1
|
Blessing E, Teichmann E, Hinz B. Anandamide Inhibits Vascular Smooth Muscle Migration, Endothelial Adhesion Protein Expression and Monocyte Adhesion of Human Coronary Artery Cells. Cells 2024; 13:2108. [PMID: 39768198 PMCID: PMC11727187 DOI: 10.3390/cells13242108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
Endocannabinoids have been shown to play a complex role in the pathophysiology of a number of cardiovascular disorders. In the present study, the effects of the two major endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG) were investigated in human coronary artery smooth muscle cells (HCASMC) and human coronary artery endothelial cells (HCAEC) with regard to potential atheroprotective and anti-inflammatory effects. In HCASMC, AEA showed an inhibitory effect on platelet-derived growth factor-induced migration, but not proliferation, independent of major cannabinoid-activatable receptors (CB1, CB2, TRPV1), while 2-AG left both responses unaffected. In HCAEC, AEA at concentrations of 6 and 10 µM significantly inhibited the interleukin (IL)-1β- and lipopolysaccharide (LPS)-stimulated expression of vascular cell adhesion molecule-1 (VCAM-1) and LPS-induced intercellular adhesion molecule-1 (ICAM-1), again independently of the abovementioned receptors. Corresponding effects were observed to a lesser extent in the presence of 2-AG, in most cases not significantly. The detection of activated phosphoproteins as well as experiments with inhibitors of corresponding signaling pathways suggest that AEA interferes with IL-1β-induced VCAM-1 expression via inhibition of protein kinase B/Akt and Src kinase activation and attenuates LPS-induced VCAM-1 and ICAM-1 expression via inhibition of signal transducer and activator of transcription 3 (STAT3) phosphorylation. As expected, AEA also led to a significant inhibition of monocyte adhesion to IL-1β- and LPS-stimulated HCAEC, with siRNA experiments confirming the functional role of VCAM-1 and ICAM-1 in this assay. 2-AG showed a comparatively weaker but, in the case of LPS stimulation, still significant inhibition of adhesion. In summary, the results emphasize the potential of AEA as a protective regulator of atherosclerotic and inflammation-related changes in HCASMC and HCAEC and encourage further corresponding preclinical studies with this endocannabinoid.
Collapse
Affiliation(s)
| | | | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany; (E.B.)
| |
Collapse
|
2
|
Yu Z, Zhang H, Li L, Li Z, Chen D, Pang X, Ji Y, Wang Y. Microglia-mediated pericytes migration and fibroblast transition via S1P/S1P3/YAP signaling pathway after spinal cord injury. Exp Neurol 2024; 379:114864. [PMID: 38866101 DOI: 10.1016/j.expneurol.2024.114864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
Platelet-derived growth factor receptor β positive (PDGFRβ+) pericytes detach from the microvascular wall and migrate into the injury center following spinal cord injury (SCI), which has been widely regarded as the main source of fibrotic scar, but the mechanism of migration and fibroblast transition remains elusive. Here we show the associated spatiotemporal distribution between microglia and pericytes at three and seven days post-injury (dpi). The increased expression of Sphingosine kinase-1 (SPHK1) in microglia significantly raised the concentration of Sphingosine-1-phosphate (S1P) in the spinal cord, which promotes migration and fibroblast transition of pericyte. In vitro experiments, we found the elevated Sphingosine 1-phosphate receptor 3 (S1P3), the S1P/S1PR3 axis inhibited the phosphorylation of YAP and promoted its nuclear translocation, which contributed to the formation of alpha-smooth muscle actin (α-SMA) and collagen type I (COL1) protein, This process can be blocked by an S1P3 specific inhibitor TY52156 in vitro. The S1P/S1P3/YAP pathway might be a potential target for treatment in SCI.
Collapse
Affiliation(s)
- Ziyuan Yu
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Huabin Zhang
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Linxi Li
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Zhi Li
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Danmin Chen
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Xiao Pang
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Yunxiang Ji
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China
| | - Yezhong Wang
- Guangzhou Med Univ, Inst Neurosci, Dept Neurosurg, Affiliated Hosp 2, Guangzhou 510260, PR China.
| |
Collapse
|
3
|
Yang S, Zhao Y, Cao S, Liu X, Feng M, Chen Y, Ma C, Zhan T, Zhang Q, Jia H, Zhao Y, Tong M, Yu Y, Liu X, Yang B, Zhang Y. Kanglexin counters vascular smooth muscle cell dedifferentiation and associated arteriosclerosis through inhibiting PDGFR. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155704. [PMID: 38759316 DOI: 10.1016/j.phymed.2024.155704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Dysregulation of vascular smooth muscle cell (VSMC) function leads to a variety of diseases such as atherosclerosis and hyperplasia after injury. However, antiproliferative drug targeting VSMC exhibits poor specificity. Therefore, there is an urgent to develop highly specific antiproliferative drugs to prevention and treatment VSMC dedifferentiation associated arteriosclerosis. Kanglexin (KLX), a new anthraquinone compound designed by our team, has potential to regulate VSMC phenotype according to the physicochemical properties. PURPOSE This project aims to evaluate the therapeutic role of KLX in VSMC dedifferentiation and atherosclerosis, neointimal formation and illustrates the underlying molecular mechanism. METHODS In vivo, the ApoE-/- mice were fed with high-fat diet (HFD) for a duration of 13 weeks to establish the atherosclerotic model. And rat carotid artery injury model was performed to establish the neointimal formation model. In vitro, PDGF-BB was used to induce VSMC dedifferentiation. RESULTS We found that KLX ameliorated the atherosclerotic progression including atherosclerotic lesion formation, lipid deposition and collagen deposition in aorta and aortic sinus in atherosclerotic mouse model. In addition, The administration of KLX effectively ameliorated neointimal formation in the carotid artery following balloon injury in SD rats. The findings derived from molecular docking and surface plasmon resonance (SPR) experiments unequivocally demonstrate that KLX had potential to bind PDGFR-β. Mechanism research work proved that KLX prevented VSMC proliferation, migration and dedifferentiation via activating the PDGFR-β-MEK -ERK-ELK-1/KLF4 signaling pathway. CONCLUSION Collectively, we demonstrated that KLX effectively attenuated the progression of atherosclerosis in ApoE-/- mice and carotid arterial neointimal formation in SD rats by inhibiting VSMC phenotypic conversion via PDGFR-β-MEK-ERK-ELK-1/KLF4 signaling. KLX exhibits promising potential as a viable therapeutic agent for the treatment of VSMC phenotype conversion associated arteriosclerosis.
Collapse
Affiliation(s)
- Shuang Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yixiu Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Shifeng Cao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xinxin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Min Feng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yi Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chunyue Ma
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Tingting Zhan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Qi Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Honglin Jia
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yu Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Ming Tong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yuanyuan Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xue Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Baofeng Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| | - Yan Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
4
|
Cauwenberghs N, Verheyen A, Sabovčik F, Ntalianis E, Vanassche T, Brguljan J, Kuznetsova T. Serum proteomic profiling of carotid arteriopathy: A population outcome study. Atherosclerosis 2023; 385:117331. [PMID: 37879154 DOI: 10.1016/j.atherosclerosis.2023.117331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/22/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND AND AIMS Circulating proteins reflecting subclinical vascular disease may improve prediction of atherosclerotic cardiovascular disease (ASCVD). We applied feature selection and unsupervised clustering on proteomic data to identify proteins associated with carotid arteriopathy and construct a protein-based classifier for ASCVD event prediction. METHODS 491 community-dwelling participants (mean age, 58 ± 11 years; 51 % women) underwent carotid ultrasonography and proteomic profiling (CVD II panel, Olink Proteomics). ASCVD outcome was collected (median follow-up time: 10.2 years). We applied partial least squares (PLS) to identify proteins linked to carotid intima-media thickness (cIMT). Next, we assessed the association between future ASCVD events and protein-based phenogroups derived by unsupervised clustering (Gaussian Mixture modelling) based on proteins selected in PLS. RESULTS PLS identified 19 proteins as important, which were all associated with cIMT in multivariable-adjusted linear regression. 8 of the 19 proteins were excluded from the clustering analysis because of high collinearity. Based on the 11 remaining proteins, the clustering algorithm subdivided the cohort into two phenogroups. Compared to the first phenogroup (n = 177), participants in the second phenogroup (n = 314) presented: i) a more unfavorable lipid profile with higher total cholesterol and triglycerides and lower HDL cholesterol (p ≤ 0.014); ii) higher cIMT (p = 0.0020); and iii) a significantly higher risk for future ASCVD events (multivariable-adjusted hazard ratio (95 % CI) versus phenogroup 1: 2.05 (1.26-3.52); p = 0.0093). The protein-based phenogrouping supplemented ACC/AHA 10-year ASCVD risk scoring for prediction of a first ASCVD event. CONCLUSIONS Focused protein-based phenogrouping identified individuals at high risk for future ASCVD and may complement current risk stratification strategies.
Collapse
Affiliation(s)
- Nicholas Cauwenberghs
- Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Belgium.
| | - Astrid Verheyen
- Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Belgium
| | - František Sabovčik
- Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Belgium
| | - Evangelos Ntalianis
- Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Belgium
| | - Thomas Vanassche
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Jana Brguljan
- Hypertension Department, University Medical Centre Ljubljana, Medical University Ljubljana, Ljubljana, Slovenia
| | - Tatiana Kuznetsova
- Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Belgium
| |
Collapse
|
5
|
Gregorczyk-Maga I, Szustkiewicz-Karoń A, Gajda M, Kapusta M, Maga W, Schönborn M. The Concentration of Pro- and Antiangiogenic Factors in Saliva and Gingival Crevicular Fluid Compared to Plasma in Patients with Peripheral Artery Disease and Type 2 Diabetes. Biomedicines 2023; 11:1596. [PMID: 37371691 DOI: 10.3390/biomedicines11061596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Several studies have investigated various biomarkers in relation to peripheral artery disease (PAD) for disease stratification and early-onset detection. In PAD, angiogenesis is required for tissue restoration and tissue perfusion. Considering changes in angiogenesis in patients with PAD, angiogenic factors could be explored as one of the new prognostic molecules. In recent studies, saliva and gingival crevicular fluid (GCF) have gained recognition as new, easily obtained diagnostic materials. This study aimed to compare the levels of selected circulating angiogenic factors (VEGF-A, PDGF-BB, and ANG-1) in unstimulated whole saliva (WS) and GCF versus plasma at three points in time to find possible correlations between their concentrations among patients with PAD and diabetes type 2 in 32 patients with Rutherford stages 5 and 6. A significant positive correlation has been demonstrated between circulating PDGF-BB levels in GCF and plasma. In most cases, comorbidities do not have an impact on the change in general correlation for the whole group. Our results clearly showed that GCF could be a good source for PDGF assessment. However, future studies with a larger number of subjects are warranted to confirm this finding and identify the most accurate angiogenic biomarkers in saliva or GCF that could be applied in clinical practice.
Collapse
Affiliation(s)
- Iwona Gregorczyk-Maga
- Faculty of Medicine, Institute of Dentistry, Jagiellonian University Medical College, 31-155 Krakow, Poland
| | | | - Mateusz Gajda
- Department of Angiology, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland
- Doctoral School of Medicine and Health Sciences, Jagiellonian University Medical College, 31-121 Krakow, Poland
| | - Maria Kapusta
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Wojciech Maga
- Faculty of Medicine, Institute of Dentistry, Jagiellonian University Medical College, 31-155 Krakow, Poland
| | - Martyna Schönborn
- Department of Angiology, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland
- Doctoral School of Medicine and Health Sciences, Jagiellonian University Medical College, 31-121 Krakow, Poland
| |
Collapse
|
6
|
O’Morain VL, Chen J, Plummer SF, Michael DR, Ramji DP. Anti-Atherogenic Actions of the Lab4b Consortium of Probiotics In Vitro. Int J Mol Sci 2023; 24:ijms24043639. [PMID: 36835055 PMCID: PMC9964490 DOI: 10.3390/ijms24043639] [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: 11/04/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Probiotic bacteria have many protective effects against inflammatory disorders, though the mechanisms underlying their actions are poorly understood. The Lab4b consortium of probiotics contains four strains of lactic acid bacteria and bifidobacteria that are reflective of the gut of newborn babies and infants. The effect of Lab4b on atherosclerosis, an inflammatory disorder of the vasculature, has not yet been determined and was investigated on key processes associated with this disease in human monocytes/macrophages and vascular smooth muscle cells in vitro. The Lab4b conditioned medium (CM) attenuated chemokine-driven monocytic migration, monocyte/macrophage proliferation, uptake of modified LDL and macropinocytosis in macrophages together with the proliferation and platelet-derived growth factor-induced migration of vascular smooth muscle cells. The Lab4b CM also induced phagocytosis in macrophages and cholesterol efflux from macrophage-derived foam cells. The effect of Lab4b CM on macrophage foam cell formation was associated with a decrease in the expression of several key genes implicated in the uptake of modified LDL and induced expression of those involved in cholesterol efflux. These studies reveal, for the first time, several anti-atherogenic actions of Lab4b and strongly implicate further studies in mouse models of the disease in vivo and in clinical trials.
Collapse
Affiliation(s)
- Victoria L. O’Morain
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Jing Chen
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Sue F. Plummer
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, UK
| | - Daryn R. Michael
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, UK
| | - Dipak P. Ramji
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
- Correspondence: ; Tel.: +44-(0)29-20876753
| |
Collapse
|
7
|
Bale BF, Doneen AL, Leimgruber PP, Vigerust DJ. The critical issue linking lipids and inflammation: Clinical utility of stopping oxidative stress. Front Cardiovasc Med 2022; 9:1042729. [PMID: 36439997 PMCID: PMC9682196 DOI: 10.3389/fcvm.2022.1042729] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/24/2022] [Indexed: 07/30/2023] Open
Abstract
The formation of an atheroma begins when lipoproteins become trapped in the intima. Entrapped lipoproteins become oxidized and activate the innate immune system. This immunity represents the primary association between lipids and inflammation. When the trapping continues, the link between lipids and inflammation becomes chronic and detrimental, resulting in atherosclerosis. When entrapment ceases, the association between lipids and inflammation is temporary and healthy, and the atherogenic process halts. Therefore, the link between lipids and inflammation depends upon lipoprotein retention in the intima. The entrapment is due to electrostatic forces uniting apolipoprotein B to polysaccharide chains on intimal proteoglycans. The genetic transformation of contractile smooth muscle cells in the media into migratory secretory smooth muscle cells produces the intimal proteoglycans. The protein, platelet-derived growth factor produced by activated platelets, is the primary stimulus for this genetic change. Oxidative stress is the main stimulus to activate platelets. Therefore, minimizing oxidative stress would significantly reduce the retention of lipoproteins. Less entrapment decreases the association between lipids and inflammation. More importantly, it would halt atherogenesis. This review will analyze oxidative stress as the critical link between lipids, inflammation, and the pathogenesis of atherosclerosis. Through this perspective, we will discuss stopping oxidative stress to disrupt a harmful association between lipids and inflammation. Numerous therapeutic options will be discussed to mitigate oxidative stress. This paper will add a new meaning to the Morse code distress signal SOS-stopping oxidative stress.
Collapse
Affiliation(s)
- Bradley Field Bale
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
| | - Amy Lynn Doneen
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
| | - Pierre P. Leimgruber
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
- Department of Medical Education and Clinical Sciences, University of Washington School of Medicine, Seattle, WA, United States
| | - David John Vigerust
- Department of Neurological Surgery, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
8
|
Snelder SM, Pouw N, Aga Y, Castro Cabezas M, Biter LU, Zijlstra F, Kardys I, van Dalen BM. Cardiovascular Biomarker Profiles in Obesity and Relation to Normalization of Subclinical Cardiac Dysfunction after Bariatric Surgery. Cells 2022; 11:cells11030422. [PMID: 35159232 PMCID: PMC8834585 DOI: 10.3390/cells11030422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
Aims: We aimed to gain insight into the underlying pathophysiology of cardiac dysfunction in obesity patients and the improvement of cardiac function after weight loss. Methods: This is a longitudinal study in which 92 cardiovascular biomarkers were measured by multiplex immunoassays in obesity patients without known cardiovascular disease, before and one year after bariatric surgery. Results: Out of 100 eligible patients, 72 patients completed the follow-up. A total of 72 (78%) biomarkers changed significantly. The biomarkers with the highest relative changes represented processes linked mainly to insulin resistance and inflammation. In the patients with persistent subclinical cardiac dysfunction, the baseline values of 10 biomarkers were different from values in patients with normalization of cardiac function. Most of these biomarkers were linked to inflammation or atherosclerosis. Finally, a model was developed to investigate the relationship between changes in the biomarkers and persistent subclinical cardiac dysfunction. Seven biomarkers were retained in this model, mainly linked to inflammation, atherosclerosis, and hypercoagulability. Conclusion: The majority (78%) of cardiovascular biomarkers changed, pointing mainly to modulation of insulin resistance and inflammation. The baseline levels of 10 biomarkers, as well as pre- to post-bariatric surgery changes in seven biomarkers, were related to persistent subclinical cardiac dysfunction after bariatric surgery.
Collapse
Affiliation(s)
- Sanne M. Snelder
- Department of Cardiology, Franciscus Gasthuis & Vlietland, 3045 PM Rotterdam, The Netherlands; (S.M.S.); (Y.A.)
| | - Nadine Pouw
- Department of Clinical Chemistry, Franciscus Gasthuis & Vlietland, 3045 PM Rotterdam, The Netherlands;
| | - Yaar Aga
- Department of Cardiology, Franciscus Gasthuis & Vlietland, 3045 PM Rotterdam, The Netherlands; (S.M.S.); (Y.A.)
| | - Manuel Castro Cabezas
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, 3045 PM Rotterdam, The Netherlands;
| | - L. Ulas Biter
- Department of Surgery, Franciscus Gasthuis & Vlietland, 3045 PM Rotterdam, The Netherlands;
| | - Felix Zijlstra
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (F.Z.); (I.K.)
| | - Isabella Kardys
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (F.Z.); (I.K.)
| | - Bas M. van Dalen
- Department of Cardiology, Franciscus Gasthuis & Vlietland, 3045 PM Rotterdam, The Netherlands; (S.M.S.); (Y.A.)
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (F.Z.); (I.K.)
- Correspondence: ; Tel.: +31-10-4616139
| |
Collapse
|
9
|
Cheng YW, Zhang ZB, Lan BD, Lin JR, Chen XH, Kong LR, Xu L, Ruan CC, Gao PJ. PDGF-D activation by macrophage-derived uPA promotes AngII-induced cardiac remodeling in obese mice. J Exp Med 2021; 218:e20210252. [PMID: 34236404 PMCID: PMC8273546 DOI: 10.1084/jem.20210252] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/03/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity-induced secretory disorder of adipose tissue-derived factors is important for cardiac damage. However, whether platelet-derived growth factor-D (PDGF-D), a newly identified adipokine, regulates cardiac remodeling in angiotensin II (AngII)-infused obese mice is unclear. Here, we found obesity induced PDGF-D expression in adipose tissue as well as more severe cardiac remodeling compared with control lean mice after AngII infusion. Adipocyte-specific PDGF-D knockout attenuated hypertensive cardiac remodeling in obese mice. Consistently, adipocyte-specific PDGF-D overexpression transgenic mice (PA-Tg) showed exacerbated cardiac remodeling after AngII infusion without high-fat diet treatment. Mechanistic studies indicated that AngII-stimulated macrophages produce urokinase plasminogen activator (uPA) that activates PDGF-D by splicing full-length PDGF-D into the active PDGF-DD. Moreover, bone marrow-specific uPA knockdown decreased active PDGF-DD levels in the heart and improved cardiac remodeling in HFD hypertensive mice. Together, our data provide for the first time a new interaction pattern between macrophage and adipocyte: that macrophage-derived uPA activates adipocyte-secreted PDGF-D, which finally accelerates AngII-induced cardiac remodeling in obese mice.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Bei Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei-Di Lan
- Department of Cardiology, First Affiliated Hospital, Xi’an Jiao Tong University, Xi’an, Shanxi, China
| | - Jing-Rong Lin
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Hui Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling-Ran Kong
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Xu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-Chao Ruan
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Wang Y, Zhang Y, Gao X, Qian J, Yang J, Sun W, Wang H, Yang Y. Resistin-like molecule beta augments phenotypic modulation of human aortic smooth muscle cell triggered by high glucose. Endocr J 2021; 68:461-468. [PMID: 33441498 DOI: 10.1507/endocrj.ej20-0343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Vascular muscle cells (VSMCs) participate in the pathophysiology of atherosclerosis. Resistin-like molecule beta (Relmβ) contributes to atherosclerosis development by activating macrophage. This study aims to investigate whether Relmβ regulates VSMC phenotypic modulation under high glucose environment. Human aortic vascular smooth muscle cells were cultured and treated with Relmβ in the presence or absence of high glucose. VSMC phenotypic modulation was assessed by expression of related markers. The migration of VSMCs was detected by wound healing assay and transwell assay. The proliferation of VSMCs was measured using CCK-8 assay. In this study, we observed that Relmβ modulated VSMC phenotypic modulation by down-regulating expression of smooth muscle α-actin (α-SMA), smooth muscle myosin heavy chain (SM-MHC), and calponin while up-regulating expression of osteopontin (OPN). Relmβ increased the expression of inflammatory genes in VSMCs. Relmβ also augmented VSMCs migration as well as proliferation. It is worth noting that all the effects of VSMCs were enhanced upon high glucose stimulation. The phosphorylation levels of p38MAPK and ERK1/2 were increased by co-treatment with Relmβ and high glucose. The p38 MAPK pathway inhibitor RWJ64809 and pERK1/2 inhibitor PD98059 significantly inhibited the proliferation of VSMCs induced by Relmβ and high glucose. Our results provide evidence that Relmβ augments phenotypic modulation and migration of human aortic smooth muscle cell induced by high glucose. Relmβ might be a potential target for treatment of atherosclerosis induced by hyperglycemia.
Collapse
Affiliation(s)
- Yi Wang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yawen Zhang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiangyu Gao
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jiali Qian
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jia Yang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wanwan Sun
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hao Wang
- Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Yehong Yang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
11
|
Enayati A, Johnston TP, Sahebkar A. Anti-atherosclerotic Effects of Spice-Derived Phytochemicals. Curr Med Chem 2021; 28:1197-1223. [PMID: 32368966 DOI: 10.2174/0929867327666200505084620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 11/22/2022]
Abstract
Cardiovascular diseases are the leading cause of death in the world. Atherosclerosis is characterized by oxidized lipid deposition and inflammation in the arterial wall and represents a significant problem in public health and medicine. Some dietary spices have been widely used in many countries; however, the mechanism of their action as it relates to the prevention and treatment of atherosclerosis is still poorly understood. In this review, we focus on the properties of various spice-derived active ingredients used in the prevention and treatment of atherosclerosis, as well as associated atherosclerotic risk factors. We provide a summary of the mechanisms of action, epidemiological analyses, and studies of various components of spice used in the clinic, animal models, and cell lines related to atherosclerosis. Most notably, we focused on mechanisms of action by which these spice-derived compounds elicit their lipid-lowering, anti-inflammatory, antioxidant, and immunomodulatory properties, as well as their involvement in selected biochemical and signal transduction pathways. It is suggested that future research should aim to design well-controlled clinical trials and more thoroughly investigate the role of spices and their active components in the prevention/treatment of atherosclerosis. Based on this literature review, it appears that spices and their active components are well tolerated and have few adverse side effects and, therefore, provide a promising adjunctive treatment strategy for patients with atherosclerosis.
Collapse
Affiliation(s)
- Ayesheh Enayati
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, United States
| | | |
Collapse
|
12
|
Chen L, Wu H, Ren C, Liu G, Zhang W, Liu W, Lu P. Inhibition of PDGF-BB reduces alkali-induced corneal neovascularization in mice. Mol Med Rep 2021; 23:238. [PMID: 33537811 PMCID: PMC7893695 DOI: 10.3892/mmr.2021.11877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to investigate the role of platelet-derived growth factor (PDGF)-BB/PDGF receptor (R)-β signaling in an experimental murine corneal neovascularization (CrNV) model. Experimental CrNV was induced by alkali injury. The intra-corneal expression of PDGF-BB was examined using immunohistochemistry. The effect of PDGF-BB on CrNV was evaluated using immunofluorescence staining. The expression levels of PDGFR-β in human retinal endothelial cells (HRECs) under normal conditions or following cobalt chloride treatment, which induced hypoxic conditions, was assessed using reverse transcription-quantitative PCR. The effect of exogenous treatment of PDGF-BB on the proliferation, migration and tube formation of HRECs under normoxic or hypoxic conditions was evaluated in vitro using Cell Counting Kit-8, wound healing and 3D Matrigel capillary tube formation assays, respectively. The results indicated that the intra-corneal expression levels of the proteins of PDGF-BB and PDGFR-β were detectable on days 2 and 7 following alkali injury. The treatment with neutralizing anti-PDGF-BB antibody resulted in significant inhibition of CrNV. The intra-corneal expression levels of vascular endothelial growth factor A, matrix metallopeptidase (MMP)-2 and MMP-9 proteins were downregulated, while the expression levels of thrombospondin (TSP)-1, TSP-2, a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-1 and ADAMTS-2 were upregulated significantly in mice treated with anti-PDGF-BB antibody. The expression levels of PDGFR-β were upregulated in HRECs under hypoxic conditions compared with those noted under normoxic conditions. Recombinant human PDGF-BB promoted the proliferation, migration and tube formation of HRECs under hypoxic conditions. The data indicated that PDGF-BB/PDGFR-β signaling was involved in CrNV and that it promoted endothelial cell proliferation, migration and tube formation. The pro-angiogenic effects of this pathway may be mediated via the induction of pro-angiogenic cytokine secretion and the suppression of anti-angiogenic cytokine secretion.
Collapse
Affiliation(s)
- Lei Chen
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongya Wu
- Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Chi Ren
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Gaoqin Liu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wenpeng Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Weiming Liu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Peirong Lu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
13
|
Wang L, Tang C. Targeting Platelet in Atherosclerosis Plaque Formation: Current Knowledge and Future Perspectives. Int J Mol Sci 2020; 21:ijms21249760. [PMID: 33371312 PMCID: PMC7767086 DOI: 10.3390/ijms21249760] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 12/23/2022] Open
Abstract
Besides their role in hemostasis and thrombosis, it has become increasingly clear that platelets are also involved in many other pathological processes of the vascular system, such as atherosclerotic plaque formation. Atherosclerosis is a chronic vascular inflammatory disease, which preferentially develops at sites under disturbed blood flow with low speeds and chaotic directions. Hyperglycemia, hyperlipidemia, and hypertension are all risk factors for atherosclerosis. When the vascular microenvironment changes, platelets can respond quickly to interact with endothelial cells and leukocytes, participating in atherosclerosis. This review discusses the important roles of platelets in the plaque formation under pro-atherogenic factors. Specifically, we discussed the platelet behaviors under disturbed flow, hyperglycemia, and hyperlipidemia conditions. We also summarized the molecular mechanisms involved in vascular inflammation during atherogenesis based on platelet receptors and secretion of inflammatory factors. Finally, we highlighted the studies of platelet migration in atherogenesis. In general, we elaborated an atherogenic role of platelets and the aspects that should be further studied in the future.
Collapse
Affiliation(s)
- Lei Wang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou 215123, China;
| | - Chaojun Tang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou 215123, China;
- Collaborative Innovation Center of Hematology of Jiangsu Province, Soochow University, Suzhou 215123, China
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 215123, China
- Correspondence: ; Tel.: +86-512-6588-0899
| |
Collapse
|
14
|
Targeting and imaging of monocyte-derived macrophages in rat's injured artery following local delivery of liposomal quantum dots. J Control Release 2020; 318:145-157. [DOI: 10.1016/j.jconrel.2019.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/26/2019] [Accepted: 12/08/2019] [Indexed: 12/27/2022]
|
15
|
The Emerging Role of Mesenchymal Stem Cells in Vascular Calcification. Stem Cells Int 2019; 2019:2875189. [PMID: 31065272 PMCID: PMC6466855 DOI: 10.1155/2019/2875189] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/12/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
Vascular calcification (VC), characterized by hydroxyapatite crystal depositing in the vessel wall, is a common pathological condition shared by many chronic diseases and an independent risk factor for cardiovascular events. Recently, VC is regarded as an active, dynamic cell-mediated process, during which calcifying cell transition is critical. Mesenchymal stem cells (MSCs), with a multidirectional differentiation ability and great potential for clinical application, play a duplex role in the VC process. MSCs facilitate VC mainly through osteogenic transformation and apoptosis. Meanwhile, several studies have reported the protective role of MSCs. Anti-inflammation, blockade of the BMP2 signal, downregulation of the Wnt signal, and antiapoptosis through paracrine signaling are possible mechanisms. This review displays the evidence both on the facilitating role and on the protective role of MSCs, then discusses the key factors determining this divergence.
Collapse
|
16
|
Lu W, Li X. PDGFs and their receptors in vascular stem/progenitor cells: Functions and therapeutic potential in retinal vasculopathy. Mol Aspects Med 2018; 62:22-32. [DOI: 10.1016/j.mam.2017.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/04/2017] [Indexed: 02/07/2023]
|
17
|
Liu R, Heiss EH, Waltenberger B, Blažević T, Schachner D, Jiang B, Krystof V, Liu W, Schwaiger S, Peña-Rodríguez LM, Breuss JM, Stuppner H, Dirsch VM, Atanasov AG. Constituents of Mediterranean Spices Counteracting Vascular Smooth Muscle Cell Proliferation: Identification and Characterization of Rosmarinic Acid Methyl Ester as a Novel Inhibitor. Mol Nutr Food Res 2018; 62:e1700860. [PMID: 29405576 DOI: 10.1002/mnfr.201700860] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/23/2018] [Indexed: 12/15/2022]
Abstract
SCOPE Aberrant vascular smooth muscle cell (VSMC) proliferation is involved in atherosclerotic plaque formation and restenosis. Mediterranean spices have been reported to confer cardioprotection, but their direct influence on VSMCs has largely not been investigated. This study aims at examining rosmarinic acid (RA) and 11 related constituents for inhibition of VSMC proliferation in vitro, and at characterizing the most promising compound for their mode of action and influence on neointima formation in vivo. METHODS AND RESULTS RA, rosmarinic acid methyl ester (RAME), and caffeic acid methyl ester inhibit VSMC proliferation in a resazurin conversion assay with IC50 s of 5.79, 3.12, and 6.78 µm, respectively. RAME significantly reduced neointima formation in vivo in a mouse femoral artery cuff model. Accordingly, RAME leads to an accumulation of VSMCs in the G0 /G1 cell-cycle phase, as indicated by blunted retinoblastoma protein phosphorylation upon mitogen stimulation and inhibition of cyclin-dependent kinase 2 in vitro. CONCLUSION RAME represses PDGF-induced VSMC proliferation in vitro and reduces neointima formation in vivo. These results recommend RAME as an interesting compound with VSMC-inhibiting potential. Future metabolism and pharmacokinetics studies might help to further evaluate the potential relevance of RAME and other spice-derived polyphenolics for vasoprotection.
Collapse
MESH Headings
- Animals
- Cardiovascular Agents/adverse effects
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Cinnamates/administration & dosage
- Cinnamates/adverse effects
- Cinnamates/pharmacology
- Cinnamates/therapeutic use
- Depsides/administration & dosage
- Depsides/adverse effects
- Depsides/pharmacology
- Depsides/therapeutic use
- Diet, Mediterranean
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Human Umbilical Vein Endothelial Cells/cytology
- Humans
- Male
- Mediterranean Region
- Methylation
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Random Allocation
- Rats
- Retinoblastoma Protein/metabolism
- Rosmarinus/chemistry
- Rosmarinus/growth & development
- Spices/analysis
- Rosmarinic Acid
Collapse
Affiliation(s)
- Rongxia Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, University of Yantai, Yantai, China
| | - Elke H Heiss
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Tina Blažević
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Daniel Schachner
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Baohong Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Vladimir Krystof
- Laboratory of Growth Regulators, Palacky University & Institute of Experimental Botany AS CR, Olomouc, Czech Republic
| | - Wanhui Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, University of Yantai, Yantai, China
| | - Stefan Schwaiger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Luis M Peña-Rodríguez
- Unidad de Biotecnología, Centro de investigación Científica de Yucatán, Mérida, México
| | - Johannes M Breuss
- Center for Physiology and Pharmacology, Institute for Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Atanas G Atanasov
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
| |
Collapse
|
18
|
Folestad E, Kunath A, Wågsäter D. PDGF-C and PDGF-D signaling in vascular diseases and animal models. Mol Aspects Med 2018; 62:1-11. [PMID: 29410092 DOI: 10.1016/j.mam.2018.01.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/14/2017] [Accepted: 01/22/2018] [Indexed: 01/06/2023]
Abstract
Members of the platelet-derived growth factor (PDGF) family are well known to be involved in different pathological conditions. The cellular and molecular mechanisms induced by the PDGF signaling have been well studied. Nevertheless, there is much more to discover about their functions and some important questions to be answered. This review summarizes the known roles of two of the PDGFs, PDGF-C and PDGF-D, in vascular diseases. There are clear implications for these growth factors in several vascular diseases, such as atherosclerosis and stroke. The PDGF receptors are broadly expressed in the cardiovascular system in cells such as fibroblasts, smooth muscle cells and pericytes. Altered expression of the receptors and the ligands have been found in various cardiovascular diseases and current studies have shown important implications of PDGF-C and PDGF-D signaling in fibrosis, neovascularization, atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Erika Folestad
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anne Kunath
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Dick Wågsäter
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
19
|
Wang T, Chen K, Hsu P, Lin H, Wang Y, Chen C, Liao Y, Juo SH. microRNA let-7g suppresses PDGF-induced conversion of vascular smooth muscle cell into the synthetic phenotype. J Cell Mol Med 2017; 21:3592-3601. [PMID: 28699690 PMCID: PMC5706591 DOI: 10.1111/jcmm.13269] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 05/02/2017] [Indexed: 01/12/2023] Open
Abstract
Platelet-derived growth factor (PDGF) can promote vascular smooth muscle cells (VSMCs) to switch from the quiescent contractile phenotype to synthetic phenotype, which contributes to atherosclerosis. We aimed to investigate the role of microRNA let-7g in phenotypic switching. Bioinformatics prediction was used to find let-7g target genes in the PDGF/mitogen-activated protein kinase kinase kinase 1 (MEKK1)/extracellular signal-regulated kinase (ERK)/Krüppel-like factor-4 (KLF4) signalling pathway that affects VSMC phenotypic switching. The luciferase reporter assay and let-7g transfection were used to confirm let-7g target genes. Two contractile proteins alpha-smooth muscle actin (α-SMA) and calponin were VSMC-specific genes and were measured as the indicators for VSMC phenotype. Lentivirus carrying the let-7g gene was injected to apolipoprotein E knockout (apoE-/- ) mice to confirm let-7g's effect on preventing atherosclerosis. Through the PDGF/MEKK1/ERK/KLF4 signalling pathway, PDGF-BB can inhibit α-SMA and calponin. The PDGFB and MEKK1 genes were predicted to harbour let-7g binding sites, which were confirmed by our reporter assays. Transfection of let-7g to VSMC also reduced PDGFB and MEKK1 levels. Moreover, we showed that let-7g decreased phosphorylated-ERK1/2 while had no effect on total ERK1/2. KLF4 can reduce VSMC-specific gene expression by preventing myocardin-serum response factor (SRF) complex from associating with these gene promoters. The immunoprecipitation assay showed that let-7g decreased the interaction between KLF4 and SRF. Further experiments demonstrated that let-7g can increase α-SMA and calponin levels to maintain VSMC in the contractile status. Injection of lentivirus carrying let-7g gene increased let-7g's levels in aorta and significantly decreased atherosclerotic plaques in the apoE-/- mice. We demonstrated that let-7g reduces the PDGF/MEKK1/ERK/KLF4 signalling to maintain VSMC in the contractile status, which further reduce VSMC atherosclerotic change.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Aorta/metabolism
- Aorta/pathology
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Becaplermin
- Binding Sites
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Gene Expression Regulation
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- MAP Kinase Kinase Kinase 1/genetics
- MAP Kinase Kinase Kinase 1/metabolism
- Mice
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Protein Binding
- Proto-Oncogene Proteins c-sis/genetics
- Proto-Oncogene Proteins c-sis/metabolism
- Signal Transduction
- Transfection
- Calponins
Collapse
Affiliation(s)
- Tzu‐Ming Wang
- Department of Medical ResearchChina Medical University HospitalTaichungTaiwan
| | - Ku‐Chung Chen
- Department of Biochemistry and Molecular Cell BiologySchool of MedicineCollege of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Po‐Yuan Hsu
- Department of Medical ResearchChina Medical University HospitalTaichungTaiwan
| | - Hsiu‐Fen Lin
- Department of NeurologyKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yung‐Song Wang
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
- Institute of Fisheries ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Chien‐Yuan Chen
- Department of Medical ResearchChina Medical University HospitalTaichungTaiwan
- Graduate Institute of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yi‐Chu Liao
- Department of NeurologyNational Yang‐Ming University School of MedicineTaipeiTaiwan
- Department of NeurologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Suh‐Hang H. Juo
- Department of Medical ResearchChina Medical University HospitalTaichungTaiwan
- Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichungTaiwan
- Brain disease research centerChina Medical UniversityTaichungTaiwan
| |
Collapse
|
20
|
Vignini A, Sabbatinelli J, Clemente N, Delli Carpini G, Tassetti M, Zagaglia G, Ciavattini A. Preperitoneal Fat Thicknesses, Lipid Profile, and Oxidative Status in Women With Uterine Fibroids. Reprod Sci 2017; 24:1419-1425. [PMID: 28122481 DOI: 10.1177/1933719116689598] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE There is growing evidence supporting a possible role for metabolic syndrome and its determinants, such as dyslipidemia, in uterine fibroid (UF) pathogenesis. The present study aims to investigate the association between UFs and visceral and subcutaneous fat thickness (SFT), lipid profile, and oxidative and antioxidative status. METHODS In this cross-sectional study, 35 patients diagnosed with UFs and 15 women without UFs were enrolled. Clinical history and anthropometric parameters were collected for every woman. Characteristics of UFs, preperitoneal fat thickness (PFT), and SFT were assessed ultrasonically. Lipid profile, glucose, thiobarbituric acid reactive substances (TBARs), and superoxide dismutase (SOD) activity were evaluated on plasma from participants. RESULTS Women with UFs showed a significantly increased PFT (11.63 ± 3.39 vs 7.01 ± 3.10 mm; P < .001), lower levels of high-density lipoprotein cholesterol (HDL-C; 45.4 ± 8.3 vs 57.2 ± 13.4 mg/dL; P = .017), higher levels of low-density lipoprotein cholesterol (LDL-C; 92.3 ± 21.5 vs 72.0 ± 14.6 mg/dL; P = .007), and oxidized LDL (65.2 ± 20.7 vs 43.0 ± 11.3 U/L; P = .002). In patients, TBARs concentration was significantly higher (9.41 ± 6.49 vs 2.92 ± 1.65 nmol malondialdehyde/100 μg prot; P < .001), whereas SOD activity was lower (1.09 ± 0.19 vs 1.37 ± 0.41 U/μL; P = .005). Preperitoneal fat thickness was positively associated with body mass index, oxidized LDL, and TBARs. At multivariate analysis, PFT and HDL-C maintained a significant correlation with the diagnosis of UFs. CONCLUSION Chronic inflammation triggered and sustained by visceral fat could play a determinant role in cell differentiation and proliferation processes, necessary for the development of UFs. Alterations in cholesterol fractions may be explained as a consequence of the increased visceral fat deposits and can reflect an increased risk of subclinical atherosclerosis in patients with UF.
Collapse
Affiliation(s)
- Arianna Vignini
- 1 Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Jacopo Sabbatinelli
- 1 Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Nicolò Clemente
- 2 Woman's Health Sciences Department, Gynecologic Section, Università Politecnica delle Marche, Ancona, Italy
| | - Giovanni Delli Carpini
- 2 Woman's Health Sciences Department, Gynecologic Section, Università Politecnica delle Marche, Ancona, Italy
| | - Marta Tassetti
- 2 Woman's Health Sciences Department, Gynecologic Section, Università Politecnica delle Marche, Ancona, Italy
| | - Giulia Zagaglia
- 2 Woman's Health Sciences Department, Gynecologic Section, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Ciavattini
- 2 Woman's Health Sciences Department, Gynecologic Section, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
21
|
Song IS, Jeong YJ, Park JH, Shim S, Jang SW. Chebulinic acid inhibits smooth muscle cell migration by suppressing PDGF-Rβ phosphorylation and inhibiting matrix metalloproteinase-2 expression. Sci Rep 2017; 7:11797. [PMID: 28924208 PMCID: PMC5603554 DOI: 10.1038/s41598-017-12221-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/06/2017] [Indexed: 01/10/2023] Open
Abstract
Excessive migration of vascular smooth muscle cells (VSMCs) after vascular injury contributes to the development of occlusive vascular disease. Inhibition of VSMC migration is a validated therapeutic modality for occlusive vascular diseases, such as atherosclerosis and restenosis. We investigated the inhibitory effect of chebulinic acid (CBA) on cell migration and matrix metalloproteinase (MMP)-2 activation in platelet-derived growth factor (PDGF)-BB-induced mouse and human VSMCs. CBA significantly inhibited PDGF-BB-induced migration in mouse and human VSMCs, without inducing cell death. Additionally, CBA significantly blocked PDGF-BB-induced phosphorylation of the PDGF receptor (PDGF-R), Akt, and extracellular signal-regulated kinase (ERK)1/2 by inhibiting the activation of the PDGF-BB signalling pathway. In both mouse and human VSMCs, CBA inhibited PDGF-induced MMP-2 mRNA and protein expression as well as the proteolytic activity of MMP-2. Moreover, CBA suppressed sprout outgrowth formation of VSMCs from endothelium-removed aortic rings as well as neointima formation following rat carotid balloon injury. Taken together, our findings indicated that CBA inhibits VSMC migration by decreasing MMP-2 expression through PDGF-R and the ERK1/2 and Akt pathways. Our data may improve the understanding of the antiatherogenic effects of CBA in VSMCs.
Collapse
Affiliation(s)
- In-Sung Song
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 138-736, Korea.,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Korea
| | - Yu Jeong Jeong
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 138-736, Korea.,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Korea
| | - Jung-Hyun Park
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 138-736, Korea.,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Korea
| | - Sungbo Shim
- Department of Biochemistry, Neuromarker Resource Bank, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea.
| | - Sung-Wuk Jang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 138-736, Korea. .,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Korea.
| |
Collapse
|
22
|
Ouyang L, Zhang K, Chen J, Wang J, Huang H. Roles of platelet-derived growth factor in vascular calcification. J Cell Physiol 2017; 233:2804-2814. [PMID: 28467642 DOI: 10.1002/jcp.25985] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is prevalent in aging, and patients with hypertension, chronic kidney disease (CKD), or diabetes. VC is regarded as an active and complex process that involves multiple mechanisms responsible for calcium deposition in vessel wall. In light of the complicated pathogenesis of VC, effective therapy for ameliorating VC is limited. Thus, it is urgent to explore the potential mechanisms and find new targets for the therapy of VC. Platelet-derived growth factor (PDGF), a potent mitogen, and chemoattractant have been found to disturb the vascular homeostasis by inducing inflammation, oxidative stress, and phenotype transition, all of which accelerate the process of VC. The aim of current review is to present a review about the roles of PDGF in affecting VC and to establish a potential target for treating VC.
Collapse
Affiliation(s)
- Liu Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Kun Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Jie Chen
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingfeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Hui Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| |
Collapse
|
23
|
Li S, Cao C, Chen H, Song J, Lee C, Zhang J, Zhang F, Geng Q, Li Z, Li J. Atheroprotective effects of statins in patients with unstable angina by regulating the blood-borne microRNA network. Mol Med Rep 2017; 16:817-827. [PMID: 28560417 PMCID: PMC5482202 DOI: 10.3892/mmr.2017.6616] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 03/28/2017] [Indexed: 12/21/2022] Open
Abstract
Experimental studies have demonstrated several effects of statins in acute coronary syndrome (ACS) that may extend their clinical benefit beyond the lipid profile modification itself. However, the precise underlying mechanism remains to be elucidated. microRNAs (miRNAs) serve significant roles in the pathophysiology of atherosclerotic plaque progression. The present study investigated the protective role of statins in patients with unstable angina (UA) by regulating the circulating miRNA network. miRNA array results demonstrated that there were 21 differentially expressed miRNAs in non-statin-treated patients with UA (n=8) compared with non-coronary artery disease controls (n=8), and 33 differentially expressed miRNAs in statin-treated patients with UA (n=8) compared with non-statin patients. TargetScan and miRanda programs were used to predict miRNAs target genes. miRNAs target genes in vascular endothelial cells and monocytes were clustered based on the CGAP SAGE library via the Database for Annotation, Visualization and Integrated Discovery (DAVID) platform, and miRNA target genes in platelets were clustered based on a UP tissue-specific library via the DAVID platform. The PANTHER database via DAVID platform was used to perform signaling pathway analysis. The miRNA-gene/pathway network was visualized by Cytoscape software. Bioinformatic analysis suggested that statin-induced miRNAs functions were primarily enriched in angiogenesis, integrin and platelet derived growth factor signaling pathways in UA patients. In endothelial cells and platelets, statin-induced miRNAs primarily targeted the integrin signaling pathway, and in monocytes primarily targeted cytoskeletal regulation by the Rho GTPase signaling pathway. These results revealed that statins may serve systematic protective roles in UA patients by influencing the circulating miRNA regulatory network. Further studies are required to verify the functions of statin-induced miRNAs in endothelial cells, platelets and monocytes.
Collapse
Affiliation(s)
- Sufang Li
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Chengfu Cao
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Hong Chen
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Junxian Song
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Chongyou Lee
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jing Zhang
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Feng Zhang
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Qiang Geng
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Zheng Li
- Department of Cardiology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jingjin Li
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
24
|
Tóth-Zsámboki E, Horváth Z, Hajtman L, Leé S, Pállinger É, Kuklis E, Tahy Á, Fekete G, Kohut L, Kiss RG. Cardiac rehabilitation programme as a non-pharmacological platelet inhibitory tool in acute coronary syndrome survivors. Eur J Prev Cardiol 2017; 24:1148-1156. [PMID: 28438028 DOI: 10.1177/2047487317704937] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Acute coronary syndrome is associated with platelet hyperactivity, which in its persistent form, promotes recurrent thrombotic events. Complex cardiac rehabilitation after acute coronary syndrome improves clinical outcome; however, its effect on platelet hyperactivity is unknown. Design and methods We enrolled 84 acute coronary syndrome patients on dual antiplatelet therapy, who underwent a new complex cardiac rehabilitation programme (NovaCord physiotherapy, lifestyle counselling, strict diet, stress management and regular coaching) and 51 control acute coronary syndrome patients with traditional cardiac rehabilitation. Platelet functionality was determined at enrolment and at three months follow-up by aggregometry, serum platelet-derived growth factor levels, total- and platelet-derived microvesicle counts (PMV; CD41a+/CD61+, CD62P+). Results Platelet aggregation parameters and platelet-derived growth factor levels were significantly decreased in the complex cardiac rehabilitation group at three months (1 µg/ml collagen, median (interquartile range): 22 (10-45) vs 14 (7.5-25.5)%, p = 0.0015; 2 µg/ml collagen: 36 (22-60) vs 26.5 (16-37)%, p = 0.0019; 1.25 µM adenosine-diphosphate: 4.5 (1-10) vs 1 (0-3)%, p = 0.0006; 5 µM adenosine-diphosphate: 27 (16-38) vs 22 (12-31)%, p = 0.0078; epinephrine: 33 (15-57) vs 27 (12-43)%, p = 0.01; platelet-derived growth factor: 434.6 (256.0-622.7) vs 224.8 (148.5-374.1) pg/ml, p = 0.0001). In contrast, these changes were absent or did not reach statistical significance in the traditional cardiac rehabilitation group. Platelet-derived microvesicle counts were significantly decreased in both groups, while total microvesicle count was significantly reduced only in the complex cardiac rehabilitation group (median (interquartile range): 3945.5 (2138-5661) vs 1739 (780-2303) count/µl; p = 0.0001). Conclusions Platelet hyperactivity three months after acute coronary syndrome significantly decreased in patients undergoing complex cardiac rehabilitation. Besides dual antiplatelet therapy, effective management and comprehensive control of cardiovascular risk factors might represent a new, non-pharmacological approach to influence platelet functionality.
Collapse
Affiliation(s)
| | - Zsófia Horváth
- 1 Department of Cardiology, Hungarian Defence Forces, Hungary.,2 School of PhD studies, Semmelweis University, Hungary
| | - László Hajtman
- 1 Department of Cardiology, Hungarian Defence Forces, Hungary
| | - Sarolta Leé
- 1 Department of Cardiology, Hungarian Defence Forces, Hungary.,2 School of PhD studies, Semmelweis University, Hungary
| | - Éva Pállinger
- 3 Department of Genetics, Cell and Immunobiology, Semmelweis University, Hungary
| | | | - Ádám Tahy
- 4 Quintess Knowledge Center, Hungary
| | | | - László Kohut
- 5 Cardiac Rehabilitation Institute, Hungarian Defence Forces, Hungary
| | - Róbert G Kiss
- 1 Department of Cardiology, Hungarian Defence Forces, Hungary
| |
Collapse
|