1
|
Chai Q, Guo C, Li L, Cao J, Liu H, Lu Z. Association of angiogenesis-associated genes with atherosclerotic plaque progression, intraplaque hemorrhage, and immune infiltration. Heliyon 2024; 10:e32692. [PMID: 39183847 PMCID: PMC11341292 DOI: 10.1016/j.heliyon.2024.e32692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 08/27/2024] Open
Abstract
Mounting evidence suggests that intraplaque angiogenesis is associated with the progression of atherosclerotic plaques and the development of intraplaque hemorrhage. The specificity of intraplaque immune cell infiltration may be associated with abnormalities in the structure and function of the nascent capillaries. Here, we analyzed expression levels of angiogenesis-associated genes in early and advanced carotid atheromatous plaque tissues as well as in stable and intraplaque hemorrhage plaques. Expression profiles of advanced arterial plaques based on angiogenesis-associated genes were classified into subtypes by performing a consensus clustering analysis. The correlation between the immune microenvironment of plaques and expression of angiogenesis-associated genes was also explored using the single sample gene set enrichment analysis method and the CIBERSORT algorithm. We identified hub angiogenesis-associated genes showing similar expression patterns throughout plaque adverse progression, and constructed a prediction model using the random forest algorithm. Receiver operating curves were constructed to evaluate efficacy in identification of intraplaque hemorrhage in a plaque. Our results suggest that heterogeneity of angiogenesis-related genes may promote the malignant development of plaques and cause plaque rupture. In conclusion, we propose a model based on expression of angiogenesis-related genes to predict the risk of plaque rupture.
Collapse
Affiliation(s)
- Quanyou Chai
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, And the Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Chunling Guo
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Long Li
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jimin Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, And the Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Huimin Liu
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311100, China
| | - Zhaoyang Lu
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, And the Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| |
Collapse
|
2
|
Biwer LA, Man JJ, Camarda ND, Carvajal BV, Karumanchi SA, Jaffe IZ. Prior Exposure to Experimental Preeclampsia Increases Atherosclerotic Plaque Inflammation in Atherogenic Mice-Brief Report. Arterioscler Thromb Vasc Biol 2024; 44:946-953. [PMID: 38450510 PMCID: PMC10978246 DOI: 10.1161/atvbaha.123.320474] [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: 12/03/2023] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Women with a history of preeclampsia have evidence of premature atherosclerosis and increased risk of myocardial infarction and stroke compared with women who had a normotensive pregnancy. Whether this is due to common risk factors or a direct impact of prior preeclampsia exposure has never been tested in a mouse atherosclerosis model. METHODS Pregnant LDLR-KO (low-density lipoprotein receptor knockout; n=35) female mice were randomized in midgestation to sFlt1 (soluble fms-like tyrosine kinase 1)-expressing adenovirus or identical control adenovirus. Postpartum, mice were fed high-fat diet for 8 weeks to induce atherogenesis. Comparison between the control and preeclampsia models was made for metabolic parameters, atherosclerosis burden and composition by histology, plaque inflammation by flow cytometry, and aortic cytokines and inflammatory markers using a cytokine array. RESULTS In pregnant LDLR-KO mice, sFlt1 adenovirus significantly induced serum sFlt1, blood pressure, renal endotheliosis, and decreased pup viability. After 8 weeks of postpartum high fat feeding, body weight, fasting glucose, plasma cholesterol, HDL (high-density lipoprotein), and LDL (low-density lipoprotein) were not significantly different between groups with no change in aortic root plaque size, lipid content, or necrotic core area. Flow cytometry demonstrated significantly increased CD45+ aortic arch leukocytes and CD3+T cells and aortic lysate contained more CCL (CC motif chemokine ligand) 22 and fetuin A and decreased expression of IGFBP6 (insulin-like growth factor-binding protein 6) and CCL21 in preeclampsia-exposed mice compared with controls. CONCLUSIONS In atherogenic LDLR-KO mice, exposure to sFlt1-induced preeclampsia during pregnancy increases future atherosclerotic plaque inflammation, supporting the concept that preeclampsia directly exacerbates atherosclerotic inflammation independent of preexisting risk factors. This mechanism may contribute to ischemic vascular disease in women after preeclampsia pregnancy.
Collapse
Affiliation(s)
- Lauren A. Biwer
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
- Department of Comparative Medicine, Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Joshua J. Man
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
| | | | | | | | - Iris Z. Jaffe
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
| |
Collapse
|
3
|
Zhou YH, Tang YZ, Guo LY, Zheng LL, Zhang D, Yang CY, Wang W. Overexpression of sFlt-1 represses ox-LDL-induced injury of HUVECs by activating autophagy via PI3K/AKT/mTOR pathway. Microvasc Res 2021; 139:104252. [PMID: 34520772 DOI: 10.1016/j.mvr.2021.104252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/26/2021] [Accepted: 09/06/2021] [Indexed: 10/20/2022]
Abstract
Soluble fms-like tyrosine kinase-1 (sFlt-1), a circulating antiangiogenic protein, is involved in the pathogenesis of atherosclerosis (AS), and the underlying mechanism is still unclear. Here, we attempted to investigate the mechanism of action of sFlt-1 in AS. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low density lipoprotein (ox-LDL) to induce cell injury. ox-LDL treatment increased LC3-II/LC3-I ratio, Beclin-1 expression and GFP-LC3 puncta in HUVECs, suggesting that ox-LDL may induce autophagic flux impairment in HUVECs. ox-LDL-treated HUVECs displayed a decrease of sFlt-1 levels. Moreover, ox-LDL treatment reduced cell proliferation and elevated apoptosis in HUVECs, which was abrogated by sFlt-1 overexpression. Up-regulation of sFlt-1 repressed the activity of PI3K/AKT/mTOR signaling pathway and enhanced autophagy in HUVECs following ox-LDL treatment. Additionally, sFlt-1 overexpression-mediated increase of autophagy in ox-LDL-treated HUVECs was abolished by 3-methyladenine (autophagy inhibitor). 3-methyladenine abrogated the impact of sFlt-1 overexpression on proliferation and apoptosis in ox-LDL-treated HUVECs. This work confirmed that overexpression of sFlt-1 activated autophagy by repressing PI3K/Akt/mTOR signaling pathway, and thus alleviated ox-LDL-induced injury of HUVECs. Therefore, this study suggests that sFlt-1 may be a potential target for AS treatment.
Collapse
Affiliation(s)
- Yi-Hua Zhou
- Department of ICU, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Yu-Zhi Tang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Liang-Yun Guo
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Li-Li Zheng
- Department of Pharmacy, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Dan Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Can-Ying Yang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Wei Wang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
4
|
Bai T, Li M, Liu Y, Qiao Z, Wang Z. Inhibition of ferroptosis alleviates atherosclerosis through attenuating lipid peroxidation and endothelial dysfunction in mouse aortic endothelial cell. Free Radic Biol Med 2020; 160:92-102. [PMID: 32768568 DOI: 10.1016/j.freeradbiomed.2020.07.026] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/24/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022]
Abstract
Atherosclerosis (AS) is the fundamental pathological state of many serious vascular diseases, characterized by disorders of lipid metabolism. Ferroptosis is a type of regulated cell death that is mainly mediated by iron-dependent lipid peroxidation. In this study, whether ferroptosis has occurred in AS and the potential effects of ferroptosis on AS were investigated. Ferroptosis inhibitor ferrostatin-1 (Fer-1) was administered to high-fat diet (HFD)-induced AS in ApoE-/- mice. The results showed that Fer-1 could alleviate AS lesion in HFD-fed ApoE-/- mice. Additionally, Fer-1 partially inhibited the iron accumulation, lipid peroxidation and reversed the expressions of ferroptosis indicators SLC7A11 and glutathione peroxidase 4 (GPX4) in HFD-fed ApoE-/- mice. Next, we evaluated the effects of inhibition of ferroptosis on oxidized-low density lipoprotein (ox-LDL)-induced mouse aortic endothelial cells (MAECs). Results showed that Fer-1 increased cell viability and reduced cell death in ox-LDL-treated MAECs. Moreover, Fer-1 decreased iron content and lipid peroxidation and up-regulated the levels of SLC7A11 and GPX4. Additionally, Fer-1 down-regulated the expressions of adhesion molecules and up-regulated eNOS expression. Iron chelator deferoxamine was used to demonstrate ferroptosis could be partially inhibited by iron complexation in ox-LDL-treated MAECs. Our results indicated that ferroptosis might occur during the initiation and development of AS. More importantly, inhibition of ferroptosis could alleviate AS through attenuating lipid peroxidation and endothelial dysfunction in AECs. Our findings might contribute to a deeper understanding regarding the pathological process of AS and provide a therapeutic target for AS.
Collapse
Affiliation(s)
- Tao Bai
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Mingxing Li
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yuanfeng Liu
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zhentao Qiao
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zhiwei Wang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
5
|
Genetic variants of VEGFR-1 gene promoter in acute myocardial infarction. Hum Genomics 2019; 13:56. [PMID: 31744542 PMCID: PMC6862733 DOI: 10.1186/s40246-019-0243-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Coronary artery disease (CAD) including acute myocardial infarction (AMI) is a common complex disease caused by atherosclerosis. Vascular epithelial growth factor receptor-1 (VEGFR-1) stimulates angiogenesis and vascular permeability, and functions as a decoy to sequester VEGF and prevent initiation of intracellular signaling. VEGFR-1 knockout mice exhibit significantly higher mortality due to heart failure, cardiac hypertrophy, and cardiac dysfunction. An evident increase in macrophage infiltration and cardiac fibrosis are also observed after transverse aortic constriction. Therefore, VEGFR-1 gene variants may be involved in CAD. In this study, VEGFR-1 gene promoter was genetically and functionally analyzed in large cohorts of AMI patients and ethnic-matched controls. RESULTS A total of 16 DNA sequence variants (DSVs) including six single-nucleotide polymorphisms (SNPs) were found in the VEGFR-1 gene promoter and 5'-untranslated region. Five novel DSVs and one SNP were only identified in AMI patients group. These DSVs and SNP significantly altered the transcriptional activity of the VEGFR-1 gene promoter in both HEK-293 and H9c2 cells (P < 0.05). Further electrophoretic mobility shift assay indicated that the DSVs and SNPs evidently affected the binding of transcription factors. CONCLUSIONS The genetic variants in VEGFR-1 gene identified in AMI patients may alter the transcriptional activity of the VEGFR-1 gene promoter and change VEGFR-1 level, contributing to AMI development.
Collapse
|
6
|
VEGF-A/VEGFR-2 and FGF-2/FGFR-1 but not PDGF-BB/PDGFR-β play important roles in promoting immature and inflammatory intraplaque angiogenesis. PLoS One 2018; 13:e0201395. [PMID: 30125282 PMCID: PMC6101364 DOI: 10.1371/journal.pone.0201395] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022] Open
Abstract
Various angiogenic factors have been shown to play important roles in intraplaque angiogenesis, while little is known about the dynamic expression change and interplay between various angiogenic factors and intraplaque angiogenesis under high cholesterol conditions. New Zealand rabbits underwent balloon injury of the abdominal artery and then were assigned to a control group (n = 15, normal chow) or high cholesterol group (n = 25, 1% high cholesterol diet). At weeks 4, 6, 8, 10, and 12 after acclimation, rabbits (high cholesterol group, n = 5; control group, n = 3) were euthanized. No lesions were observed in the control group. From week 4 to week 12, the expression of vascular endothelial growth factor A (VEGF-A), VEGF receptor 2 (VEGFR-2), fibroblast growth factor 2 (FGF-2), FGF receptor 1 (FGFR-1), platelet-derived growth factor-BB (PDGF-BB), and tumor necrosis factor alpha (TNF-α), the vulnerability index (VI) and the microvessel density (MVD) were significantly elevated in the high cholesterol group; however, PDGF receptor β (PDGFR-β) expression showed little change. Analysis by double-label immunofluorescence (CD31 and Ng2) and FITC-dextran indicated that the neovessels within the plaque were leaky due to a lack of pericytes. As indicated by Pearson’s correlation analysis, there was a highly positive correlation between the VI, MVD, macrophage content, and TNF-α level, and the levels of VEGF-A/VEGFR-2 and FGF-2/FGFR-1. However, no correlations were observed between PDGFR-β levels and the VI or MVD. High expression of VEGF-A/VEGFR-2 and FGF-2/FGFR-1 but not of PDGF-BB/PDGFR-β may contribute to immature and inflammatory intraplaque angiogenesis and plaque instability in a rabbit model of atherosclerosis.
Collapse
|
7
|
Chen R, Huang Z, Wang J, Chen X, Fu Y, Wang W. Silent Information Regulator 1 Negatively Regulates Atherosclerotic Angiogenesis via Mammalian Target of Rapamycin Complex 1 Signaling Pathway. Am J Med Sci 2018; 356:168-176. [PMID: 30219159 DOI: 10.1016/j.amjms.2018.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND This study aimed to investigate the interactions between silent information regulator 1 (SIRT1) and mammalian target of rapamycin (mTOR) in intraplaque angiogenesis and their potential mechanisms through in vivo and in vitro studies. METHODS An atherosclerosis model was established in 12 rabbits on a high-cholesterol diet. The rabbits were equally divided into 3 groups: a control group (high-lipid diet), RAP group (high-lipid diet supplemented with rapamycin) and RAP + NAM group (high-lipid diet supplemented with rapamycin and nicotinamide). At the end of 4 weeks, the area of plaques in the aorta was determined and the protein expression of CD31 and vascular endothelial growth factor (VEGF) was detected through hematoxylin and eosin staining and immunohistochemical staining, respectively. For in vitro study, a hypoxia model was established in human umbilical vein endothelial cells (HUVECs) by using the chemical method (CoCl2). The MTT assay, scratch assay and tube formation assay were performed to evaluate the proliferation and angiogenesis abilities of HUVECs. Reverse transcription polymerase chain reaction was used to examine the mRNA levels of SIRT1, hypoxia-inducible factor-1α (HIF-1α), mTOR and p70 ribosomal S6 kinase (p70S6K). Western blotting was used to examine the protein levels of SIRT1, HIF-1α, mTOR, p-mTOR, p-raptor and p-p70S6K. RESULTS The results of the in vivo study indicated a significant inhibitory effect of rapamycin on plaque size and intraplaque angiogenesis (0.05 ± 0.02mm2 versus 5.44 ± 0.50mm2, P < 0.05). This effect was attenuated by nicotinamide (0.76 ± 0.15mm2 versus 0.05 ± 0.02mm2, P < 0.05). Compared with the RAP group, CD31- and VEGF-positive vessels were abundant in the RAP + NAM group. The RAP group showed lower expression of p-mTOR, p-p70S6K and HIF-1α than did the control group (P < 0.05), whereas the RAP + NAM group showed slightly higher expression of these factors than did the RAP group (P < 0.05). Furthermore, in vitro studies revealed that the inhibitory effect of rapamycin on the angiogenic ability of HUVECs and its significant inhibitory effects on the protein level of HIF-1α and the phosphorylation of proteins involved in the mTORC1 pathway, including mTOR, raptor and p70S6K (P < 0.05), were enhanced by cotreatment with SRT1720 and rapamycin (P < 0.05). In contrast to mTOR and SIRT1, the mRNA levels of p70S6K and HIF-1α were reduced by rapamycin (P < 0.05) and further reduced by cotreatment with SRT1720 and rapamycin. CONCLUSIONS The study results indicate that SIRT1 might negatively regulate atherosclerotic angiogenesis via mTORC1 and HIF-1α signaling pathway and cointervention of SIRT1 and mTOR may serve as a crucial therapeutic strategy in cardiovascular medicine.
Collapse
Affiliation(s)
- Runtai Chen
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Zhenchun Huang
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Junyi Wang
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Xiaoying Chen
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Yucai Fu
- Laboratory of Cell Senescence, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Wei Wang
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China.
| |
Collapse
|
8
|
de Jager SCA, Meeuwsen JAL, van Pijpen FM, Zoet GA, Barendrecht AD, Franx A, Pasterkamp G, van Rijn BB, Goumans MJ, den Ruijter HM. Preeclampsia and coronary plaque erosion: Manifestations of endothelial dysfunction resulting in cardiovascular events in women. Eur J Pharmacol 2017; 816:129-137. [PMID: 28899695 DOI: 10.1016/j.ejphar.2017.09.012] [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: 05/22/2017] [Revised: 08/31/2017] [Accepted: 09/08/2017] [Indexed: 12/29/2022]
Abstract
Atherosclerosis is the major underlying pathology of cardiovascular disease (CVD). The risk for CVD is increased in women with a history of preeclampsia. Multiple studies have indicated that accelerated atherosclerosis underlies this increased CVD risk. Furthermore, it has been suggested that endothelial dysfunction and inflammation play an important role in the increased CVD risk of women with preeclampsia. Rupture or erosion of atherosclerotic plaques can induce the formation of thrombi that underlie the onset of acute clinical CVD such as myocardial infarction and stroke. In relatively young women, cardiovascular events are mainly due to plaque erosions. Eroded plaques have a distinct morphology compared to ruptured plaques, but have been understudied as a substrate for CVD. The currently available evidence points towards lesions with features of stability such as high collagen content and smooth muscle cells and with distinct mechanisms that further promote the pro-thrombotic environment such as Toll Like Receptor (TLR) signaling and endothelial apoptosis. These suggested mechanisms, that point to endothelial dysfunction and intimal thickening, may also play a role in preeclampsia. Pregnancy is considered a stress test for the cardiovascular system with preeclampsia as an additional pathological substrate for earlier manifestation of vascular disease. This review provides a summary of the possible common mechanisms involved in preeclampsia and accelerated atherosclerosis in young females and highlights plaque erosion as a likely substrate for CVD events in women with a history of preeclampsia.
Collapse
Affiliation(s)
- Saskia C A de Jager
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, The Netherlands.
| | - John A L Meeuwsen
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, The Netherlands
| | - Freeke M van Pijpen
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, The Netherlands
| | - Gerbrand A Zoet
- Wilhelmina Children's Hospital Birth Centre, Division of Woman and Baby, University Medical Center Utrecht, The Netherlands
| | - Arjan D Barendrecht
- Laboratory of Clinical Chemistry and Haematology, University Medical Center Utrecht, The Netherlands
| | - Arie Franx
- Wilhelmina Children's Hospital Birth Centre, Division of Woman and Baby, University Medical Center Utrecht, The Netherlands
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry and Haematology, University Medical Center Utrecht, The Netherlands
| | - Bas B van Rijn
- Wilhelmina Children's Hospital Birth Centre, Division of Woman and Baby, University Medical Center Utrecht, The Netherlands; Academic Unit of Human Development and Health, Institute for Life Sciences, University of Southampton, United Kingdom
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, The Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
9
|
Abstract
In this article, we introduce briefly several strategies for preventing atherosclerotic cardiovascular disease and promoting healthcare for non-communicable diseases (NCDs). These novel strategies include four core elements of health – sleep, emotion, exercise, and diet – and consist of SEED intervention (SEEDi) and E(e)SEEDi due to supplementation of the environment as a core element, and Hu's healthy lifestyles intervention (HHLi) which originates from E(e)SEED-BasED healthy lifestyles. They are suitable for the early evaluation of risk factors, and play a key role in the prevention and management of human NCDs when combined with the RT-ABCDEF strategy and the Grade 210 prevention, which include obesity-OSA-hypertension syndrome and C-type hypertension, especially in halting cardiovascular, diabetes and cancer (CDC) strips we first discovered. After successful clinical practice, we may expect our novel strategies for controlling these chronic diseases according to the conception of mass prevention and treatment.
Collapse
|
10
|
Parma L, Baganha F, Quax PHA, de Vries MR. Plaque angiogenesis and intraplaque hemorrhage in atherosclerosis. Eur J Pharmacol 2017; 816:107-115. [PMID: 28435093 DOI: 10.1016/j.ejphar.2017.04.028] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/31/2017] [Accepted: 04/20/2017] [Indexed: 12/15/2022]
Abstract
Acute cardiovascular events, due to rupture or erosion of an atherosclerotic plaque, represent the major cause of morbidity and mortality in patients. Growing evidence suggests that plaque neovascularization is an important contributor to plaque growth and instability. The vessels' immaturity, with profound structural and functional abnormalities, leads to recurrent intraplaque hemorrhage. This review discusses new insights of atherosclerotic neovascularization, including the effects of leaky neovessels on intraplaque hemorrhage, both in experimental models and humans. Furthermore, modalities for in vivo imaging and therapeutic interventions to target plaque angiogenesis will be discussed.
Collapse
Affiliation(s)
- Laura Parma
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Fabiana Baganha
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Paul H A Quax
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - Margreet R de Vries
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
11
|
Cystathionine γ-lyase is expressed in human atherosclerotic plaque microvessels and is involved in micro-angiogenesis. Sci Rep 2016; 6:34608. [PMID: 27708362 PMCID: PMC5052587 DOI: 10.1038/srep34608] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 05/13/2016] [Indexed: 01/09/2023] Open
Abstract
Atherosclerotic plaques are classically divided into stable and vulnerable plaques. Vulnerable plaques are prone to rupture with a risk for infarction. High intraplaque microvessel density predisposes to plaque vulnerability. Hydrogen sulfide (H2S) is a proangiogenic gasotransmitter which is endogenously produced by cystathionine γ-lyase (CSE), and is believed to have vasculoprotective effects. However, due to its proangiogenic effects, H2S may result in pathological angiogenesis in atherosclerotic plaques, thereby increasing plaque vulnerability. The aim of this study was to determine CSE expression pattern in atherosclerotic plaques, and investigate whether CSE is involved in micro-angiogenesis in vitro. Endarterectomy plaques were studied for CSE expression, and the role of CSE in micro-angiogenesis was studied in vitro. CSE is expressed in plaques with similar levels in both stable and vulnerable plaques. CSE co-localized with von Willebrand Factor-positive microvessel endothelial cells and alpha-smooth-muscle actin-positive SMCs. In vitro, inhibition of CSE in HMEC-1 reduced tube formation, cell viability/proliferation, and migration which was restored after culture in the presence of H2S donor GYY4137. CSE is expressed in intraplaque microvessels, and H2S is a stimulator of micro-angiogenesis in vitro. Due to this pro-angiogenic effect, high levels of CSE in atherosclerotic plaques may be a potential risk for plaque vulnerability.
Collapse
|
12
|
Seno A, Takeda Y, Matsui M, Okuda A, Nakano T, Nakada Y, Kumazawa T, Nakagawa H, Nishida T, Onoue K, Somekawa S, Watanabe M, Kawata H, Kawakami R, Okura H, Uemura S, Saito Y. Suppressed Production of Soluble Fms-Like Tyrosine Kinase-1 Contributes to Myocardial Remodeling and Heart Failure. Hypertension 2016; 68:678-87. [PMID: 27480835 DOI: 10.1161/hypertensionaha.116.07371] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/26/2016] [Indexed: 01/17/2023]
Abstract
Soluble fms-like tyrosine kinase-1 (sFlt-1), an endogenous inhibitor of vascular endothelial growth factor and placental growth factor, is involved in the pathogenesis of cardiovascular disease. However, the significance of sFlt-1 in heart failure has not been fully elucidated. We found that sFlt-1 is decreased in renal failure and serves as a key molecule in atherosclerosis. In this study, we aimed to investigate the role of the decreased sFlt-1 production in heart failure, using sFlt-1 knockout mice. sFlt-1 knockout mice and wild-type mice were subjected to transverse aortic constriction and evaluated after 7 days. The sFlt-1 knockout mice had significantly higher mortality (52% versus 15%; P=0.0002) attributable to heart failure and showed greater cardiac hypertrophy (heart weight to body weight ratio, 8.95±0.45 mg/g in sFlt-1 knockout mice versus 6.60±0.32 mg/g in wild-type mice; P<0.0001) and cardiac dysfunction, which was accompanied by a significant increase in macrophage infiltration and cardiac fibrosis, than wild-type mice after transverse aortic constriction. An anti-placental growth factor-neutralizing antibody prevented pressure overload-induced cardiac hypertrophy, fibrosis, and cardiac dysfunction. Moreover, monocyte chemoattractant protein-1 expression was significantly increased in the hypertrophied hearts of sFlt-1 knockout mice compared with wild-type mice. Monocyte chemoattractant protein-1 inhibition with neutralizing antibody ameliorated maladaptive cardiac remodeling in sFlt-1 knockout mice after transverse aortic constriction. In conclusion, decreased sFlt-1 production plays a key role in the aggravation of cardiac hypertrophy and heart failure through upregulation of monocyte chemoattractant protein-1 expression in pressure-overloaded heart.
Collapse
Affiliation(s)
- Ayako Seno
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Yukiji Takeda
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Masaru Matsui
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Aya Okuda
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Tomoya Nakano
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Yasuki Nakada
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Takuya Kumazawa
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Hitoshi Nakagawa
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Taku Nishida
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Kenji Onoue
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Satoshi Somekawa
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Makoto Watanabe
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Hiroyuki Kawata
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Rika Kawakami
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Hiroyuki Okura
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Shiro Uemura
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Yoshihiko Saito
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.).
| |
Collapse
|
13
|
The therapeutic effect of bevacizumab on plaque neovascularization in a rabbit model of atherosclerosis during contrast-enhanced ultrasonography. Sci Rep 2016; 6:30417. [PMID: 27452862 PMCID: PMC4958919 DOI: 10.1038/srep30417] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/05/2016] [Indexed: 01/06/2023] Open
Abstract
The purpose of the study was to assess the therapeutic effect of the angiogenesis inhibitor bevacizumab on plaques of various stages in rabbit models using contrast-enhanced ultrasonography (CEUS). Abdominal aortic atherosclerosis was induced in 55 rabbits. Thirty-six randomly selected rabbits were divided into 2 groups according to the timing of the bevacizumab injection: an early-stage plaque group (Group ESP) and a later-stage plaque group (Group LSP). The remainder were considered the control group. Standard ultrasonography and CEUS imaging of the abdominal aorta were performed. The animals were euthanized after CEUS, and plaque specimens were harvested for histological staining of CD31. The control group exhibited a substantially higher enhanced intensity, a higher ratio of enhanced intensity in the plaque to that in the lumen, and an increased number of CD31-positive microvessels in the plaque sections than Groups ESP and LSP (P < 0.05 for all). A higher enhanced intensity (P = 0.044), a higher ratio of enhanced intensity in the plaque to that in the lumen (P = 0.023) and more CD31-positive microvessels in the plaque sections (P = 0.006) were found in Group LSP than in Group ESP. Bevacizumab demonstrated more advanced inhibition of neovascularization in early-stage plaques in rabbits.
Collapse
|
14
|
McKittrick CM, Kennedy S, Oldroyd KG, McGinty S, McCormick C. Modelling the Impact of Atherosclerosis on Drug Release and Distribution from Coronary Stents. Ann Biomed Eng 2016; 44:477-87. [PMID: 26384667 PMCID: PMC4764635 DOI: 10.1007/s10439-015-1456-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/09/2015] [Indexed: 11/24/2022]
Abstract
Although drug-eluting stents (DES) are now widely used for the treatment of coronary heart disease, there remains considerable scope for the development of enhanced designs which address some of the limitations of existing devices. The drug release profile is a key element governing the overall performance of DES. The use of in vitro, in vivo, ex vivo, in silico and mathematical models has enhanced understanding of the factors which govern drug uptake and distribution from DES. Such work has identified the physical phenomena determining the transport of drug from the stent and through tissue, and has highlighted the importance of stent coatings and drug physical properties to this process. However, there is limited information regarding the precise role that the atherosclerotic lesion has in determining the uptake and distribution of drug. In this review, we start by discussing the various models that have been used in this research area, highlighting the different types of information they can provide. We then go on to describe more recent methods that incorporate the impact of atherosclerotic lesions.
Collapse
Affiliation(s)
- C M McKittrick
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, UK
| | - S Kennedy
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - K G Oldroyd
- West of Scotland Region Heart and Lung Centre, Golden Jubilee National Hospital, Dunbartonshire, UK
| | - S McGinty
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, UK
| | - C McCormick
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, UK.
| |
Collapse
|
15
|
Abstract
Injury of arterial endothelium by abnormal shear stress and other insults induces migration and proliferation of vascular smooth muscle cells (VSMCs), which in turn leads to intimal thickening, hypoxia, and vasa vasorum angiogenesis. The resultant new blood vessels extend from the tunica media into the outer intima, allowing blood-borne oxidized low-density lipoprotein (oxLDL) particles to accumulate in outer intimal tissues by extravasation through local capillaries. In response to oxLDL accumulation, monocytes infiltrate into arterial wall tissues, where they differentiate into macrophages and subsequently evolve into foam cells by uptaking large quantities of oxLDL particles, the latter process being stimulated by hypoxia. Increased oxygen demand due to expanding macrophage and foam cell populations contributes to persistent hypoxia in plaque lesions, whereas hypoxia further promotes plaque growth by stimulating angiogenesis, monocyte infiltration, and oxLDL uptake into macrophages. Molecularly, the accumulation of hypoxia-inducible factor (HIF)-1α and the expression of its target genes mediate many of the hypoxia-induced processes during plaque initiation and growth. It is hoped that further understanding of the underlying mechanisms may lead to novel therapies for effective intervention of atherosclerosis.
Collapse
Affiliation(s)
- Guo-Hua Fong
- Center for Vascular Biology and Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06030, USA,
| |
Collapse
|
16
|
van Hinsbergh VWM, Eringa EC, Daemen MJAP. Neovascularization of the atherosclerotic plaque: interplay between atherosclerotic lesion, adventitia-derived microvessels and perivascular fat. Curr Opin Lipidol 2015; 26:405-11. [PMID: 26241102 DOI: 10.1097/mol.0000000000000210] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Neovascularization is a prominent feature in advanced human atherosclerotic plaques. This review surveys recent evidence for and remaining uncertainties regarding a role of neovascularization in atherosclerotic plaque progression. Specific emphasis is given to hypoxia, angiogenesis inhibition, and perivascular adipose tissue (PVAT). RECENT FINDINGS Immunohistochemical and imaging studies showed a strong association between hypoxia, inflammation and neovascularization, and the progression of the atherosclerotic plaque both in humans and mice. Whereas in humans, a profound invasion of microvessels from the adventitia into the plaque occurs, neovascularization in mice is found mainly (peri)adventitially. Influencing neovascularization in mice affected plaque progression, possibly by improving vessel perfusion, but supportive clinical data are not available. Whereas plaque neovascularization contributes to monocyte/macrophage accumulation in the plaque, lymphangiogenesis may facilitate egress of cells and waste products. A specific role for PVAT and its secreted factors is anticipated and wait further clinical evaluation. SUMMARY Hypoxia, inflammation, and plaque neovascularization are associated with plaque progression as underpinned by recent imaging data in humans. Recent studies provide new insights into modulation of adventitia-associated angiogenesis, PVAT, and plaque development in mice, but there is still a need for detailed information on modulating human plaque vascularization in patients.
Collapse
Affiliation(s)
- Victor W M van Hinsbergh
- aLaboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center bDepartment of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
17
|
Wu WK, Georgiadis A, Copland DA, Liyanage S, Luhmann UFO, Robbie SJ, Liu J, Wu J, Bainbridge JW, Bates DO, Ali RR, Nicholson LB, Dick AD. IL-4 regulates specific Arg-1(+) macrophage sFlt-1-mediated inhibition of angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2324-35. [PMID: 26079814 DOI: 10.1016/j.ajpath.2015.04.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/17/2015] [Accepted: 04/23/2015] [Indexed: 12/14/2022]
Abstract
One of the main drivers for neovascularization in age-related macular degeneration is activation of innate immunity in the presence of macrophages. Here, we demonstrate that T helper cell type 2 cytokines and, in particular, IL-4 condition human and murine monocyte phenotype toward Arg-1(+), and their subsequent behavior limits angiogenesis by increasing soluble fms-like tyrosine kinase 1 (sFlt-1) gene expression. We document that T helper cell type 2 cytokine-conditioned murine macrophages neutralize vascular endothelial growth factor-mediated endothelial cell proliferation (human umbilical vein endothelial cell and choroidal vasculature) in a sFlt-1-dependent manner. We demonstrate that in vivo intravitreal administration of IL-4 attenuates laser-induced choroidal neovascularization (L-CNV) due to specific IL-4 conditioning of macrophages. IL-4 induces the expression of sFlt-1 by resident CD11b(+) retinal microglia and infiltrating myeloid cells but not from retinal pigment epithelium. IL-4-induced suppression of L-CNV is not prevented when sFlt-1 expression is attenuated in retinal pigment epithelium. IL-4-mediated suppression of L-CNV was abrogated in IL-4R-deficient mice and in bone marrow chimeras reconstituted with myeloid cells that had undergone lentiviral-mediated shRNA silencing of sFlt-1, demonstrating the critical role of this cell population. Together, these data establish how lL-4 directly drives macrophage sFlt-1 production expressing an Arg-1(+) phenotype and support the therapeutic potential of targeted IL-4 conditioning within the tissue to regulate disease conditions such as neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Wei-Kang Wu
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | | | - David A Copland
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Sidath Liyanage
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Ulrich F O Luhmann
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Scott J Robbie
- Institute of Ophthalmology, University College London, London, United Kingdom; National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, United Kingdom
| | - Jian Liu
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Jiahui Wu
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - James W Bainbridge
- Institute of Ophthalmology, University College London, London, United Kingdom; National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, United Kingdom
| | - David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Queen's Medical Centre, The University of Nottingham, Nottingham, United Kingdom
| | - Robin R Ali
- Institute of Ophthalmology, University College London, London, United Kingdom; National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, United Kingdom
| | - Lindsay B Nicholson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom; School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Andrew D Dick
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom; Institute of Ophthalmology, University College London, London, United Kingdom; School of Clinical Sciences, University of Bristol, Bristol, United Kingdom; National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, United Kingdom.
| |
Collapse
|
18
|
Gishti O, Jaddoe VW, Felix JF, Reiss I, Hofman A, Ikram MK, Steegers EA, Gaillard R. Influence of Maternal Angiogenic Factors During Pregnancy on Microvascular Structure in School-Age Children. Hypertension 2015; 65:722-8. [DOI: 10.1161/hypertensionaha.114.05008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Olta Gishti
- From the Generation R Study Group (O.G., V.W.V.J., J.F.F., R.G.), and Departments of Pediatrics (O.G., V.W.V.J., I.R., R.G.), Epidemiology (O.G., V.W.V.J., J.F.F., A.H., R.G.), Ophthalmology (M.K.I.), Obstetrics and Gynecology (E.A.P.S.), and Neonatology (I.R.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Singapore Eye Research Institute and Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore (M.K.I.); and Memory Aging & Cognition
| | - Vincent W.V. Jaddoe
- From the Generation R Study Group (O.G., V.W.V.J., J.F.F., R.G.), and Departments of Pediatrics (O.G., V.W.V.J., I.R., R.G.), Epidemiology (O.G., V.W.V.J., J.F.F., A.H., R.G.), Ophthalmology (M.K.I.), Obstetrics and Gynecology (E.A.P.S.), and Neonatology (I.R.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Singapore Eye Research Institute and Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore (M.K.I.); and Memory Aging & Cognition
| | - Janine F. Felix
- From the Generation R Study Group (O.G., V.W.V.J., J.F.F., R.G.), and Departments of Pediatrics (O.G., V.W.V.J., I.R., R.G.), Epidemiology (O.G., V.W.V.J., J.F.F., A.H., R.G.), Ophthalmology (M.K.I.), Obstetrics and Gynecology (E.A.P.S.), and Neonatology (I.R.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Singapore Eye Research Institute and Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore (M.K.I.); and Memory Aging & Cognition
| | - Irwin Reiss
- From the Generation R Study Group (O.G., V.W.V.J., J.F.F., R.G.), and Departments of Pediatrics (O.G., V.W.V.J., I.R., R.G.), Epidemiology (O.G., V.W.V.J., J.F.F., A.H., R.G.), Ophthalmology (M.K.I.), Obstetrics and Gynecology (E.A.P.S.), and Neonatology (I.R.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Singapore Eye Research Institute and Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore (M.K.I.); and Memory Aging & Cognition
| | - Albert Hofman
- From the Generation R Study Group (O.G., V.W.V.J., J.F.F., R.G.), and Departments of Pediatrics (O.G., V.W.V.J., I.R., R.G.), Epidemiology (O.G., V.W.V.J., J.F.F., A.H., R.G.), Ophthalmology (M.K.I.), Obstetrics and Gynecology (E.A.P.S.), and Neonatology (I.R.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Singapore Eye Research Institute and Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore (M.K.I.); and Memory Aging & Cognition
| | - Mohammad Kamran Ikram
- From the Generation R Study Group (O.G., V.W.V.J., J.F.F., R.G.), and Departments of Pediatrics (O.G., V.W.V.J., I.R., R.G.), Epidemiology (O.G., V.W.V.J., J.F.F., A.H., R.G.), Ophthalmology (M.K.I.), Obstetrics and Gynecology (E.A.P.S.), and Neonatology (I.R.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Singapore Eye Research Institute and Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore (M.K.I.); and Memory Aging & Cognition
| | - Eric A.P. Steegers
- From the Generation R Study Group (O.G., V.W.V.J., J.F.F., R.G.), and Departments of Pediatrics (O.G., V.W.V.J., I.R., R.G.), Epidemiology (O.G., V.W.V.J., J.F.F., A.H., R.G.), Ophthalmology (M.K.I.), Obstetrics and Gynecology (E.A.P.S.), and Neonatology (I.R.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Singapore Eye Research Institute and Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore (M.K.I.); and Memory Aging & Cognition
| | - Romy Gaillard
- From the Generation R Study Group (O.G., V.W.V.J., J.F.F., R.G.), and Departments of Pediatrics (O.G., V.W.V.J., I.R., R.G.), Epidemiology (O.G., V.W.V.J., J.F.F., A.H., R.G.), Ophthalmology (M.K.I.), Obstetrics and Gynecology (E.A.P.S.), and Neonatology (I.R.), Erasmus MC, University Medical Center, Rotterdam, the Netherlands; Singapore Eye Research Institute and Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore (M.K.I.); and Memory Aging & Cognition
| |
Collapse
|
19
|
Biological mechanisms of microvessel formation in advanced atherosclerosis: The big Five. Trends Cardiovasc Med 2013; 23:153-64. [DOI: 10.1016/j.tcm.2012.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 10/08/2012] [Accepted: 10/10/2012] [Indexed: 12/19/2022]
|
20
|
Vascular endothelial growth factors (VEGFs) and stroke. Cell Mol Life Sci 2013; 70:1753-61. [PMID: 23475070 DOI: 10.1007/s00018-013-1282-8] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/17/2022]
Abstract
Vascular endothelial growth factors (VEGFs) have been shown to participate in atherosclerosis, arteriogenesis, cerebral edema, neuroprotection, neurogenesis, angiogenesis, postischemic brain and vessel repair, and the effects of transplanted stem cells in experimental stroke. Most of these actions involve VEGF-A and the VEGFR-2 receptor, but VEGF-B, placental growth factor, and VEGFR-1 have been implicated in some cases as well. VEGF signaling pathways represent important potential targets for the acute and chronic treatment of stroke.
Collapse
|
21
|
Pre-eclampsia and offspring cardiovascular health: mechanistic insights from experimental studies. Clin Sci (Lond) 2012; 123:53-72. [PMID: 22455350 PMCID: PMC3315178 DOI: 10.1042/cs20110627] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Pre-eclampsia is increasingly recognized as more than an isolated disease of pregnancy. Women who have had a pregnancy complicated by pre-eclampsia have a 4-fold increased risk of later cardiovascular disease. Intriguingly, the offspring of affected pregnancies also have an increased risk of higher blood pressure and almost double the risk of stroke in later life. Experimental approaches to identify the key features of pre-eclampsia responsible for this programming of offspring cardiovascular health, or the key biological pathways modified in the offspring, have the potential to highlight novel targets for early primary prevention strategies. As pre-eclampsia occurs in 2–5% of all pregnancies, the findings are relevant to the current healthcare of up to 3 million people in the U.K. and 15 million people in the U.S.A. In the present paper, we review the current literature that concerns potential mechanisms for adverse cardiovascular programming in offspring exposed to pre-eclampsia, considering two major areas of investigation: first, experimental models that mimic features of the in utero environment characteristic of pre-eclampsia, and secondly, how, in humans, offspring cardiovascular phenotype is altered after exposure to pre-eclampsia. We compare and contrast the findings from these two bodies of work to develop insights into the likely key pathways of relevance. The present review and analysis highlights the pivotal role of long-term changes in vascular function and identifies areas of growing interest, specifically, response to hypoxia, immune modification, epigenetics and the anti-angiogenic in utero milieu.
Collapse
|
22
|
Theilade S, Lajer M, Jorsal A, Tarnow L, Parving HH, Rossing P. Evaluation of placental growth factor and soluble Fms-like tyrosine kinase 1 as predictors of all-cause and cardiovascular mortality in patients with Type 1 diabetes with and without diabetic nephropathy. Diabet Med 2012; 29:337-44. [PMID: 21988672 DOI: 10.1111/j.1464-5491.2011.03482.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS Placental growth factor is a vascular endothelial growth factor involved in angiogenesis, vascular inflammation and plaque formation. Soluble Fms-like tyrosine kinase 1 is a decoy receptor for placental growth factor, reducing its activity. The aim of this study is to evaluate the predictive value of placental growth factor and soluble Fms-like tyrosine kinase 1 in relation to all-cause and cardiovascular mortality and decline in kidney function in Type 1 diabetes. METHODS This was a prospective, observational follow-up study with 8 (0-13) years [median (range)] of follow-up, including patients with Type 1 diabetes, of whom 458 had diabetic nephropathy [278 men; age 42 ± 11 years (mean ± sd), diabetes duration 28 ± 9 years, glomerular filtration rate 76 ± 33 ml min(-1) 1.73 m(-2) ] and 442 had long-standing normoalbuminuria (234 men; age 45 ± 12 years, diabetes duration 28 ± 10 years). RESULTS Placental growth factor and soluble Fms-like tyrosine kinase 1 levels measured at baseline were higher in patients with diabetic nephropathy compared with patients with long-standing normoalbuminuria [median (range)] 15 (4-131) vs. 11 (7-64) ng/l, (P < 0.001) and 86 (42-3462) vs. 77 (43-1557) ng/l (P < 0.001), respectively. In patients with diabetic nephropathy, high levels of placental growth factor predicted all-cause and cardiovascular mortality [hazard ratio 1.94 (1.16-3.24) and hazard ratio 2.91 (1.45-5.85)] after adjustment for sex, age, smoking, systolic blood pressure, HbA(1c) , cholesterol, glomerular filtration rate and previous cardiovascular disease. High levels of placental growth factor predicted increased risk of end-stage renal disease [hazard ratio 2.77 (1.47-5.14)], but covariate adjustments attenuated the association [hazard ratio 1.89 (0.91-3.95)]. Among patients with long-standing normoalbuminuria, placental growth factor levels predicted fatal and non-fatal cardiovascular events [hazard ratio 1.97 (1.03-3.76)], but not all-cause mortality. Baseline soluble Fms-like tyrosine kinase 1 levels did not predict outcome in either group after adjustment. CONCLUSION Placental growth factor is elevated in patients with Type 1 diabetes and diabetic nephropathy and predicts all-cause and cardiovascular mortality, but not deterioration of kidney function.
Collapse
Affiliation(s)
- S Theilade
- Steno Diabetes Center, Gentofte, Denmark.
| | | | | | | | | | | |
Collapse
|
23
|
Vuorio T, Jauhiainen S, Ylä-Herttuala S. Pro- and anti-angiogenic therapy and atherosclerosis with special emphasis on vascular endothelial growth factors. Expert Opin Biol Ther 2011; 12:79-92. [DOI: 10.1517/14712598.2012.641011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|