
- Gene Expression/drug effects
- Hypoglycemic Agents/pharmacology
- Lipid Metabolism/drug effects
- Mice
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- PPAR alpha/genetics
- PPAR gamma/agonists
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Platelet-Derived Growth Factor/pharmacology
- Prostaglandin D2/analogs & derivatives
- Prostaglandin D2/pharmacology
- Rats
- Rats, Sprague-Dawley
- Thiazolidinediones/pharmacology
- Transfection
- Troglitazone
- Tropomyosin/genetics
Collapse
Affiliation(s)
- Hyun-Joung Lim
- Division of Cardiovascular Diseases, Center for Biomedical Sciences, NIH, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
50
|
Jones SA. Directing transition from innate to acquired immunity: defining a role for IL-6. THE JOURNAL OF IMMUNOLOGY 2005; 175:3463-8. [PMID: 16148087 DOI: 10.4049/jimmunol.175.6.3463] [Citation(s) in RCA: 565] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Appropriate control of leukocyte recruitment and activation is a fundamental requirement for competent host defense and resolving inflammation. A pivotal event that defines the successful outcome of any inflammatory event is the transition from innate to acquired immunity. In IL-6 deficiency, this process appears defective, and a series of in vivo studies have documented important roles for IL-6 in both the resolution of innate immunity and the development of acquired immune responses. Within this review, particular attention will be given to the regulatory properties of the soluble IL-6 receptor and how its activity may affect chronic disease progression.
Collapse
Affiliation(s)
- Simon A Jones
- Department of Medical Biochemistry and Immunology, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
| |
Collapse
|