1
|
Kowara M, Kopka M, Kopka K, Głowczyńska R, Mitrzak K, Kim DA, Sadowski KA, Cudnoch-Jędrzejewska A. MicroRNA Inhibiting Atheroprotective Proteins in Patients with Unstable Angina Comparing to Chronic Coronary Syndrome. Int J Mol Sci 2024; 25:10621. [PMID: 39408950 PMCID: PMC11476700 DOI: 10.3390/ijms251910621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Patients with unstable angina present clinical characteristics of atherosclerotic plaque vulnerability, contrary to chronic coronary syndrome patients. The process of athersclerotic plaque destabilization is also regulated by microRNA particles. In this study, the investigation on expression levels of microRNAs inhibiting the expression of proteins that protect from atherosclerotic plaque progression (miR-92a inhibiting KLF2, miR-10b inhibiting KLF4, miR-126 inhibiting MerTK, miR-98 inhibiting IL-10, miR-29b inhibiting TGFβ1) was undertaken. A number of 62 individuals were enrolled-unstable angina (UA, n = 14), chronic coronary syndrome (CCS, n = 38), and healthy volunteers (HV, n = 10). Plasma samples were taken, and microRNAs expression levels were assessed by qRT-PCR. As a result, the UA patients presented significantly increased miR-10b levels compared to CCS patients (0.097 vs. 0.058, p = 0.033). Moreover, in additional analysis when UA patients were grouped together with stable patients with significant plaque in left main or proximal left anterior descending ("UA and LM/proxLAD" group, n = 29 patients) and compared to CCS patients with atherosclerotic lesions in other regions of coronary circulation ("CCS other" group, n = 25 patients) the expression levels of both miR-10b (0.104 vs. 0.046; p = 0.0032) and miR-92a (92.64 vs. 54.74; p = 0.0129) were significantly elevated. In conclusion, the study revealed significantly increased expression levels of miR-10b and miR-92a, a regulator of endothelial protective KLF factors (KLF4 and KLF2, respectively) in patients with more vulnerable plaque phenotypes.
Collapse
Affiliation(s)
- Michał Kowara
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland (A.C.-J.)
| | - Michał Kopka
- Department of Methodology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| | - Karolina Kopka
- Department of Methodology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland
| | - Renata Głowczyńska
- 1st Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Karolina Mitrzak
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland (A.C.-J.)
- 1st Department of Cardiology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Dan-ae Kim
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland (A.C.-J.)
| | - Karol Artur Sadowski
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland (A.C.-J.)
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1b Banacha Street, 02-097 Warsaw, Poland (A.C.-J.)
| |
Collapse
|
2
|
Edsfeldt A, Singh P, Matthes F, Tengryd C, Cavalera M, Bengtsson E, Dunér P, Volkov P, Karadimou G, Gisterå A, Orho-Melander M, Nilsson J, Sun J, Gonçalves I. Transforming growth factor-β2 is associated with atherosclerotic plaque stability and lower risk for cardiovascular events. Cardiovasc Res 2023; 119:2061-2073. [PMID: 37200403 PMCID: PMC10478752 DOI: 10.1093/cvr/cvad079] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 01/27/2023] [Accepted: 02/21/2023] [Indexed: 05/20/2023] Open
Abstract
AIMS Transforming growth factor-beta (TGF-β) exists in three isoforms TGF-β1, -β2, and -β3. TGF-β1 has been suggested to be important for maintaining plaque stability, yet the role of TGF-β2 and -β3 in atherosclerosis remains to be investigated.This study explores the association of the three isoforms of TGF-β with plaque stability in the human atherosclerotic disease. METHODS AND RESULTS TGF-β1, -β2, and -β3 proteins were quantified in 223 human carotid plaques by immunoassays. Indications for the endarterectomy were: symptomatic carotid plaque with stenosis >70% or without symptoms and >80% stenosis. Plaque mRNA levels were assessed by RNA sequencing. Plaque components and extracellular matrix were measured histologically and biochemically. Matrix metalloproteinases and monocyte chemoattractant protein-1 (MCP-1) was measured with immunoassays. The effect of TGF-β2 on inflammation and protease activity was investigated in vitro using THP-1 and RAW264.7 macrophages. Patients were followed longitudinally for cardiovascular (CV) events.TGF-β2 was the most abundant isoform and was increased at both protein and mRNA levels in asymptomatic plaques. TGF-β2 was the main determinant separating asymptomatic plaques in an Orthogonal Projections to Latent Structures Discriminant Analysis. TGF-β2 correlated positively to features of plaque stability and inversely to markers of plaque vulnerability. TGF-β2 was the only isoform inversely correlated to the matrix-degrading matrix metalloproteinase-9 and inflammation in the plaque tissue. In vitro, TGF-β2 pre-treatment reduced MCP-1 gene and protein levels as well as matrix metalloproteinase-9 gene levels and activity. Patients with plaques with high TGF-β2 levels had a lower risk to suffer from future CV events. CONCLUSIONS TGF-β2 is the most abundant TGF-β isoform in human plaques and may maintain plaque stability by decreasing inflammation and matrix degradation.
Collapse
Affiliation(s)
- Andreas Edsfeldt
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Pratibha Singh
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Frank Matthes
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | | | - Michele Cavalera
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms—Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Pontus Dunér
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Petr Volkov
- Department of Clinical Sciences, LUDC Bioinformatics Unit, Malmö, Lund University, Lund, Sweden
- Data Science and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Glykeria Karadimou
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Anton Gisterå
- Department of Medicine, Center for Molecular Medicine, Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Nilsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Jiangming Sun
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Isabel Gonçalves
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
3
|
Affiliation(s)
- Lingfeng Luo
- Division of Vascular Surgery, Department of Surgery (L.L., N.J.L.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute, CA (L.L., N.J.L.)
| | - Nicholas J Leeper
- Division of Vascular Surgery, Department of Surgery (L.L., N.J.L.), Stanford University School of Medicine, CA
- Division of Cardiovascular Medicine, Department of Medicine (N.J.L.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute, CA (L.L., N.J.L.)
| |
Collapse
|
4
|
Deng Y, Jiang S, Lin X, Wang B, Chen B, Tong J, Shi W, Yu B, Tang J. Differential expression profile of miRNAs between stable and vulnerable plaques of carotid artery stenosis patients. Genes Genet Syst 2023. [PMID: 37121730 DOI: 10.1266/ggs.22-00123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Plaque vulnerability is associated with the degree of carotid artery stenosis (CS) and the risk of stroke. MicroRNAs (miRNAs) exert critical functions in disease progression, although only a few miRNAs have been well identified in CS. Therefore, this study aimed to investigate the differential expression profile of miRNAs and their potential functions in plaques of CS patients. Three CS patients with stable plaques and three patients with vulnerable plaques who underwent carotid endarterectomy were enrolled in this study. Differentially expressed miRNAs (DEmiRNAs) between patients with stable and vulnerable plaques were determined using small RNA sequencing. Target genes of DEmiRNAs were predicted and submitted to functional analyses. Validation of dysregulated DEmiRNAs was determined using quantitative real-time polymerase chain reaction (qRT-PCR). After sequencing, 76 DEmiRNAs were identified in vulnerable plaques, including 53 upregulated miRNAs and 23 downregulated miRNAs. Next, 23,495 target genes of the identified DEmiRNAs were predicted and functionally analyzed. This indicated that the target genes of the identified DEmiRNAs were mainly enriched in protein phosphorylation, transcription, nitrogen compound metabolism, endocytosis and autophagy, and related to signaling pathways of Hippo, MAPK, insulin, TGF-β, FoxO, AMPK and p53. Furthermore, qRT-PCR results for six miRNAs showed that five (83%) of them (hsa-miR-511-5p, hsa-miR-150-5p, hsa-miR-378a-5p, hsa-miR-365b-5p and hsa-miR-6511b-5p) were consistent with the sequencing results. Differential expression profiles and potential function of miRNAs associated with plaque stability in CS patients are identified for the first time, which should help to understand the regulatory mechanism of plaque stability in CS.
Collapse
Affiliation(s)
- Ying Deng
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Shuai Jiang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Xueguang Lin
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Bo Wang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Bo Chen
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Jindong Tong
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Weijun Shi
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Bo Yu
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Jingdong Tang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| |
Collapse
|
5
|
Ahmadi J, Hosseini E, Kargar F, Ghasemzadeh M. Stable CAD patients show higher levels of platelet-borne TGF-β1 associated with a superior pro-inflammatory state than the pro-aggregatory status; Evidence highlighting the importance of platelet-derived TGF-β1 in atherosclerosis. J Thromb Thrombolysis 2023; 55:102-115. [PMID: 36352058 DOI: 10.1007/s11239-022-02729-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2022] [Indexed: 11/10/2022]
Abstract
Activated platelets are involved in the atherogenic stage of atherosclerosis, while they can also progress it to atherothrombosis which may cause an ischemic state and organ failure. In general, coronary artery disease (CAD) is considered as common and severe clinical consequence of atherosclerosis, manifesting as a chronic inflammatory condition with the release of platelet mediators, among which the importance of platelet-borne TGF-β1 is not yet well understood. Hence, for the first time, this study aimed to examine platelet level of TGF-β1 (latent/mature) in CAD-patients and its association with the expression of platelet pro-inflammatory molecules. Platelet from stable CAD-patients candidate for CABG and healthy controls were subjected to flowcytometry analysis to evaluate P-selectin and CD40L expressions and PAC-1 binding. Platelet-borne and soluble TGF-β1, both mature/active and latent forms were also examined with western blotting. Higher expression levels of P-selectin and CD40L in patients with CAD than in controls were associated with comparable levels of PAC-1 binding in both groups. Platelet TGF-β1 levels were also significantly higher in patients, while their platelets showed clear bands of mature TGF-β1 that were barely visible in healthy individuals. Soluble TGF-β1 was also higher in patients. Significant correlations between mature/active TGF-β1 and platelet pro-inflammatory markers (P-selectin and CD40L) as well as common indicators of inflammation (CRP and ESR) were observed in CAD patients. In this study, given the insignificant changes in pro-aggregatory potentials in stable CAD, the pro-inflammatory state of platelets may be more involved in disease development and progression. Direct correlations between active platelet-borne TGF-β1 and pro-inflammatory markers with its presence in CAD-patients, which was almost absent in the platelets of healthy individuals, may also underscore the significant contribution of platelet-borne TGF-β1 to the pathogenesis of the disease.
Collapse
Affiliation(s)
- Javad Ahmadi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ehteramolsadat Hosseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Faranak Kargar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Mehran Ghasemzadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran. .,Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Next to the Milad Tower, Hemmat Exp. Way, P.O.Box:14665-1157, Tehran, Iran.
| |
Collapse
|
6
|
Alajbegovic A, Daoud F, Ali N, Kawka K, Holmberg J, Albinsson S. Transcription factor GATA6 promotes migration of human coronary artery smooth muscle cells in vitro. Front Physiol 2022; 13:1054819. [PMID: 36523548 PMCID: PMC9744938 DOI: 10.3389/fphys.2022.1054819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
Vascular smooth muscle cell plasticity plays a pivotal role in the pathophysiology of vascular diseases. Despite compelling evidence demonstrating the importance of transcription factor GATA6 in vascular smooth muscle, the functional role of GATA6 remains poorly understood. The aim of this study was to elucidate the role of GATA6 on cell migration and to gain insight into GATA6-sensitive genes in smooth muscle. We found that overexpression of GATA6 promotes migration of human coronary artery smooth muscle cells in vitro, and that silencing of GATA6 in smooth muscle cells resulted in reduced cellular motility. Furthermore, a complete microarray screen of GATA6-sensitive gene transcription resulted in 739 upregulated and 248 downregulated genes. Pathways enrichment analysis showed involvement of transforming growth factor beta (TGF-β) signaling which was validated by measuring mRNA expression level of several members. Furthermore, master regulators prediction based on microarray data revealed several members of (mitogen activated protein kinase) MAPK pathway as a master regulators, reflecting involvement of MAPK pathway also. Our findings provide further insights into the functional role of GATA6 in vascular smooth muscle and suggest that targeting GATA6 may constitute as a new approach to inhibit vascular smooth muscle migration.
Collapse
Affiliation(s)
- Azra Alajbegovic
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Fatima Daoud
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Neserin Ali
- Department of Clinical Sciences Lund, Orthopedics, Clinical Epidemiology Unit, Lund University, Lund, Sweden
| | - Katarzyna Kawka
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Johan Holmberg
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | |
Collapse
|
7
|
Bachmann JC, Baumgart SJ, Uryga AK, Bosteen MH, Borghetti G, Nyberg M, Herum KM. Fibrotic Signaling in Cardiac Fibroblasts and Vascular Smooth Muscle Cells: The Dual Roles of Fibrosis in HFpEF and CAD. Cells 2022; 11:1657. [PMID: 35626694 PMCID: PMC9139546 DOI: 10.3390/cells11101657] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022] Open
Abstract
Patients with heart failure with preserved ejection fraction (HFpEF) and atherosclerosis-driven coronary artery disease (CAD) will have ongoing fibrotic remodeling both in the myocardium and in atherosclerotic plaques. However, the functional consequences of fibrosis differ for each location. Thus, cardiac fibrosis leads to myocardial stiffening, thereby compromising cardiac function, while fibrotic remodeling stabilizes the atherosclerotic plaque, thereby reducing the risk of plaque rupture. Although there are currently no drugs targeting cardiac fibrosis, it is a field under intense investigation, and future drugs must take these considerations into account. To explore similarities and differences of fibrotic remodeling at these two locations of the heart, we review the signaling pathways that are activated in the main extracellular matrix (ECM)-producing cells, namely human cardiac fibroblasts (CFs) and vascular smooth muscle cells (VSMCs). Although these signaling pathways are highly overlapping and context-dependent, effects on ECM remodeling mainly act through two core signaling cascades: TGF-β and Angiotensin II. We complete this by summarizing the knowledge gained from clinical trials targeting these two central fibrotic pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kate M. Herum
- Research and Early Development, Novo Nordisk A/S, Novo Nordisk Park, 2760 Maaloev, Denmark; (J.C.B.); (S.J.B.); (A.K.U.); (M.H.B.); (G.B.); (M.N.)
| |
Collapse
|
8
|
Yurdagul A. Crosstalk Between Macrophages and Vascular Smooth Muscle Cells in Atherosclerotic Plaque Stability. Arterioscler Thromb Vasc Biol 2022; 42:372-380. [PMID: 35172605 PMCID: PMC8957544 DOI: 10.1161/atvbaha.121.316233] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Most acute cardiovascular events are due to plaque rupture, with atheromas containing large necrotic cores and thin fibrous caps being more susceptible to rupture and lesions with small necrotic cores and thick fibrous caps being more protected from rupture. Atherosclerotic plaques are comprised various extracellular matrix proteins, modified lipoprotein particles, and cells of different origins, that is, vascular cells and leukocytes. Although much has been revealed about the mechanisms that lead to plaque instability, several key areas remain incompletely understood. This In-Focus Review highlights processes related to cellular crosstalk and the role of the tissue microenvironment in determining cell function and plaque stability. Recent advances highlight critical underpinnings of atherosclerotic plaque vulnerability, particularly impairments in the ability of macrophages to clear dead cells and phenotypic switching of vascular smooth muscle cells. However, these processes do not occur in isolation, as crosstalk between macrophages and vascular smooth muscle cells and interactions with their surrounding microenvironment play a significant role in determining plaque stability. Understanding these aspects of cellular crosstalk within an atherosclerotic plaque may shed light on how to modify cell behavior and identify novel approaches to transform rupture-prone atheromas into stable lesions.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences, Shreveport
| |
Collapse
|
9
|
Ma J, Tan Q, Nie X, Zhou M, Wang B, Wang X, Cheng M, Ye Z, Xie Y, Wang D, Chen W. Longitudinal relationships between polycyclic aromatic hydrocarbons exposure and heart rate variability: Exploring the role of transforming growth factor-β in a general Chinese population. JOURNAL OF HAZARDOUS MATERIALS 2022; 425:127770. [PMID: 34823955 DOI: 10.1016/j.jhazmat.2021.127770] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/16/2021] [Accepted: 11/09/2021] [Indexed: 06/13/2023]
Abstract
We aim to investigate the long-term adverse effects of polycyclic aromatic hydrocarbons (PAHs) exposure on heart rate variability (HRV) reduction, and to assess the potential role of transforming growth factor-β1 (TGF-β1) in such relationship. We enrolled 2985 adult residents with 4100 observations who participated at baseline and 6-years follow-up from Wuhan-Zhuhai cohort. Ten detectable urinary PAHs metabolites and two HRV indices were repeatedly measured at baseline and follow-up; and plasma TGF-β1 levels were also determined for all subjects. We observed that both total urinary low molecular weight PAHs (ΣLWM OH-PAH) and total urinary high molecular weight PAHs (ΣHWM OH-PAH) were negatively associated with HRV reductions (P < 0.05). Subjects with persistent high levels of ΣHWM OH-PAH had a significant reduction in HRV over 6 years, and the incensement of TGF-β1 could aggravate above adverse effects in a dose-response manner. All kinds of PAHs were positively associated with plasma TGF-β1 elevation, which in turn, were negatively related to HRV indices. Increased TGF-β1 significant mediated 1.34-3.62% of PAHs-associated HRV reduction. Our findings demonstrated that long-term high levels of PAHs exposure could cause HRV reductions, and TGF-β1 may play an essential role in such association.
Collapse
Affiliation(s)
- Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Qiyou Tan
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiuquan Nie
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Min Zhou
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bin Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xing Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Man Cheng
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zi Ye
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yujia Xie
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Dongming Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weihong Chen
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
10
|
You M, Liu Y, Wang B, Li L, Zhang H, He H, Zhou Q, Cao T, Wang L, Zhao Z, Zhu Z, Gao P, Yan Z. Asprosin induces vascular endothelial-to-mesenchymal transition in diabetic lower extremity peripheral artery disease. Cardiovasc Diabetol 2022; 21:25. [PMID: 35168605 PMCID: PMC8848671 DOI: 10.1186/s12933-022-01457-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/25/2022] [Indexed: 12/03/2022] Open
Abstract
Background Altered adipokine secretion in dysfunctional adipose tissue facilitates the development of atherosclerotic diseases including lower extremity peripheral artery disease (PAD). Asprosin is a recently identified adipokine and displays potent regulatory role in metabolism, but the relationship between asprosin and lower extremity PAD remains uninvestigated. Methods 33 type 2 diabetes mellitus (T2DM) patients (DM), 51 T2DM patients with PAD (DM + PAD) and 30 healthy normal control (NC) volunteers were recruited and the blood samples were collected for detecting the circulatory asprosin level and metabolomic screening. RNA sequencing was performed using the aorta tissues from the type 2 diabetic db/db mice and human umbilical vein endothelial cells (HUVECs) were treated with asprosin to determine its impact on the endothelial-to-mesenchymal transition (EndMT). Results The circulating levels of asprosin in DM + PAD group were significantly higher than that of NC group and the DM group. Circulating asprosin level was remarkably negatively correlated with ankle-brachial index (ABI), even after adjusting for age, sex, body mass index (BMI) and other traditional risk factors of PAD. Logistic regression analysis revealed that asprosin is an independent risk factor for PAD and receiver-operator characteristic (ROC) curve determined a good sensitivity (74.5%) and specificity (74.6%) of asprosin to distinguish PAD. Data from metabolomics displayed a typical characteristics of de novo amino acid synthesis in collagen protein production by myofibroblasts in patients with PAD and activation of TGF-β signaling pathway appeared in the aortic tissue of db/db mice. Asprosin directly induces EndMT in HUVECs in a TGF-β-dependent manner as TGF-β signaling pathway inhibitor SB431542 erased the promotional effect of asprosin on EndMT. Conclusions Elevated circulatory asprosin level is an independent risk factor of lower extremity PAD and might serve as a diagnostic marker. Mechanistically, asprosin directly induces EndMT that participates in vascular injury via activation of TGF-β signaling pathway. Trial registration This trial was registered at clinicaltrials.gov as NCT05068895 Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01457-0.
Collapse
Affiliation(s)
- Mei You
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Yushuang Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Bowen Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Li Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Hexuan Zhang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Hongbo He
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Qing Zhou
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Tingbing Cao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Lijuan Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Zhigang Zhao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China.
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China.
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, 10 Chang Jiang Zhi Lu, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
11
|
Rostami Rayeni N, Abdollahzad H, Alibakhshi P, Morvaridzadeh M, Heydari H, Dehnad A, Khorshidi M, Izadi A, Shidfar F, Dulce Estêvão M, Omidi A, Heshmati J. Effects of body weight regain on leptin levels: A systematic review and meta-analysis. Cytokine 2021; 148:155647. [PMID: 34344588 DOI: 10.1016/j.cyto.2021.155647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND There are different changes observed before and after diet therapy, and also after weight regain. However, there is not sufficient information regarding weight regain and hormonal changes. PURPOSE The purpose of this study was to review the connection between weight regain and leptin concentration levels. METHODS MEDLINE, SCOPUS, Web of Science, and the Cochrane Library were searched for interventional articles published from January 1, 1980, to June 30, 2020. Randomized clinical trials with parallel or cross over design assessing leptin concentrations at the baseline and at the end of study were reviewed. Two independent reviewers extracted data related to study design, year of publication, country, age, gender, body mass index (BMI), duration of the following up period and mean ± SD of other intended variables. RESULTS Four articles were included, published between 2004 and 2016. Three of them were conducted in the US and one of them in Netherland. Sample size of the studies ranged between 25 and 148 participants. The range of following up period was from13 to 48 weeks. The age range of participants was from 34 to 44 years. Our analysis shows that weight regain could reduce leptin levels, but this change is not statistically significant. CONCLUSION This review suggests that weight regain may induce a non-significant reduction in leptin level. However, the limited number and great heterogeneity between the included studies may affect the presented results and there are still need to well-designed, large population studies to determine the relationship between weight regain and leptin levels.
Collapse
Affiliation(s)
- Najme Rostami Rayeni
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Hadi Abdollahzad
- Department of Nutritional Science, School of Nutritional Science and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pooya Alibakhshi
- Department of Internal Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Morvaridzadeh
- Songhor Healthcare Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hafez Heydari
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Afsaneh Dehnad
- Department of English Language, School of Health Management and Information Sciences, Center for Educational Research in Medical Sciences (CERMS), Iran University of Medical Sciences, Iran.
| | - Masoud Khorshidi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran; Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azimeh Izadi
- Iran Football Medical Assessment and Rehabilitation Center (IFMARC), Tehran, Iran; Department of Biochemistry and Diet Therapy, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| | - M Dulce Estêvão
- Universidade do Algarve, Escola Superior de Saúde, Campus de Gambelas, Faro, Portugal.
| | - Amirhosein Omidi
- Songhor Healthcare Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javad Heshmati
- Songhor Healthcare Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
12
|
Holm Nielsen S, Jonasson L, Kalogeropoulos K, Karsdal MA, Reese-Petersen AL, Auf dem Keller U, Genovese F, Nilsson J, Goncalves I. Exploring the role of extracellular matrix proteins to develop biomarkers of plaque vulnerability and outcome. J Intern Med 2020; 287:493-513. [PMID: 32012358 DOI: 10.1111/joim.13034] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/04/2019] [Accepted: 01/13/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease (CVD) is the most common cause of death in industrialized countries. One underlying cause is atherosclerosis, which is a systemic disease characterized by plaques of retained lipids, inflammatory cells, apoptotic cells, calcium and extracellular matrix (ECM) proteins in the arterial wall. The biologic composition of an atherosclerotic plaque determines whether the plaque is more or less vulnerable, that is prone to rupture or erosion. Here, the ECM and tissue repair play an important role in plaque stability, vulnerability and progression. This review will focus on ECM remodelling in atherosclerotic plaques, with focus on how ECM biomarkers might predict plaque vulnerability and outcome.
Collapse
Affiliation(s)
- S Holm Nielsen
- From the, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - L Jonasson
- Department of Medical and Health Sciences, Division of Cardiovascular Medicine, Linköping University, Linköping, Sweden
| | - K Kalogeropoulos
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - M A Karsdal
- From the, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark
| | | | - U Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - F Genovese
- From the, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark
| | - J Nilsson
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - I Goncalves
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
13
|
Bashir AZ, Bashir K, Hunter WJ, Agrawal DK. Cathepsin L expression in the carotid arteries of atherosclerotic swine. Arch Med Sci Atheroscler Dis 2019; 4:e264-e267. [PMID: 32373754 PMCID: PMC7197027 DOI: 10.5114/amsad.2019.90153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/05/2019] [Indexed: 11/17/2022] Open
Affiliation(s)
- Ayisha Zaka Bashir
- Department of Clinical Translational Sciences, School of Medicine, Creighton University, Omaha. Nebraska, USA
| | - Khalid Bashir
- Department of Clinical Translational Sciences, School of Medicine, Creighton University, Omaha. Nebraska, USA
| | - William J. Hunter
- Department of Clinical Translational Sciences, School of Medicine, Creighton University, Omaha. Nebraska, USA
| | - Devendra K. Agrawal
- Department of Translational Research, Western University of Health Sciences, California, USA
| |
Collapse
|
14
|
Affiliation(s)
- Kathryn L Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada.
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
15
|
Scicchitano P, Cortese F, Gesualdo M, De Palo M, Massari F, Giordano P, Ciccone MM. The role of endothelial dysfunction and oxidative stress in cerebrovascular diseases. Free Radic Res 2019; 53:579-595. [PMID: 31106620 DOI: 10.1080/10715762.2019.1620939] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Pietro Scicchitano
- Department of Cardiology, Hospital “F. Perinei”, Altamura, Italy
- Section of Cardiovascular Diseases, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | - Francesca Cortese
- Section of Cardiovascular Diseases, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| | | | - Micaela De Palo
- Department of Cardiac Surgery, Mater Dei Hospital, Bari, Italy
| | | | - Paola Giordano
- Department of Biomedical Sciences and Human Oncology – Paediatric Unit, Policlinico Hospital, Bari, Italy
| | - Marco Matteo Ciccone
- Section of Cardiovascular Diseases, Department of Emergency and Organ Transplantation, School of Medicine, University of Bari, Bari, Italy
| |
Collapse
|
16
|
Shi Y, Zhang J, Xu W, Yi J, Li Y, Chen Y. The correlation of TGFβ1 gene polymorphisms with congenital heart disease susceptibility. Gene 2018; 686:160-163. [PMID: 30321660 DOI: 10.1016/j.gene.2018.10.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/30/2018] [Accepted: 10/11/2018] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the relationship between single nucleotide polymorphisms (SNPs) in transforming growth factor beta 1 (TGFβ1) gene (rs1982073 and rs1800471) and congenital heart disease (CHD) susceptibility. METHODS Totally, 145 CHD patients and 140 healthy controls were enrolled in this case-control study. The case and control groups were matched in age and gender. Genotyping for TGFβ1 gene SNPs rs1982073 and rs1800471 was conducted via polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) approach. Chi-square test was used to analyze the association of TGFβ1 polymorphisms with CHD risk. The results were presented as odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS The frequencies of the CT genotype and T allele of TGFβ1 gene SNP rs1982073 were significantly different between cases and controls (P < 0.05), revealing their associations with reduced susceptibility to CHD (OR = 0.521, 95%CI = 0.302-0.897; OR = 0.706, 95%CI = 0.507-0.983). Nevertheless, TGFβ1 gene SNP rs1800471 had no significant association with CHD susceptibility (P > 0.05). CONCLUSION TGFβ1 gene SNP rs1982073 might be correlated with CHD susceptibility, and the T allele might decrease the disease risk. However, TGFβ1 gene polymorphism rs1800471 was not related to CHD risk.
Collapse
Affiliation(s)
- Yujie Shi
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Jian Zhang
- Department of Cardiology, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Wei Xu
- Cardiovascular Disease Institute, PLA Army General Hospital, Beijing 100853, China
| | - Jun Yi
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yang Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
17
|
Augstein A, Mierke J, Poitz DM, Strasser RH. Sox9 is increased in arterial plaque and stenosis, associated with synthetic phenotype of vascular smooth muscle cells and causes alterations in extracellular matrix and calcification. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2526-2537. [PMID: 29777903 DOI: 10.1016/j.bbadis.2018.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/23/2018] [Accepted: 05/15/2018] [Indexed: 12/20/2022]
Abstract
Vascular smooth muscle cells (VSMC) exhibit a dual role in progression and maintenance of arteriosclerosis. They are fundamental for plaque stability but also can drive plaque progression. During pathogenic vascular remodeling, VSMC transdifferentiate into a phenotype with enhanced proliferation and migration. Moreover, they exert an increased capacity to generate extracellular matrix proteins. A special lineage of transdifferentiated VSMC expresses Sox9, a multi-functional transcription factor. The aim of the study was to examine the role of Sox9 in phenotypic alterations leading to arteriosclerosis. Using mouse models for arterial stenosis, Sox9 induction in diseased vessels was verified. The phenotypic switch of VSMC from contractile to proliferative nature caused a significant increase of Sox9 expression. Various factors known to be involved in the progression of arteriosclerosis were examined for their ability to modulate Sox9 expression in VSMC. While PDGF-BB resulted in a strong transient upregulation of Sox9, TGF-β1 appeared to be responsible for a moderate, but prolonged increase of Sox9 expression. Beside the regulation, functional studies focused on knockout and overexpression of Sox9. A Sox9-dependent alteration of extracellular matrix could be revealed and was associated with an upregulated calcium deposition. Taken together, Sox9 is identified as important factor of VSMC function by modulation the extracellular matrix composition and calcium deposition, which are important processes in plaque development.
Collapse
Affiliation(s)
- Antje Augstein
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany.
| | - Johannes Mierke
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| | - David M Poitz
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| | - Ruth H Strasser
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| |
Collapse
|
18
|
Muñoz R, Santamaría E, Rubio I, Ausín K, Ostolaza A, Labarga A, Roldán M, Zandio B, Mayor S, Bermejo R, Mendigaña M, Herrera M, Aymerich N, Olier J, Gállego J, Mendioroz M, Fernández-Irigoyen J. Mass Spectrometry-Based Proteomic Profiling of Thrombotic Material Obtained by Endovascular Thrombectomy in Patients with Ischemic Stroke. Int J Mol Sci 2018; 19:ijms19020498. [PMID: 29414888 PMCID: PMC5855720 DOI: 10.3390/ijms19020498] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 12/25/2022] Open
Abstract
Thrombotic material retrieved from acute ischemic stroke (AIS) patients represents a valuable source of biological information. In this study, we have developed a clinical proteomics workflow to characterize the protein cargo of thrombi derived from AIS patients. To analyze the thrombus proteome in a large-scale format, we developed a workflow that combines the isolation of thrombus by endovascular thrombectomy and peptide chromatographic fractionation coupled to mass-spectrometry. Using this workflow, we have characterized a specific proteomic expression profile derived from four AIS patients included in this study. Around 1600 protein species were unambiguously identified in the analyzed material. Functional bioinformatics analyses were performed, emphasizing a clustering of proteins with immunological functions as well as cardiopathy-related proteins with blood-cell dependent functions and peripheral vascular processes. In addition, we established a reference proteomic fingerprint of 341 proteins commonly detected in all patients. Protein interactome network of this subproteome revealed protein clusters involved in the interaction of fibronectin with 14-3-3 proteins, TGFβ signaling, and TCP complex network. Taken together, our data contributes to the repertoire of the human thrombus proteome, serving as a reference library to increase our knowledge about the molecular basis of thrombus derived from AIS patients, paving the way toward the establishment of a quantitative approach necessary to detect and characterize potential novel biomarkers in the stroke field.
Collapse
Affiliation(s)
- Roberto Muñoz
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Enrique Santamaría
- Clinical Neuroproteomics Laboratory, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
- Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Idoya Rubio
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Karina Ausín
- Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Aiora Ostolaza
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Alberto Labarga
- Bioinformatics Laboratory, Navarrabiomed-Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Miren Roldán
- Neuroepigenetics Laboratory, Navarrabiomed-Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Beatriz Zandio
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Sergio Mayor
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Rebeca Bermejo
- Department of Interventional Neuroradiology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Mónica Mendigaña
- Department of Interventional Neuroradiology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - María Herrera
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Nuria Aymerich
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Jorge Olier
- Department of Interventional Neuroradiology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Jaime Gállego
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
| | - Maite Mendioroz
- Department of Neurology, Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
- Neuroepigenetics Laboratory, Navarrabiomed-Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Laboratory, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
- Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, IDISNA, Navarra Institute for Health Research, Pamplona 31008, Spain.
| |
Collapse
|
19
|
Bentzon JF, Daemen M, Falk E, Garcia-Garcia HM, Herrmann J, Hoefer I, Jukema JW, Krams R, Kwak BR, Marx N, Naruszewicz M, Newby A, Pasterkamp G, Serruys PWJC, Waltenberger J, Weber C, Tokgözoglu L, Ylä-Herttuala S. Stabilisation of atherosclerotic plaques. Thromb Haemost 2017; 106:1-19. [DOI: 10.1160/th10-12-0784] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 04/29/2011] [Indexed: 01/04/2023]
Abstract
SummaryPlaque rupture and subsequent thrombotic occlusion of the coronary artery account for as many as three quarters of myocardial infarctions. The concept of plaque stabilisation emerged about 20 years ago to explain the discrepancy between the reduction of cardiovascular events in patients receiving lipid lowering therapy and the small decrease seen in angiographic evaluation of atherosclerosis. Since then, the concept of a vulnerable plaque has received a lot of attention in basic and clinical research leading to a better understanding of the pathophysiology of the vulnerable plaque and acute coronary syndromes. From pathological and clinical observations, plaques that have recently ruptured have thin fibrous caps, large lipid cores, exhibit outward remodelling and invasion by vasa vasorum. Ruptured plaques are also focally inflamed and this may be a common denominator of the other pathological features. Plaques with similar characteristics, but which have not yet ruptured, are believed to be vulnerable to rupture. Experimental studies strongly support the validity of anti-inflammatory approaches to promote plaque stability. Unfortunately, reliable non-invasive methods for imaging and detection of such plaques are not yet readily available. There is a strong biological basis and supportive clinical evidence that low-density lipoprotein lowering with statins is useful for the stabilisation of vulnerable plaques. There is also some clinical evidence for the usefulness of antiplatelet agents, beta blockers and renin-angiotensin-aldosterone system inhibitors for plaque stabilisation. Determining the causes of plaque rupture and designing diagnostics and interventions to prevent them are urgent priorities for current basic and clinical research in cardiovascular area.
Collapse
|
20
|
Salem M, Butt H, Choke E, Moore D, West K, Robinson T, Sayers R, Naylor A, Bown M. Gene and Protein Expression of Chemokine (C-C-Motif) Ligand 19 is Upregulated in Unstable Carotid Atherosclerotic Plaques. Eur J Vasc Endovasc Surg 2016; 52:427-436. [DOI: 10.1016/j.ejvs.2016.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/19/2016] [Indexed: 12/13/2022]
|
21
|
Immune-inflammatory responses in atherosclerosis: Role of an adaptive immunity mainly driven by T and B cells. Immunobiology 2016; 221:1014-33. [PMID: 27262513 DOI: 10.1016/j.imbio.2016.05.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/06/2016] [Accepted: 05/23/2016] [Indexed: 01/22/2023]
Abstract
Adaptive immune response plays an important role in atherogenesis. In atherosclerosis, the proinflammatory immune response driven by Th1 is predominant but the anti-inflammatory response mediated mainly by regulatory T cells is also present. The role of Th2 and Th17 cells in atherogenesis is still debated. In the plaque, other T helper cells can be observed such as Th9 and Th22 but is little is known about their impact in atherosclerosis. Heterogeneity of CD4(+) T cell subsets presented in the plaque may suggest for plasticity of T cell that can switch the phenotype dependening on the local microenvironment and activating/blocking stimuli. Effector T cells are able to recognize self-antigens released by necrotic and apoptotic vascular cells and induce a humoral immune reaction. Tth cells resided in the germinal centers help B cells to switch the antibody class to the production of high-affinity antibodies. Humoral immunity is mediated by B cells that release antigen-specific antibodies. A variety of B cell subsets were found in human and murine atherosclerotic plaques. In mice, B1 cells could spontaneously produce atheroprotective natural IgM antibodies. Conventional B2 lymphocytes secrete either proatherogenic IgG, IgA, and IgE or atheroprotective IgG and IgM antibodies reactive with oxidation-specific epitopes on atherosclerosis-associated antigens. A small population of innate response activator (IRA) B cells, which is phenotypically intermediate between B1 and B2 cells, produces IgM but possesses proatherosclerotic properties. Finally, there is a minor subset of splenic regulatory B cells (Bregs) that protect against atherosclerotic inflammation through support of generation of Tregs and production of anti-inflammatory cytokines IL-10 and TGF-β and proapoptotic molecules.
Collapse
|
22
|
Rai V, Rao VH, Shao Z, Agrawal DK. Dendritic Cells Expressing Triggering Receptor Expressed on Myeloid Cells-1 Correlate with Plaque Stability in Symptomatic and Asymptomatic Patients with Carotid Stenosis. PLoS One 2016; 11:e0154802. [PMID: 27148736 PMCID: PMC4858252 DOI: 10.1371/journal.pone.0154802] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/19/2016] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease with atherosclerotic plaques containing inflammatory cells, including T-lymphocytes, dendritic cells (DCs) and macrophages that are responsible for progression and destabilization of atherosclerotic plaques. Stressed cells undergoing necrosis release molecules that act as endogenous danger signals to alert and activate innate immune cells. In atherosclerotic tissue the number of DCs increases with the progression of the lesion and produce several inflammatory cytokines and growth factors. Triggering receptor expressed on myeloid cells (TREM)-1 plays a crucial role in inflammation. However, relationship of DCs and the role of TREM-1 with the stability of atherosclerotic plaques have not been examined. In this study, we investigated the heterogeneity of the plaque DCs, myeloid (mDC1 and mDC2) and plasmacytoid (pDCs), and examined the expression of TREM-1 and their co-localization with DCs in the plaques from symptomatic (S) and asymptomatic (AS) patients with carotid stenosis. We found increased expression of HLA-DR, fascin, and TREM-1 and decreased expression of TREM-2 and α-smooth muscle actin in S compared to AS atherosclerotic carotid plaques. Both TREM-1 and fascin were co-localized suggesting increased expression of TREM-1 in plaque DCs of S compared to AS patients. These data were supported by increased mRNA transcripts of TREM-1 and decreased mRNA transcripts of TREM-2 in carotid plaques of S compared to AS patients. There was higher density of both CD1c+ mDC1 and CD141+ mDC2 in the carotid plaques from AS compared to S patients, where as the density of CD303+ pDCs were higher in the carotid plaques of S compared to AS patients. These findings suggest a potential role of pDCs and TREM-1 in atherosclerotic plaque vulnerability. Thus, newer therapies could be developed to selectively block TREM-1 for stabilizing atherosclerotic plaques.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska 68178, United States of America
| | - Velidi H. Rao
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska 68178, United States of America
| | - Zhifei Shao
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska 68178, United States of America
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska 68178, United States of America
- * E-mail:
| |
Collapse
|
23
|
Association of transforming growth factor-β1 gene C509T, G800A and T869C polymorphisms with intracerebral hemorrhage in North Indian Population: a case-control study. Neurol Sci 2015; 37:353-9. [PMID: 26621360 DOI: 10.1007/s10072-015-2426-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/18/2015] [Indexed: 10/22/2022]
Abstract
Transforming growth factor-β1 (TGF-β1) is a multifunctional pro-inflammatory cytokine involved in inflammation and pathogenesis of cerebrovascular disease. As per our knowledge, there is no published study investigating the association between variations within the TGF-β1 gene polymorphisms and risk of intracerebral hemorrhage (ICH). The aim of this study was to investigate the association of the TGF-β1 gene (C509T, G800A and T869C) polymorphisms, and their haplotypes with the risk of ICH in North Indian population. 100 ICH patients and 100 age- and sex-matched controls were studied. Genotyping was performed using SNaPshot method. Conditional logistic regression analysis was used to calculate the strength of association between TGF-β1 gene polymorphisms and risk of ICH. Hypertension, diabetes, dyslipidemia, low socioeconomic status, smoking, physical activity were found to be associated with the risk of ICH. The distribution of C509T, G800A and T869C genotypes was consistent with Hardy-Weinberg Equilibrium (HWE) in the ICH and control group. Adjusted conditional logistic regression analysis showed an independent association of TGF-β1 G800A (OR 9.07; 95% CI 2.3-35.6; P = 0.002) and T869C (OR 5.1; 95 % CI 1.9-13.2; P = 0.001) with the risk of ICH under dominant model. Haplotype analysis showed that C509-G800-C869 and C509-A800-C869 haplotypes were significantly associated with the increased risk of ICH. C509T and T869C were in strong linkage disequilibrium (D' = 0.53, r(2) = 0.23). Our results suggest that TGF-β1 (G800A, T869C) gene polymorphisms and their haplotypes are significantly associated with the risk of ICH in North Indian population. Further prospective studies with large sample size are required for independent validation. Our findings could be helpful in identifying individuals at increased risk for developing ICH.
Collapse
|
24
|
Serum Galectin-9 Levels Are Associated with Coronary Artery Disease in Chinese Individuals. Mediators Inflamm 2015; 2015:457167. [PMID: 26663989 PMCID: PMC4667018 DOI: 10.1155/2015/457167] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 09/10/2015] [Accepted: 09/13/2015] [Indexed: 01/12/2023] Open
Abstract
Background. Recently, several studies suggest that galectin-9 (Gal-9) might play a pivotal role in the pathogenesis of autoimmune diseases. However, the exact role of Gal-9 in atherosclerosis remains to be elucidated. Methods. Serum Gal-9, high-sensitivity C-reactive protein (hs-CRP), interferon- (IFN-) γ, interleukin- (IL-) 4, IL-17, and transforming growth factor- (TGF-) β1 were measured. The effect of Gal-9 on peripheral blood mononuclear cells (PBMC) was investigated in patients with normal coronary artery (NCA). Results. The lowest level of Gal-9 was found in the ST-segment elevation myocardial infarction (STEMI) group, followed by the non-ST-segment elevation ACS (NSTEACS), the NCA, and the stable angina pectoris (SAP) groups, respectively. Additionally, Gal-9 was found to be independently associated with hs-CRP, lipoprotein(a), and creatinine. Notably, Gal-9 was also noted to be an independent predictor of the Gensini score. Moreover, Gal-9 suppressed T-helper 17 (Th17) and expanded regulatory T cells (Tregs), resulting in decreased IL-17 production and increased secretion of TGF-β1. Conclusions. Serum Gal-9 is associated with not only coronary artery disease (CAD), but also the severity of coronary arteries stenosis. Gal-9 can expand Tregs and suppress Th17 development in activated PBMC, implying that Gal-9 has the potential to dampen the development of atherosclerosis and may be a new therapy for CAD.
Collapse
|
25
|
Curtis A, Smith T, Ziganshin BA, Elefteriades JA. Ascending Aortic Proaneurysmal Genetic Mutations with Antiatherogenic Effects. Int J Angiol 2015; 24:189-97. [PMID: 26417187 DOI: 10.1055/s-0035-1556075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Thoracic aortic aneurysms are common and are associated with a high morbidity and mortality. Despite this lethal diagnosis, there is an increasing body of evidence to suggest that the diagnosis of an aneurysm, specifically in the ascending thoracic aorta, may significantly reduce the risk of developing systemic atherosclerosis. Clinical observations in the operating room have shown pristine blood vessels in patients undergoing surgery for thoracic aortic aneurysms. There is now evidence that both the carotid intima-media thickness and arterial calcification, which are early and late signs of atherosclerosis respectively, are decreased in those with thoracic aortic aneurysms. These clinical studies are supported by molecular, genetic, and pharmacological evidence. Two principle mechanisms have been identified to explain the relationship of a proaneurysmal state conferring protection from atherosclerosis. These include an excess proteolytic balance of matrix metalloproteinase activity, leading to fragmentation of elastic lamellae and disordered collagen deposition. In addition, transforming growth factor β modulates vascular smooth muscle cells, extracellular matrix, and leukocytes. This confers protection from the initial plaque formation and, later provides stability to the plaque possibly through alteration of the types I and II transforming growth factor β receptor ratio. Furthermore, studies are now beginning to establish an important role for statins and estradiol in modulating these complex pathways. In the future, as our understanding of these complex mechanisms underlying aneurysmal protection against atherosclerosis increases, corresponding therapies may be developed to offer protection from atherosclerosis.
Collapse
Affiliation(s)
- Alexander Curtis
- Aortic Institute at Yale-New Haven, Yale University School of Medicine, New Haven, Connecticut
| | - Tanya Smith
- Aortic Institute at Yale-New Haven, Yale University School of Medicine, New Haven, Connecticut
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven, Yale University School of Medicine, New Haven, Connecticut ; Department of Surgical Diseases No. 2, Kazan State Medical University, Kazan, Russia
| | - John A Elefteriades
- Aortic Institute at Yale-New Haven, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
26
|
Talasaz AH, Khalili H, Jenab Y, Salarifar M, Broumand MA, Darabi F. N-Acetylcysteine effects on transforming growth factor-β and tumor necrosis factor-α serum levels as pro-fibrotic and inflammatory biomarkers in patients following ST-segment elevation myocardial infarction. Drugs R D 2014; 13:199-205. [PMID: 24048773 PMCID: PMC3784054 DOI: 10.1007/s40268-013-0025-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background and Aims Ischemia following acute myocardial infarction (AMI) increases the level of pro-fibrotic and inflammatory cytokines, including transforming growth factor (TGF)-β and tumor necrosis factor (TNF)-α. N-acetylcysteine (NAC) has therapeutic benefits in the management of patients with AMI. To the best of our knowledge, this is the first study that has evaluated the effect of NAC on TNF-α and TGF-β levels in patients with AMI. Methods Following confirmation of AMI, 88 patients were randomly administered NAC 600 mg (Fluimucil®, Zambon, Ticino, Switzerland) or placebo orally twice daily for 3 days. For quantification of TGF-β and TNF-α serum levels after 24 and 72 h of NAC or placebo administration, peripheral venous blood (10 mL) samples were collected at these time points. Results Comparisons between levels of TGF-β and TNF-α after 24 and 72 h within the NAC or placebo groups revealed that there was not any significant difference except for TGF-β levels in the placebo group, which increased significantly over time (p = 0.042). Significant relationships existed between patients’ ejection fraction (p = 0.005) and TGF-β levels. Conclusions Receiving NAC could prevent TGF-β levels from increasing after 72 h as compared with not receiving NAC. As TGF-β had strong correlations with the ejection fraction, its antagonism seems to be important in the prevention of remodeling.
Collapse
Affiliation(s)
- Azita Hajhossein Talasaz
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran,
| | | | | | | | | | | |
Collapse
|
27
|
Inflammatory biomarkers for predicting cardiovascular disease. Clin Biochem 2013; 46:1353-71. [PMID: 23756129 DOI: 10.1016/j.clinbiochem.2013.05.070] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 05/27/2013] [Accepted: 05/30/2013] [Indexed: 02/07/2023]
Abstract
The pathology of cardiovascular disease (CVD) is complex; multiple biological pathways have been implicated, including, but not limited to, inflammation and oxidative stress. Biomarkers of inflammation and oxidative stress may serve to help identify patients at risk for CVD, to monitor the efficacy of treatments, and to develop new pharmacological tools. However, due to the complexities of CVD pathogenesis there is no single biomarker available to estimate absolute risk of future cardiovascular events. Furthermore, not all biomarkers are equal; the functions of many biomarkers overlap, some offer better prognostic information than others, and some are better suited to identify/predict the pathogenesis of particular cardiovascular events. The identification of the most appropriate set of biomarkers can provide a detailed picture of the specific nature of the cardiovascular event. The following review provides an overview of existing and emerging inflammatory biomarkers, pro-inflammatory cytokines, anti-inflammatory cytokines, chemokines, oxidative stress biomarkers, and antioxidant biomarkers. The functions of each biomarker are discussed, and prognostic data are provided where available.
Collapse
|
28
|
Aldhoon B, Kučera T, Smorodinová N, Martínek J, Melenovský V, Kautzner J. Associations between cardiac fibrosis and permanent atrial fibrillation in advanced heart failure. Physiol Res 2013; 62:247-55. [PMID: 23489193 DOI: 10.33549/physiolres.932409] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Atrial fibrosis is considered as the basis in the development of long-standing atrial fibrillation (AF). However, in advanced heart failure (HF), the independent role of fibrosis for AF development is less clear since HF itself leads to atrial scarring. Our study aimed to differentiate patients with AF from patients without AF in a population consisting of patients with advanced HF. Myocardial samples from the right atrial and the left ventricular wall were obtained during heart transplantation from the explanted hearts of 21 male patients with advanced HF. Long-standing AF was present in 10 of them and the remaining 11 patients served as sinus rhythm controls. Echocardiographic and hemodynamic measurements were recorded prior to heart transplantation. Collagen volume fraction (CVF), transforming growth factor-beta (TGF-beta), and connective tissue growth factor (CTGF) expression in myocardial specimens were assessed histologically and immunohistochemically. The groups were well matched according to age (51.9+/-8.8 vs. 51.3+/-9.3 y) and co-morbidities. The AF group had higher blood pressure in the right atrium (13.6+/-7.7 vs. 6.0+/-5.0 mmHg; p=0.02), larger left atrium diameter (56.1+/-7.7 vs. 50+/-5.1 mm; p=0.043), higher left atrium wall stress (18.1+/-2.1 vs. 16.1+/-1.7 kdynes/m(2); p=0.04), and longer duration of HF (5.0+/-2.9 vs. 2.0+/-1.6 y, p=0.008). There were no significant differences in CVF (p=0.12), in CTGF (p=0.60), and in TGF-beta expression (p=0.66) in the atrial myocardium between the two study groups. In conclusions, in advanced HF, atrial fibrosis expressed by CVF is invariably present regardless of occurrence of AF. In addition to atrial wall fibrosis, increased wall stress might contribute to AF development in long-standing AF.
Collapse
Affiliation(s)
- B Aldhoon
- Department of Cardiology, Institute for Clinical and Experimental Medicine - IKEM, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
29
|
Yang DH, Hsu CF, Lin CY, Guo JY, Yu WC, Chang VH. Krüppel-like factor 10 upregulates the expression of cyclooxygenase 1 and further modulates angiogenesis in endothelial cell and platelet aggregation in gene-deficient mice. Int J Biochem Cell Biol 2013. [DOI: 10.1016/j.biocel.2012.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
30
|
Riccioni G, Sblendorio V. Atherosclerosis: from biology to pharmacological treatment. J Geriatr Cardiol 2012; 9:305-17. [PMID: 23097661 PMCID: PMC3470030 DOI: 10.3724/sp.j.1263.2012.02132] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/23/2012] [Accepted: 06/01/2012] [Indexed: 12/13/2022] Open
Abstract
A recent explosion in the amount of cardiovascular risk has swept across the globe. Primary prevention is the preferred method to lower cardiovascular risk. Lowering the prevalence of obesity is the most urgent matter, and is pleiotropic since it affects blood pressure, lipid profiles, glucose metabolism, inflammation, and atherothrombotic disease progression. Given the current obstacles, success of primary prevention remains uncertain. At the same time, the consequences of delay and inaction will inevitably be disastrous, and the sense of urgency mounts. Pathological and epidemiological data confirm that atherosclerosis begins in early childhood, and advances seamlessly and inexorably throughout life. Risk factors in childhood are similar to those in adults, and track between stages of life. When indicated, aggressive treatment should begin at the earliest indication, and be continued for many years. For those patients at intermediate risk according to global risk scores, C-reactive protein, coronary artery calcium, and carotid intima-media thickness are available for further stratification. Using statins for primary prevention is recommended by guidelines, is prevalent, but remains under prescribed. Statin drugs are unrivaled, evidence-based, major weapons to lower cardiovascular risk. Even when low density lipoprotein cholesterol targets are attained, over half of patients continue to have disease progression and clinical events. Though clinical evidence is incomplete, altering or raising the blood high density lipoprotein cholesterol level continues to be pursued. The aim of this review is to point out the attention of key aspects of vulnerable plaques regarding their pathogenesis and treatment.
Collapse
Affiliation(s)
- Graziano Riccioni
- Cardiology Unit, San Camillo de Lellis Hospital, Manfredonia, Via Isonzo 71043 Manfredonia (FG), Italy
| | - Valeriana Sblendorio
- Department of General Surgery and Surgical Specialties, University of Modena and Reggio Emilia Medical School, Surgical Clinic, Via Università, 41121 Modena, Italy
| |
Collapse
|
31
|
Rocha CDLJV, Rocha Júnior AM, Aarestrup BJV, Aarestrup FM. Inibição da expressão de ciclooxigenase 2 em feridas cutâneas de camundongos NOD submetidos à terapia a laser de baixa intensidade. J Vasc Bras 2012. [DOI: 10.1590/s1677-54492012000300002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
CONTEXTO: A terapia a laser de baixa intensidade (LLLT) tem sido relatada como importante moduladora da cicatrização de feridas cutâneas aumentando a proliferação fibroblástica associada ao aumento da expressão da citocina fator transformador de crescimento- β2 (TGF-βB2). OBJETIVO: No presente estudo foram avaliados os efeitos da LLLT sobre a expressão da enzima ciclooxigenase 2 (COX2) no sítio do reparo tecidual utilizando o modelo experimental com camundongos diabéticos não obesos (NOD) para estudar a cicatrização de feridas cutâneas. MÉTODOS: Foram utilizados 30 camundongos NOD, destes 14 ficaram diabéticos e foram divididos em dois grupos: o grupo I (n=7) foi submetido a um procedimento cirúrgico de feridas cutâneas e o grupo II (n=7) foi submetido a um procedimento cirúrgico de feridas cutâneas e tratados com LLLT. O grupo II foi submetido à LLLT nos seguintes parâmetros: 15 mW de potência, dose de 3,8 J/cm² e tempo de aplicação de 20 segundos. Após sete dias do ato cirúrgico e após aplicação do laser, os animais foram eutanasiados com sobredose de anestesia e amostras das feridas foram colhidas para posterior análise histopatológica, histomorfométrica e imuno-histoquímica. RESULTADOS: A LLLT promoveu a inibição da expressão da COX2 em feridas cutâneas de camundongos diabéticos. CONCLUSÃO: Em conjunto, os resultados sugeriram que a LLLT é capaz de modular negativamente a expressão da enzima COX2 contribuindo para o controle da resposta inflamatória em feridas cutâneas de camundongos NOD.
Collapse
|
32
|
Iso H, Maruyama K, Ikehara S, Yamagishi K, Tamakoshi A. Cellular growth factors in relation to mortality from cardiovascular disease in middle-aged Japanese: the JACC study. Atherosclerosis 2012; 224:154-60. [PMID: 22858286 DOI: 10.1016/j.atherosclerosis.2012.05.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 05/18/2012] [Accepted: 05/19/2012] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Limited evidence has been available on the relationships of cellular growth factors with cardiovascular disease in population-based samples. METHODS We conducted a nested case-control study under a large prospective cohort study (JACC study) where a total of 39,242 subjects aged 40-79 years provided serum sample. We measured cellular growth factors [insulin-like growth factors I, II and binding protein-3 (IGF-I, IGF-II and IGFBP-3) and transforming growth factor (TGF-β1)] among cases and controls, matched for sex, age, area of residence and year of serum storage. RESULTS AND CONCLUSIONS During the follow-up for 9 years, there were 233 deaths from total stroke (49 subarachnoid hemorrhages, 55 intraparenchymal hemorrhages, 71 ischemic strokes), and 97 deaths from coronary heart disease. The multivariable odds ratio (95%CI) of intraparenchymal hemorrhage associated with a 1-SD increment of IGF-I (men:4 8 ng/ml, women: 61 ng/ml) was 0.31 (0.14-0.71). That of ischemic stroke associated with a 1-SD increment of TGF-β1 (men: 8.0 ng/ml, women: 10.9 ng/ml) was 0.58 (0.34-0.98). Serum IGF-II and IGFBP-3 were not associated with mortality from any outcomes. In conclusion, IGF-I was inversely associated with mortality from intraparenchymal hemorrhage while TGF-β1 was so with ischemic stroke, suggesting potential roles of cellular proliferation in the development or prognosis of stroke.
Collapse
Affiliation(s)
- Hiroyasu Iso
- Public Health, Department of Social and Environmental Medicine, Osaka University Graduate School of Medicine, Japan.
| | | | | | | | | |
Collapse
|
33
|
Guo X, Chen SY. Transforming growth factor-β and smooth muscle differentiation. World J Biol Chem 2012; 3:41-52. [PMID: 22451850 PMCID: PMC3312200 DOI: 10.4331/wjbc.v3.i3.41] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 01/19/2012] [Accepted: 01/26/2012] [Indexed: 02/05/2023] Open
Abstract
Transforming growth factor (TGF)-β family members are multifunctional cytokines regulating diverse cellular functions such as growth, adhesion, migration, apoptosis, and differentiation. TGF-βs elicit their effects via specific type I and type II serine/threonine kinase receptors and intracellular Smad transcription factors. Knockout mouse models for the different components of the TGF-β signaling pathway have revealed their critical roles in smooth muscle cell (SMC) differentiation. Genetic studies in humans have linked mutations in these signaling components to specific cardiovascular disorders such as aorta aneurysm and congenital heart diseases due to SMC defects. In this review, the current understanding of TGF-β function in SMC differentiation is highlighted, and the role of TGF-β signaling in SMC-related diseases is discussed.
Collapse
Affiliation(s)
- Xia Guo
- Xia Guo, Shi-You Chen, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, United States
| | | |
Collapse
|
34
|
Najar RA, Ghaderian SMH, Panah AST. Association of transforming growth factor-β1 gene polymorphisms with genetic susceptibility to acute myocardial infarction. Am J Med Sci 2012; 342:365-70. [PMID: 21685787 DOI: 10.1097/maj.0b013e318215908a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Transforming growth factor beta 1 (TGF-β1) gene plays an important role in acute myocardial infarction (AMI); however, little is known about the relation of variations within the gene and risk of cardiovascular diseases. In this study, the authors evaluated the influence of TGF-β1 polymorphisms on the onset and progression of AMI in Iranian patients comparing with healthy individuals. METHODS Genomic DNA and peripheral blood mononuclear cells of 900 enrolled patients with AMI and 900 control subjects were extracted. The -509 C/T, 868T/C, 913G/C and 11929C/T TGF-β1 polymorphisms were detected. The messenger RNA (mRNA) expression and serum levels of TGF-β1 were analyzed by real-time reverse-transcriptase polymerase chain reaction and ELISA, respectively. RESULTS The frequency of "T" allele in -509 C/T, "C" allele in 868T/C, "C" allele in 913G/C and "T" allele in 11929C/T polymorphisms were significantly higher in the patients than control subjects (P < 0.001). There were significant differences in circulating levels of TGF-β1 in the patients than in control subjects (P < 0.001). These concentrations are associated with its gene polymorphism. The mRNA expression levels of TGF-β1 were significantly higher in the patient serums compared with controls (P < 0.001). CONCLUSIONS Our results confirmed the association between the TGF-β1 polymorphisms and risk of AMI, which suggest that genetic polymorphisms in TGF-β1 might be helpful for determining susceptibility to AMI in Iranian patients. There are also significant relationship between serum TGF-β1 and occurrence of AMI. In addition, susceptibility to AMI might be related to TGF-β1 gene expression, which affects its serum levels.
Collapse
Affiliation(s)
- Reza Akbarzadeh Najar
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences and Health Services, Tehran, Iran
| | | | | |
Collapse
|
35
|
Toma I, McCaffrey TA. Transforming growth factor-β and atherosclerosis: interwoven atherogenic and atheroprotective aspects. Cell Tissue Res 2012; 347:155-75. [PMID: 21626289 PMCID: PMC4915479 DOI: 10.1007/s00441-011-1189-3] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 05/06/2011] [Indexed: 12/15/2022]
Abstract
Age-related progression of cardiovascular disease is by far the largest health problem in the US and involves vascular damage, progressive vascular fibrosis and the accumulation of lipid-rich atherosclerotic lesions. Advanced lesions can restrict flow to key organs and can trigger occlusive thrombosis resulting in a stroke or myocardial infarction. Transforming growth factor-beta (TGF-β) is a major orchestrator of the fibroproliferative response to tissue damage. In the early stages of repair, TGF-β is released from platelets and activated from matrix reservoirs; it then stimulates the chemotaxis of repair cells, modulates immunity and inflammation and induces matrix production. At later stages, it negatively regulates fibrosis through its strong antiproliferative and apoptotic effects on fibrotic cells. In advanced lesions, TGF-β might be important in arterial calcification, commonly referred to as "hardening of the arteries". Because TGF-β can signal through multiple pathways, namely the SMADs, a MAPK pathway and the Rho/ROCK pathways, selective defects in TGF-β signaling can disrupt otherwise coordinated pathways of tissue regeneration. TGF-β is known to control cell proliferation, cell migration, matrix synthesis, wound contraction, calcification and the immune response, all being major components of the atherosclerotic process. However, many of the effects of TGF-β are essential to normal tissue repair and thus, TGF-β is often thought to be "atheroprotective". The present review attempts to parse systematically the known effects of TGF-β on both the major risk factors for atherosclerosis and to isolate the role of TGF-β in the many component pathways involved in atherogenesis.
Collapse
Affiliation(s)
- Ian Toma
- Department of Medicine, Division of Genomic Medicine, The George Washington University Medical Center, 2300 I Street NW. Ross Hall 443, Washington DC 20037, USA
| | - Timothy A. McCaffrey
- Department of Medicine, Division of Genomic Medicine, The George Washington University Medical Center, 2300 I Street NW. Ross Hall 443, Washington DC 20037, USA
| |
Collapse
|
36
|
Katakami N, Kaneto H, Osonoi T, Kawai K, Ishibashi F, Imamura K, Maegawa H, Kashiwagi A, Watada H, Kawamori R, Shimomura I, Yamasaki Y. Transforming growth factor β1 T868C gene polymorphism is associated with cerebral infarction in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract 2011; 94:e57-60. [PMID: 21885147 DOI: 10.1016/j.diabres.2011.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 08/01/2011] [Indexed: 11/30/2022]
Abstract
It is likely that the C allele of the polymorphism at position 29 of the translated sequence of transforming growth factor (TGF)-β1 gene, which codes a pleiotropic cytokine expressed in a variety of cells, is a susceptibility allele for cerebral infarction in Japanese type 2 diabetic patients.
Collapse
Affiliation(s)
- N Katakami
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ohyama Y, Tanaka T, Shimizu T, Matsui H, Sato H, Koitabashi N, Doi H, Iso T, Arai M, Kurabayashi M. Runx2/Smad3 complex negatively regulates TGF-β-induced connective tissue growth factor gene expression in vascular smooth muscle cells. J Atheroscler Thromb 2011; 19:23-35. [PMID: 21986102 DOI: 10.5551/jat.9753] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Connective tissue growth factor (CTGF), a direct target gene of transforming growth factor-β (TGF-β) signaling, plays an important role in the development of atherosclerosis. We previously showed that Runx2, a key transcription factor in osteoblast differentiation, regulates osteogenic conversion and dedifferentiation of vascular smooth muscle cells (VSMCs). In this study, we investigated the hypothesis that Runx2 modulates CTGF gene expression via the regulation of TGF-β signaling. METHODS AND RESULTS Expression of the Runx2 gene was decreased, and CTGF mRNA levels were reciprocally increased by TGF-β in a time-dependent manner in cultured human aortic smooth muscle cells (HASMCs) and C3H10T1/2 cells. Forced expression of Runx2 decreased and the reduction of Runx2 expression by small interfering RNA enhanced both basal and TGF-β-stimulated CTGF gene expression in HASMCs. Site-directed mutation analysis of the CTGF promoter indicated that transcriptional repression by Runx2 was mediated by the Smad-binding element (SBE) under basal and TGF-β-stimulated conditions. Data obtained from immunoblots of Runx2-, Smad3- or Smad4-transfected cells and chromatin immunoprecipitation analysis indicated that Runx2 interacts with Smad3 at the SBE. Immunohistochemistry revealed that the expression of Runx2 and CTGF was distinct and almost mutually exclusive in human atherosclerotic plaque. CONCLUSIONS These results for the first time demonstrate that Runx2/Smad3 complex negatively regulates endogenous and TGF-β-induced CTGF gene expression in VSMCs. Thus, the induction of Runx2 expression contributes to the phenotypic modulation of VSMCs, in which the TGF-β/Smad pathway plays a major role.
Collapse
Affiliation(s)
- Yoshiaki Ohyama
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Peng Z, Zhan L, Chen S, Xu E. Association of transforming growth factor-β1 gene C-509T and T869C polymorphisms with atherosclerotic cerebral infarction in the Chinese: a case-control study. Lipids Health Dis 2011; 10:100. [PMID: 21679448 PMCID: PMC3129580 DOI: 10.1186/1476-511x-10-100] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 06/16/2011] [Indexed: 12/12/2022] Open
Abstract
Background Transforming growth factor-β1 (TGF-β1) is a multifunctional cytokine involved in inflammation and pathogenesis of atherosclerosis. There is scant information on the relation between variations within the TGF-β1 gene polymorphisms and risks of ischemic cerebrovascular diseases. Therefore, this case-controlled study was carried out to investigate the possible association of the TGF-β1 gene C-509T and T869C polymorphisms, and their combined genotypes with the risk of atherosclerotic cerebral infarction (CI) in the Chinese population. Results We recruited 164 CI patients and 167 healthy control subjects who were frequency-matched for age and gender. The frequencies of the -509TT genotype and T allele gene were significantly higher in the CI group (P = 0.007, P = 0.006). The frequencies of +869CC genotype and C allele were higher in the CI group (P = 0.002, P = 0.004). In the CI group, the individuals with -509TT genotype had a significantly higher level of plasma triglyceride (TG) (P = 0.017). +869CC genotype correlated significantly with higher level of plasma low density lipoprotein cholesterol (LDL-c) in the CI group (P = 0.015). With haplotype analysis, the frequency of the -509T/+869C combined genotype was significantly higher in the CI group than in controls (P < 0.001). Conclusions Our study suggests that C-509T and T869C gene polymorphisms in TGF-β1 may be a critical risk factor of genetic susceptibility to CI in the Chinese population.
Collapse
Affiliation(s)
- Zhongxing Peng
- Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical College, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, P.R. China
| | | | | | | |
Collapse
|
39
|
Aihara KI, Ikeda Y, Yagi S, Akaike M, Matsumoto T. Transforming Growth Factor-β1 as a Common Target Molecule for Development of Cardiovascular Diseases, Renal Insufficiency and Metabolic Syndrome. Cardiol Res Pract 2010; 2011:175381. [PMID: 21234356 PMCID: PMC3018616 DOI: 10.4061/2011/175381] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 12/08/2010] [Indexed: 01/25/2023] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is a polypeptide member of the transforming growth factor β superfamily of cytokines. It is a secreted protein that performs many cellular functions including control of cell growth, cell proliferation, cell differentiation and apoptosis. In the cardiovascular system, TGF-β1 plays pivotal roles in the pathogenesis of hypertension, restenosis after percutaneous coronary intervention, atherosclerosis, cardiac hypertrophy and heart failure. In addition, TGF-β1 has been shown to be increased in adipose tissue of obese subjects with insulin resistance. Furthermore, TGF-β1 is a potent initiator of proliferation of renal mesangial cells leading to chronic kidney disease. Some currently available agents can manipulate TGF-β1 expression leading to amelioration of cardiovascular diseases. Thus, an understanding of interactions between chronic kidney disease and metabolic syndrome and the development of cardiovascular diseases is an important issue, and attention should be given to TGF-β1 as a crucial factor for regulation and modulation of those pathological conditions.
Collapse
Affiliation(s)
- Ken-Ichi Aihara
- Department of Medicine and Bioregulatory Sciences, The University of Tokushima, Graduate School of Health Biosciences, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | | | | | | | | |
Collapse
|
40
|
Deng HB, Jiang CQ, Tomlinson B, Liu B, Lin JM, Wong KS, Cheung BMY, Lam TH, Thomas GN. A polymorphism in transforming growth factor-β1 is associated with carotid plaques and increased carotid intima-media thickness in older Chinese men: the Guangzhou Biobank Cohort Study-Cardiovascular Disease Subcohort. Atherosclerosis 2010; 214:391-6. [PMID: 21167485 DOI: 10.1016/j.atherosclerosis.2010.11.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 11/15/2010] [Accepted: 11/15/2010] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Polymorphisms of the transforming growth factor-β1 (TGFB1) gene have not been associated with asymptomatic atherosclerosis previously. We investigated the relationship between a single nucleotide polymorphism (SNP) rs4803455 in TGFB1 and atherosclerosis identified by the presence of carotid plaque and increased intima-media thickness (IMT) in an older Chinese population. METHODS 1996 subjects (992 (49.7%) men aged 50-85 years) from the Guangzhou Biobank Cohort Study-Cardiovascular Subcohort (GBCS-CVD) were genotyped. Carotid plaque and IMT were assessed by B-mode ultrasonography. RESULTS In male subjects, the C allele of TGFB1 rs4803455 was significantly associated with prevalence of carotid plaque (adjusted OR: 2.49, 95% CI: 1.16-5.36, P = 0.03). The C allele was related to increased number of common carotid artery (CCA) plaques (P=0.03) and larger carotid plaque area (P = 0.02) in men. The homozygous carriers of allele C in male subjects also had a higher risk of having carotid IMT ≥ 1 mm (adjusted OR: 1.75, 95% CI: 1.05-2.93, P = 0.03). These associations were independent of age, smoking, physical activity, body mass index, blood pressure, lipid profile, fasting glucose and high sensitivity C-reactive protein. CONCLUSION This is the first study to show that the C allele in TGFB1 was associated with increased risk of atherosclerosis in older Chinese men. Further investigations on the linkage between the TGFB1 gene and progression of atherosclerosis in asymptomatic populations are warranted.
Collapse
Affiliation(s)
- Han-Bing Deng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Hermus L, Lefrandt JD, Tio RA, Breek JC, Zeebregts CJ. Carotid plaque formation and serum biomarkers. Atherosclerosis 2010; 213:21-9. [DOI: 10.1016/j.atherosclerosis.2010.05.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 05/08/2010] [Accepted: 05/10/2010] [Indexed: 10/19/2022]
|
42
|
The role of nucleotides in apoptotic cell clearance: implications for disease pathogenesis. J Mol Med (Berl) 2010; 89:13-22. [PMID: 20809090 DOI: 10.1007/s00109-010-0673-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 07/02/2010] [Accepted: 07/29/2010] [Indexed: 12/20/2022]
Abstract
Apoptosis occurs in many tissues, during both normal and pathogenic processes. Normally, apoptotic cells are rapidly cleared, either by neighboring or recruited phagocytes. The prompt clearance of apoptotic cells requires that the apoptotic cells announce their presence through the release of chemotactic factors, known as "find-me" signals, to recruit phagocytes to the site of death, and through the exposure of so-called "eat-me" signals, which are ligands for phagocytic uptake. The importance of prompt apoptotic cell clearance is revealed by findings that decreasing the efficiency of engulfment results in the persistence of apoptotic cells, which is often associated with chronic inflammation and autoimmunity. Additionally, the proper clearance of apoptotic cells is actively anti-inflammatory, which is thought to play a crucial role in immunologic tolerance. Therefore, defects associated with clearance of apoptotic cells may contribute to the pathogenesis of several inflammatory diseases, including autoimmunity and atherosclerosis. Here, we review the role of nucleotides in the apoptotic cell clearance process and discuss their implications for disease pathogenesis.
Collapse
|
43
|
TGF-β and microvessel homeostasis. Microvasc Res 2010; 80:166-73. [DOI: 10.1016/j.mvr.2010.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 02/23/2010] [Accepted: 03/04/2010] [Indexed: 12/17/2022]
|
44
|
Buday A, Orsy P, Godó M, Mózes M, Kökény G, Lacza Z, Koller A, Ungvári Z, Gross ML, Benyó Z, Hamar P. Elevated systemic TGF-beta impairs aortic vasomotor function through activation of NADPH oxidase-driven superoxide production and leads to hypertension, myocardial remodeling, and increased plaque formation in apoE(-/-) mice. Am J Physiol Heart Circ Physiol 2010; 299:H386-95. [PMID: 20511416 DOI: 10.1152/ajpheart.01042.2009] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The role of circulating, systemic TGF-beta levels in endothelial function is not clear. TGF-beta(1) may cause endothelial dysfunction in apolipoprotein E-deficient (apoE(-/-)) mice via stimulation of reactive oxygen species (ROS) production by the NADPH oxidase (NOX) system and aggravate aortic and heart remodeling and hypertension. Thoracic aorta (TA) were isolated from 4-mo-old control (C57Bl/6), apoE(-/-), TGF-beta(1)-overexpressing (TGFbeta(1)), and crossbred apoE(-/-) x TGFbeta(1) mice. Endothelium-dependent relaxation was measured before and after incubation with apocynin (NOX inhibitor) or superoxide dismutase (SOD; ROS scavenger). Superoxide production within the vessel wall was determined by dihydroethidine staining under confocal microscope. In 8-mo-old mice, aortic and myocardial morphometric changes, plaque formation by en face fat staining, and blood pressure were determined. Serum TGF-beta(1) levels (ELISA) were elevated in TGFbeta(1) mice without downregulation of TGF-beta-I receptor (immunohistochemistry). In the aortic wall, superoxide production was enhanced and NO-dependent relaxation diminished in apoE(-/-) x TGFbeta(1) mice but improved significantly after apocynin or SOD. Myocardial capillary density was reduced, fibrocyte density increased, aortic wall was thicker, combined lesion area was greater, and blood pressure was higher in the apoE(-/-) x TGFbeta vs. C57Bl/6 mice. Our results demonstrate that elevated circulating TGF-beta(1) causes endothelial dysfunction through NOX activation-induced oxidative stress, accelerating atherosclerosis and hypertension in apoE(-/-) mice. These findings may provide a mechanism explaining accelerated atherosclerosis in patients with elevated plasma TGFbeta(1).
Collapse
Affiliation(s)
- Anna Buday
- Institute of Pathophysiology, Dept. of Medicine, Semmelweis Univ., Budapest, 1089 Nagyvárad tér 4, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Xu S, Liu AC, Gotlieb AI. Common pathogenic features of atherosclerosis and calcific aortic stenosis: role of transforming growth factor-beta. Cardiovasc Pathol 2009; 19:236-47. [PMID: 19942455 DOI: 10.1016/j.carpath.2009.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2009] [Revised: 09/22/2009] [Accepted: 09/24/2009] [Indexed: 12/20/2022] Open
Abstract
Calcific aortic stenosis and atherosclerosis have been investigated separately in experimental in vitro and in vivo studies and in clinical studies. The similarities identified in both diseases suggest that similar pathogenic pathways are involved in both conditions. Most current therapeutic studies are focused on statins. The evidence suggests that statin effects on valves may, in large part, be independent of the lipid lowering effects of the drug. There are several molecules that play significant regulatory roles on the development and progression of valve sclerosis and calcification and on growth and complications of atherosclerotic plaques. The purpose of this review is to discuss the pathogenic features of the two conditions, highlight the important similarities, and then review the data that suggest that transforming growth factor-beta may play a key regulatory role in both diseases and that this is worthy of study as a potential therapeutic target for both conditions.
Collapse
Affiliation(s)
- Songyi Xu
- Toronto General Research Institute and Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| | | | | |
Collapse
|
46
|
Park JD, Kim YY, Lee CY. Identification of epistasis in ischemic stroke using multifactor dimensionality reduction and entropy decomposition. BMB Rep 2009; 42:617-22. [DOI: 10.5483/bmbrep.2009.42.9.617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
47
|
Spectroscopy to improve identification of vulnerable plaques in cardiovascular disease. Int J Cardiovasc Imaging 2009; 26:111-9. [PMID: 19760516 DOI: 10.1007/s10554-009-9500-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Accepted: 08/19/2009] [Indexed: 10/20/2022]
Abstract
Many apparent healthy persons die from cardiovascular disease, despite major advances in prevention and treatment of cardiovascular disease. Traditional cardiovascular risk factors are able to predict cardiovascular events in the long run, but fail to assess current disease activity or nearby cardiovascular events. There is a clear relation between the occurrence of cardiovascular events and the presence of so-called vulnerable plaques. These vulnerable plaques are characterized by active inflammation, a thin cap and a large lipid pool. Spectroscopy is an optical imaging technique which depicts the interaction between light and tissues, and thereby shows the biochemical composition of tissues. In recent years, impressive advances have been made in spectroscopy technology and intravascular spectroscopy is able to assess the composition of plaques of interest and thereby to identify and actually quantify plaque vulnerability. This review summarizes the current evidence for spectroscopy as a measure of plaque vulnerability and discusses the potential role of intravascular spectroscopic imaging techniques.
Collapse
|
48
|
Kona S, Chellamuthu P, Xu H, Hills SR, Nguyen KT. Effects of cyclic strain and growth factors on vascular smooth muscle cell responses. Open Biomed Eng J 2009; 3:28-38. [PMID: 19812708 PMCID: PMC2757671 DOI: 10.2174/1874120700903010028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Revised: 07/06/2009] [Accepted: 07/09/2009] [Indexed: 01/09/2023] Open
Abstract
Under physiological and pathological conditions, vascular smooth muscle cells (SMC) are exposed to different biochemical factors and biomechanical forces. Previous studies pertaining to SMC responses have not investigated the effects of both factors on SMCs. Thus, in our research we investigated the combined effects of growth factors like Bfgf (basic fibroblast growth factor), TGF-β (transforming growth factor β) and PDGF (platelet-derived growth factor) along with physiological cyclic strain on SMC responses. Physiological cyclic strain (10% strain) significantly reduced SMC proliferation compared to static controls while addition of growth factors bFGF, TGF-β or PDGF-AB had a positive influence on SMC growth compared to strain alone. Microarray analysis of SMCs exposed to these growth factors and cyclic strain showed that several bioactive genes (vascular endothelial growth factor, epidermal growth factor receptor, etc.) were altered upon exposure. Further work involving biochemical and pathological cyclic strain stimulation will help us better understand the role of cyclic strain and growth factors in vascular functions and development of vascular disorders.
Collapse
Affiliation(s)
- Soujanya Kona
- Department of Bioengineering, University of Texas at Arlington and University of Texas Southwestern Medical Center at Dallas, USA
| | | | | | | | | |
Collapse
|
49
|
Churchman AT, Anwar AA, Li FYL, Sato H, Ishii T, Mann GE, Siow RCM. Transforming growth factor-beta1 elicits Nrf2-mediated antioxidant responses in aortic smooth muscle cells. J Cell Mol Med 2009; 13:2282-2292. [PMID: 19674192 DOI: 10.1111/j.1582-4934.2009.00874.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The anti-inflammatory properties of transforming growth factor-beta(1) (TGF-beta(1)) account for its protection against atherosclerotic plaque rupture. This study investigates whether activation of the Nrf2 (nuclear factor erythroid 2 [NF-E2]-related factor 2) transcription pathway is involved in TGF-beta(1) mediated induction of the antioxidant enzyme heme oxygenase-1 (HO-1) in smooth muscle cells (SMC). Human aortic smooth muscle cells (HAoSMC) or wild-type and Nrf2-deficient mouse (MAoSMC) aortic SMC were treated with TGF-beta(1) (2.5-10 ng/ml, 0-24 hrs). We report the first evidence that TGF-beta(1) induces Nrf2 mediated HO-1 expression and antioxidant response element activity, which was paralleled by enhanced superoxide production and expression of the NAD(P)H oxidase subunit p22(phox). TGF-beta(1) failed to induce HO-1 expression in MAoSMC derived from Nrf2-deficient mice, and HO-1 induction by TGF-beta(1) in HAoSMC was attenuated by inhibition of extracellular signal regulated kinase or c-jun-N-terminal kinase but not p38 mitogen activated protein kinase. Inhibition of NAD(P)H oxidase or scavenging of superoxide diminished HO-1 induction in response to TGF-beta(1). The oxidative stress agents glucose oxidase (GOx) and diethylmaleate enhanced TGF-beta(1) generation and HO-1 expression in HAoSMC, while antagonism of TGF-beta(1) signalling by adenoviral Smad7 overexpression attenuated their induction of HO-1. Pre-treatment of HAoSMC with TGF-beta(1) reduced nuclear translocation of the pro-apoptotic mediator p53 elicited by GOx. Our findings demonstrate that Nrf2 is a new target of TGF-beta(1) signalling in the vasculature which may contribute to the atheroprotective properties attributed to this growth factor.
Collapse
Affiliation(s)
| | - Anila A Anwar
- Cardiovascular Division, School of Medicine, King's College London, UK
| | - Francois Y L Li
- Cardiovascular Division, School of Medicine, King's College London, UK
| | - Hideyo Sato
- Department of Bioresources, Faculty of Agriculture, Yamagata University, Tsuruoka, Japan
| | - Tetsuro Ishii
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tuskuba, Japan
| | - Giovanni E Mann
- Cardiovascular Division, School of Medicine, King's College London, UK
| | - Richard C M Siow
- Cardiovascular Division, School of Medicine, King's College London, UK
| |
Collapse
|
50
|
Khanna AK. Enhanced susceptibility of cyclin kinase inhibitor p21 knockout mice to high fat diet induced atherosclerosis. J Biomed Sci 2009; 16:66. [PMID: 19604372 PMCID: PMC2720941 DOI: 10.1186/1423-0127-16-66] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 07/15/2009] [Indexed: 11/10/2022] Open
Abstract
Cyclin kinase inhibitor p21 is one of the most potent inhibitors of aortic smooth muscle cell proliferation, a key mediator of atherosclerosis. This study tests if p2l deficiency will result in severe atherosclerosis in a mouse model. p21-/- and strain matched wild type mice were fed with high fat diet for 21 weeks. Analysis for biochemical parameters (cholesterol, triglycerides) in serum and mRNA expression of CD36, HO-1, TGF-beta, IFN-gamma, TNF-alpha, PPAR-gamma and NADPH oxidase components (p22phox, NOX-1 and Rac-1) was performed in aortic tissues by Real Time PCR. p21-/- mice gained significantly (p < 0.01) more weight than wild type mice, triglycerides (p < 0.05) and cholesterol levels (p < 0.01) were more pronounced in the sera of p21-/- compared to wild type mice fed with high fat diet. High fat diet resulted in significantly decreased TGF-beta (p < 0.02), HO-l (p < 0.02) and increased CD36 (p < 0.03) mRNA expression in aortic tissues of p21-/- mice compared to animal fed with regular diet. IFN-gamma mRNA expression (235 +/- 11 folds) increased significantly in high fat diet fed p21-/- mice and a multifold modulation of PPAR-gamma(136 +/- 7), p22phox, NOX-1 and Rac-1 (15-35-folds) mRNA in aortic tissues from p21-/- mice compared to the wild type mice. Severity of atherosclerotic lesions was significantly higher in p21-/- compared to wild type mice. The results demonstrate that the deficiency of p21 leads to altered expression of pro-atherogenic genes, and severe atherosclerosis in mice fed with high fat diet. This opens the possibility of p21 protein as a therapeutic tool to control progression of atherosclerosis.
Collapse
Affiliation(s)
- Ashwani K Khanna
- Department of Medicine (Cardiology), University of Maryland, Baltimore, USA.
| |
Collapse
|