1
|
Yang J, Yang L, Wang Y, Huai L, Shi B, Zhang D, Xu W, Cui D. Interleukin-6 related signaling pathways as the intersection between chronic diseases and sepsis. Mol Med 2025; 31:34. [PMID: 39891057 PMCID: PMC11783753 DOI: 10.1186/s10020-025-01089-6] [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: 08/28/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Sepsis is associated with immune dysregulated and organ dysfunction due to severe infection. Clinicians aim to restore organ function, rather than prevent diseases that are prone to sepsis, resulting in high mortality and a heavy public health burden. Some chronic diseases can induce sepsis through inflammation cascade reaction and Cytokine Storm (CS). Interleukin (IL)-6, the core of CS, and its related signaling pathways have been considered as contributors to sepsis. Therefore, it is important to study the relationship between IL-6 and its related pathways in sepsis-related chronic diseases. This review generalized the mechanism of sepsis-related chronic diseases via IL-6 related pathways with the purpose to take rational management for these diseases. IL-6 related signaling pathways were sought in Kyoto Encyclopedia of Genes and Genomes (KEGG), and retrieved protein-protein interaction in the Search for Interaction Genes tool (STRING). In PubMed and Google Scholar, the studies were searched out, which correlating to IL-6 related pathways and associating with the pathological process of sepsis. Focused on the interactions of sepsis and IL-6 related pathways, some chronic diseases have been studied for association with sepsis, containing insulin resistance, Alcoholic liver disease (ALD), Alzheimer disease (AD), and atherosclerosis. This article summarized the inflammatory mechanisms of IL-6 cross-talked with other mediators of some chronic diseases in vitro, animal models, and human experiments, leading to the activation of pathways and accelerating the progression of sepsis. The clinicians should be highlight to this kind of diseases and more clinical trials are needed to provide more reliable theoretical basis for health policy formulation.
Collapse
Affiliation(s)
- Jie Yang
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China.
| | - Lin Yang
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Yanjiao Wang
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Lu Huai
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Bohan Shi
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Di Zhang
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Wei Xu
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Di Cui
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| |
Collapse
|
2
|
Zeng Y, Fu BM. Angiogenesis and Microvascular Permeability. Cold Spring Harb Perspect Med 2025; 15:a041163. [PMID: 38692737 PMCID: PMC11694756 DOI: 10.1101/cshperspect.a041163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Angiogenesis, the formation of new blood microvessels, is a necessary physiological process for tissue generation and repair. Sufficient blood supply to the tissue is dependent on microvascular density, while the material exchange between the circulating blood and the surrounding tissue is controlled by microvascular permeability. We thus begin this article by reviewing the key signaling factors, particularly vascular endothelial growth factor (VEGF), which regulates both angiogenesis and microvascular permeability. We then review the role of angiogenesis in tissue growth (bone regeneration) and wound healing. Finally, we review angiogenesis as a pathological process in tumorigenesis, intraplaque hemorrhage, cerebral microhemorrhage, pulmonary fibrosis, and hepatic fibrosis. Since the glycocalyx is important for both angiogenesis and microvascular permeability, we highlight the role of the glycocalyx in regulating the interaction between tumor cells and endothelial cells (ECs) and VEGF-containing exosome release and uptake by tumor-associated ECs, all of which contribute to tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Bingmei M Fu
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York 10031, USA
| |
Collapse
|
3
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [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: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
4
|
Han J, Ye L, Wang Y. Pyroptosis: An Accomplice in the Induction of Multisystem Complications Triggered by Obstructive Sleep Apnea. Biomolecules 2024; 14:1349. [PMID: 39595526 PMCID: PMC11592050 DOI: 10.3390/biom14111349] [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: 07/25/2024] [Revised: 10/05/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Obstructive sleep apnea (OSA) is a common respiratory disorder, primarily characterized by two pathological features: chronic intermittent hypoxia (CIH) and sleep deprivation (SD). OSA has been identified as a risk factor for numerous diseases, and the inflammatory response related to programmed cell necrosis is believed to play a significant role in the occurrence and progression of multisystem damage induced by OSA, with increasing attention being paid to pyroptosis. Recent studies have indicated that OSA can elevate oxidative stress levels in the body, activating the process of pyroptosis within different tissues, ultimately accelerating organ dysfunction. However, the molecular mechanisms of pyroptosis in the multisystem damage induced by OSA remain unclear. Therefore, this review focuses on four major systems that have received concentrated attention in existing research in order to explore the role of pyroptosis in promoting renal diseases, cardiovascular diseases, neurocognitive diseases, and skin diseases in OSA patients. Furthermore, we provide a comprehensive overview of methods for inhibiting pyroptosis at different molecular levels, with the goal of identifying viable targets and therapeutic strategies for addressing OSA-related complications.
Collapse
Affiliation(s)
- Jingwen Han
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, China; (J.H.); (L.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Lisong Ye
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, China; (J.H.); (L.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Yan Wang
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
- Department of Preventive Dentistry, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, China
| |
Collapse
|
5
|
Nasir I, McGuinness C, Poh AR, Ernst M, Darcy PK, Britt KL. Tumor macrophage functional heterogeneity can inform the development of novel cancer therapies. Trends Immunol 2023; 44:971-985. [PMID: 37995659 DOI: 10.1016/j.it.2023.10.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/25/2023]
Abstract
Macrophages represent a key component of the tumor microenvironment (TME) and are largely associated with poor prognosis. Therapeutic targeting of macrophages has historically focused on inhibiting their recruitment or reprogramming their phenotype from a protumor (M2-like) to an antitumor (M1-like) one. Unfortunately, this approach has not provided clinical breakthroughs that have changed practice. Emerging studies utilizing single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics have improved our understanding of the ontogeny, phenotype, and functional plasticity of macrophages. Overlaying the wealth of current information regarding macrophage molecular subtypes and functions has also identified novel therapeutic vulnerabilities that might drive better control of tumor-associated macrophages (TAMs). Here, we discuss the functional profiling of macrophages and provide an update of novel macrophage-targeted therapies in development.
Collapse
Affiliation(s)
- Ibraheem Nasir
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Conor McGuinness
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia; La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia; La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Phillip K Darcy
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia; Cancer Immunology Research Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Kara L Britt
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia.
| |
Collapse
|
6
|
Arief Waskito B, Sargowo D, Kalsum U, Tjokroprawiro A. Anti-atherosclerotic activity of aqueous extract of Ipomoea batatas (L.) leaves in high-fat diet-induced atherosclerosis model rats. J Basic Clin Physiol Pharmacol 2023; 34:725-734. [PMID: 34986543 DOI: 10.1515/jbcpp-2021-0080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Cardiovascular diseases, especially atherosclerosis, are the leading cause of human mortality in Indonesia. Ipomoea batatas (L.) is a food plant used in Indonesian traditional medicine to treat cardiovascular diseases and related conditions. We assessed the anti-atherosclerotic activity of the aqueous extract of I. batatas leaves in a rat model of high-fat diet-induced atherosclerosis and its mechanism. METHODS The presence of amino acid content in the I. batatas L. purple variant was determined by liquid chromatography high-resolution mass spectrometry (LC-HRMS). Thirty male Wistar rats were divided into five groups (n=6/group), i.e., standard diet group (SD), high-fat diet group (HF), and HF plus I. batatas L. extracts orally (625; 1,250; or 2,500 mg/kg) groups. The numbers of macrophages and aortic wall thickness were analyzed histologically. Immunohistochemical analyses were performed to assess foam cells-oxidized low-density lipoprotein (oxLDL), endothelial nitric oxide synthase (eNOS), and vascular endothelial growth factor (VEGF) expression in the aorta. RESULTS LC-HRMS analysis showed nine amino acid content were identified from I. batatas L. In vivo study revealed that oral administration of I. batatas L. leaf extract alleviated foam cells-oxLDL formation and aortic wall thickness caused by high-fat diet atherosclerosis rats. Further, I. batatas L. leaf extract promoted the number of macrophages and modulated VEGF and eNOS expression in the aorta. CONCLUSIONS I. batatas L. leaf extract shows a positive anti-atherosclerosis effect. Furthermore, the mechanism may promote the macrophages, eNOS, VEGF expressions, and inhibition of foam cells-oxLDL formation and aortic wall thickness with the best dosage at 2,500 mg/kg. This could represent a novel approach to prevent cardiovascular diseases.
Collapse
Affiliation(s)
- Budi Arief Waskito
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, Indonesia
- Department of Internal Medicine, Faculty of Medicine, Wijaya Kusuma University, Surabaya, East Java, Indonesia
| | - Djanggan Sargowo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Brawijaya, Dr. Saiful Anwar General Hospital, Malang, East Java, Indonesia
| | - Umi Kalsum
- Department of Pharmacology, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, Indonesia
| | - Askandar Tjokroprawiro
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo Hospital, Surabaya, East Java, Indonesia
| |
Collapse
|
7
|
Boswell-Patterson CA, Hétu MF, Pang SC, Herr JE, Zhou J, Jain S, Bambokian A, Johri AM. Novel theranostic approaches to neovascularized atherosclerotic plaques. Atherosclerosis 2023; 374:1-10. [PMID: 37149970 DOI: 10.1016/j.atherosclerosis.2023.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/05/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023]
Abstract
As the global burden of atherosclerotic cardiovascular disease continues to rise, there is an increased demand for improved imaging techniques for earlier detection of atherosclerotic plaques and new therapeutic targets. Plaque lesions, vulnerable to rupture and thrombosis, are thought to be responsible for the majority of cardiovascular events, and are characterized by a large lipid core, a thin fibrous cap, and neovascularization. In addition to supplying the plaque core with increased inflammatory factors, these pathological neovessels are tortuous and leaky, further increasing the risk of intraplaque hemorrhage. Clinically, plaque neovascularization has been shown to be a significant and independent predictor of adverse cardiovascular outcomes. Microvessels can be detected through contrast-enhanced ultrasound (CEUS) imaging, however, clinical assessment in vivo is generally limited to qualitative measures of plaque neovascularization. There is no validated standard for quantitative assessment of the microvessel networks found in plaques. Advances in our understanding of the pathological mechanisms underlying plaque neovascularization and its significant role in the morbidity and mortality associated with atherosclerosis have made it an attractive area of research in translational medicine. Current areas of research include the development of novel therapeutic and diagnostic agents to target plaque neovascularization stabilization. With recent progress in nanotechnology, nanoparticles have been investigated for their ability to specifically target neovascularization. Contrast microbubbles have been similarly engineered to carry loads of therapeutic agents and can be visualized using CEUS. This review summarizes the pathogenesis, diagnosis, clinical significance of neovascularization, and importantly the emerging areas of theranostic tool development.
Collapse
Affiliation(s)
| | - Marie-France Hétu
- Department of Medicine, Cardiovascular Imaging Network at Queen's (CINQ), Queen's University, Canada
| | - Stephen C Pang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Julia E Herr
- Department of Medicine, Cardiovascular Imaging Network at Queen's (CINQ), Queen's University, Canada
| | - Jianhua Zhou
- Department of Biomedical Engineering, Sun Yat-sen University, Guangzhou, China
| | - Shagun Jain
- Department of Medicine, Cardiovascular Imaging Network at Queen's (CINQ), Queen's University, Canada
| | - Alexander Bambokian
- Department of Medicine, Cardiovascular Imaging Network at Queen's (CINQ), Queen's University, Canada
| | - Amer M Johri
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada; Department of Medicine, Cardiovascular Imaging Network at Queen's (CINQ), Queen's University, Canada.
| |
Collapse
|
8
|
Díaz-García E, Sanz-Rubio D, García-Tovar S, Alfaro E, Cubero P, Gil AV, Marin JM, Cubillos-Zapata C, García-Río F. Inflammasome activation mediated by oxidised low-density lipoprotein in patients with sleep apnoea and early subclinical atherosclerosis. Eur Respir J 2023; 61:13993003.01401-2022. [PMID: 36517180 DOI: 10.1183/13993003.01401-2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/20/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Atherosclerosis is a common comorbidity of obstructive sleep apnoea (OSA) patients, caused by the interaction of dyslipidaemia and systemic inflammation. The OSA pro-inflammatory response is mediated by NLRP3 inflammasome activation, which requires a priming signal mediated by intermittent hypoxia (IH) and an activation signal provided by soluble stimulus present in plasma. Our objectives were to study oxidised low-density lipoprotein (oxLDL) expression in OSA patients with or without early subclinical atherosclerosis (eSA) as well as its contribution to NLRP3 activation and tissue factor (TF) release. METHODS We analysed oxLDL, key components of the NLRP3 inflammasome cascade and TF in plasma and monocytes from OSA patients and non-apnoeic subjects, with or without eSA as determined by increased carotid intima-media thickness without the appearance of atherosclerotic plaques. The oxLDL contribution to NLRP3 inflammasome activation was assessed using in vitro models. RESULTS High levels of oxLDL were identified in plasma from OSA patients, particularly in those with eSA, as well as an overexpression of NLRP3 cascade components and TF. Furthermore, in vitro models showed that both oxLDL and plasma from OSA patients with eSA act synergistically with IH as a priming and activation signal of NLRP3 that enhances the inflammatory response, pyroptosis and TF release. CONCLUSIONS OSA patients with eSA exhibit NLRP3 activation by IH and the presence of oxLDL capable of releasing TF, constituting a pathway for the interaction between dyslipidaemia and systemic inflammation in the development of atherosclerotic lesions.
Collapse
Affiliation(s)
- Elena Díaz-García
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - David Sanz-Rubio
- Translational Research Unit, Miguel Servet University Hospital - IISAragon, Zaragoza, Spain
| | - Sara García-Tovar
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Enrique Alfaro
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Pablo Cubero
- Translational Research Unit, Miguel Servet University Hospital - IISAragon, Zaragoza, Spain
| | - Ana V Gil
- Translational Research Unit, Miguel Servet University Hospital - IISAragon, Zaragoza, Spain
| | - José M Marin
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Translational Research Unit, Miguel Servet University Hospital - IISAragon, Zaragoza, Spain
- Department of Medicine, University of Zaragoza School of Medicine, Zaragoza, Spain
| | - Carolina Cubillos-Zapata
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- C. Cubillos-Zapata and F. García-Río contributed equally to this article as lead authors and supervised the work
| | - Francisco García-Río
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
- C. Cubillos-Zapata and F. García-Río contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
9
|
CXCR6 Mediates Pressure Overload-Induced Aortic Stiffness by Increasing Macrophage Recruitment and Reducing Exosome-miRNA29b. J Cardiovasc Transl Res 2022; 16:271-286. [PMID: 36018423 DOI: 10.1007/s12265-022-10304-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
Abstract
Aortic stiffness is an independent risk factor for aortic diseases such as aortic dissection which commonly occurred with aging and hypertension. Chemokine receptor CXCR6 is critically involved in vascular inflammation and remodeling. Here, we investigated whether and how CXCR6 plays a role in aortic stiffness caused by pressure overload. CXCR6-/- and WT mice underwent transverse aortic constriction (TAC) surgery for 8 weeks. CXCR6 deficiency significantly improved TAC-induced aortic remodeling and endothelial dysfunction by decreasing CD11c+ macrophage infiltration, suppressing VCAM-1 and ICAM-1, reducing collagen deposition, and downregulating MMP12 and osteopontin in the aorta. Consistently, blocking the CXCL16/CXCR6 axis also reduced aortic accumulation of CD11c+ macrophages and vascular stiffness but without affecting the release of TNF-α and IL-6 from the aorta. Furthermore, pressure overload inhibited aortic release of exosomes, which could be reversed by suppressing CXCR6 or CXCL16. Inhibition of exosome release by GW4869 significantly aggravated TAC-induced aortic calcification and stiffness. By exosomal microRNA microarray analysis, we found that microRNA-29b was significantly reduced in aortic endothelial cells (AECs) receiving TAC. Intriguingly, blocking the CXCL16/CXCR6 axis restored the expression of miR-29b in AECs. Finally, overexpression of miR-29b significantly increased eNOS and reduced MMPs and collagen in AECs. By contrast, antagonizing miR-29b in vivo further enhanced TAC-induced expressions of MMP12 and osteopontin, aggravated aortic fibrosis, calcification, and stiffness. Our study demonstrated a key role of the CXCL16/CXCR6 axis in macrophage recruitment and macrophage-mediated aortic stiffness under pressure overload through an exosome-miRNAs-dependent manner.
Collapse
|
10
|
Cimmino G, Di Serafino L, Cirillo P. Pathophysiology and mechanisms of Acute Coronary Syndromes: atherothrombosis, immune-inflammation, and beyond. Expert Rev Cardiovasc Ther 2022; 20:351-362. [PMID: 35510629 DOI: 10.1080/14779072.2022.2074836] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/04/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The pathophysiology of atherosclerosis and its acute complications, such as the Acute Coronary Syndromes (ACS), is continuously under investigation. Immunity and inflammation seem to play a pivotal role in promoting formation and grow of atherosclerotic plaques. At the same time, plaque rupture followed by both platelets' activation and coagulation cascade induction lead to intracoronary thrombus formation. Although these phenomena might be considered responsible of about 90% of ACS, in up to 5-10% of acute syndromes, a non-obstructive coronary artery disease (MINOCA) might be documented. This paper gives an overview on atherothrombosis and immuno-inflammation processes involved in ACS pathophysiology, also emphasizing the pathological mechanisms potentially involved in MINOCA. AREAS COVERED The relationship between immuno-inflammation and atherothrombosis is continuously updated by recent findings. At the same time, pathophysiology of MINOCA still remains a partially unexplored field, stimulating the research of potential links between these two aspects of ACS pathophysiology. EXPERT OPINION Pathophysiology of ACS has been extensively investigated; however, several gray areas still remain. MINOCA represents one of these areas. At the same time, many aspects of immune-inflammation processes are still unknown. Thus, research should be continued to shed a brighter light on both these sides of "ACS" moon.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luigi Di Serafino
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Thomas C, Leleu D, Masson D. Cholesterol and HIF-1α: Dangerous Liaisons in Atherosclerosis. Front Immunol 2022; 13:868958. [PMID: 35386720 PMCID: PMC8977597 DOI: 10.3389/fimmu.2022.868958] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
HIF-1α exerts both detrimental and beneficial actions in atherosclerosis. While there is evidence that HIF-1α could be pro-atherogenic within the atheromatous plaque, experimental models of atherosclerosis suggest a more complex role that depends on the cell type expressing HIF-1α. In atheroma plaques, HIF-1α is stabilized by local hypoxic conditions and by the lipid microenvironment. Macrophage exposure to oxidized LDLs (oxLDLs) or to necrotic plaque debris enriched with oxysterols induces HIF-1α -dependent pathways. Moreover, HIF-1α is involved in many oxLDL-induced effects in macrophages including inflammatory response, angiogenesis and metabolic reprogramming. OxLDLs activate toll-like receptor signaling pathways to promote HIF-1α stabilization. OxLDLs and oxysterols also induce NADPH oxidases and reactive oxygen species production, which subsequently leads to HIF-1α stabilization. Finally, recent investigations revealed that the activation of liver X receptor, an oxysterol nuclear receptor, results in an increase in HIF-1α transcriptional activity. Reciprocally, HIF-1α signaling promotes triglycerides and cholesterol accumulation in macrophages. Hypoxia and HIF-1α increase the uptake of oxLDLs, promote cholesterol and triglyceride synthesis and decrease cholesterol efflux. In conclusion, the impact of HIF-1α on cholesterol homeostasis within macrophages and the feedback activation of the inflammatory response by oxysterols via HIF-1α could play a deleterious role in atherosclerosis. In this context, studies aimed at understanding the specific mechanisms leading to HIF-1α activation within the plaque represents a promising field for research investigations and a path toward development of novel therapies.
Collapse
Affiliation(s)
- Charles Thomas
- Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,LipSTIC LabEx, Dijon, France
| | - Damien Leleu
- Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,LipSTIC LabEx, Dijon, France.,CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, Dijon, France
| | - David Masson
- Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,LipSTIC LabEx, Dijon, France.,CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, Dijon, France
| |
Collapse
|
12
|
Reactive Oxygen Species and Oxidative Stress in Vascular-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7906091. [PMID: 35419169 PMCID: PMC9001081 DOI: 10.1155/2022/7906091] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022]
Abstract
Oxidative stress (OS) refers to the enhancement of oxidation and the decreased of related antioxidant enzymes activity under pathological conditions, resulting in relatively excess reactive oxygen species (ROS), causing cytotoxicity, which leads to tissue damage and is linked to neurodegenerative diseases, cardiovascular diseases, diabetes, cancers, and many other pathologies. As an important intracellular signaling molecule, ROS can regulate numerous physiological actions, such as vascular reactivity and neuronal function. According to several studies, the uncontrolled production of ROS is related to vascular injury. The growing evidence revealing how traditional risk factors translate into ROS and lead to vasculitis and other vascular diseases. In this review, we sought to mainly discuss the role of ROS and antioxidant mechanisms in vascular-related diseases, especially cardiovascular and common macrovascular diseases.
Collapse
|
13
|
Plicosepalus acacia Extract and Its Major Constituents, Methyl Gallate and Quercetin, Potentiate Therapeutic Angiogenesis in Diabetic Hind Limb Ischemia: HPTLC Quantification and LC-MS/MS Metabolic Profiling. Antioxidants (Basel) 2021; 10:antiox10111701. [PMID: 34829572 PMCID: PMC8614836 DOI: 10.3390/antiox10111701] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/23/2022] Open
Abstract
Plicosepalus acacia (Fam. Loranthaceae) has been reported to possess hypoglycemic, antioxidant, antimicrobial, and anti-inflammatory effects. Liquid chromatography combined with tandem mass spectrometry (LC-MS/MS) analysis revealed the presence of a high content of polyphenolic compounds that are attributed to the therapeutic effects of the crude extract. In addition, methyl gallate and quercetin were detected as major phytomedicinal agents at concentrations of 1.7% and 0.062 g%, respectively, using high-performance thin layer chromatography (HPTLC). The present study investigated the effect of the P. acacia extract and its isolated compounds, methyl gallate and quercetin, on hind limb ischemia induced in type 1 diabetic rats. Histopathological examination revealed that treatment with P. acacia extract, methyl gallate, and quercetin decreased degenerative changes and inflammation in the ischemic muscle. Further biochemical assessment of the hind limb tissue showed decreased oxidative stress, increased levels of nitric oxide and endothelial nitric oxide synthase (eNOS), and enhancement of the levels of heme oxygenase-1 (HO-1) and vascular endothelial growth factor (VEGF) in the groups treated with methyl gallate and quercetin. Expression levels of hypoxia inducible factor-1 alpha (HIF-1α), VEGF, fibroblast growth factor-2 (FGF-2), and miR-146a were upregulated in the muscle tissue of methyl gallate- and quercetin-treated groups along with downregulation of nuclear factor kappa B (NF-κB). In conclusion, P. acacia extract and its isolated compounds, methyl gallate and quercetin, mediated therapeutic angiogenesis in diabetic hind limb ischemia.
Collapse
|
14
|
Liang P, Mao L, Ma Y, Ren W, Yang S. A systematic review on Zhilong Huoxue Tongyu capsule in treating cardiovascular and cerebrovascular diseases: Pharmacological actions, molecular mechanisms and clinical outcomes. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114234. [PMID: 34044079 DOI: 10.1016/j.jep.2021.114234] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/29/2021] [Accepted: 05/20/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiovascular and cerebrovascular diseases have become a severe threat for human health worldwide, however, optimal therapeutic options are still developed. Zhilong Huoxue Tongyu capsule (ZL capsule) is mainly composed of Astragalus membranaceus, Leech, Earthworm, Cinnamomum cassia and Sargentodoxa cuneata, having functions of replenishing qi and activating blood, dispelling wind and reducing phlegm. It is an expanded application on the basis of traditional uses of above TCMs, acquiring a satisfactory curative effect on cardiovascular and cerebrovascular diseases over twenty years. AIM OF THE STUDY To comprehensively summarize the main components of ZL capsule, understand the mechanisms of ZL capsule, and conclude clinical regimens of ZL capsule for cardiovascular and cerebrovascular diseases. MATERIALS AND METHODS We selected network pharmacology technology to analyze main active compounds and predict underlying mechanism of ZL capsule against atherosclerosis. Molecular docking was performed to simulate the interaction pattern between the active components of ZL capsule and putative targets. Further, PubMed, Web of Science, China National Knowledge Infrastructure and Google Scholar were used to search literatures, with the key words of "Zhilong Huoxue Tongyu capsule", "cardiovascular and cerebrovascular diseases", "atherosclerosis", "clinical study" and their combinations, mainly from 2000 to 2020. RESULTS Both network pharmacology analysis, molecular docking and animal experiments studies confirmed that mechanisms of ZL capsule plays the role of anti-inflammatory, anti-apoptosis and promoting angiogenesis in treating cardiovascular and cerebrovascular diseases by multi-components acting on multi-targets via multi-pathways. Over 1000 clinical cases were benefited from the treatment of ZL capsule, suggesting a holistic concept of "the same therapy for different myocardial and cerebral diseases". CONCLUSIONS For the first time, this systematic review may supply meaningful information for further studies to explore material basis and pharmacodynamics of ZL capsule and also provide a basis for sharing the "Chinese patent medicine" for cardiovascular and cerebrovascular diseases.
Collapse
Affiliation(s)
- Pan Liang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Drug Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Linshen Mao
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Drug Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yue Ma
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Drug Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Drug Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Drug Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
15
|
Knutson AK, Williams AL, Boisvert WA, Shohet RV. HIF in the heart: development, metabolism, ischemia, and atherosclerosis. J Clin Invest 2021; 131:137557. [PMID: 34623330 DOI: 10.1172/jci137557] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The heart forms early in development and delivers oxygenated blood to the rest of the embryo. After birth, the heart requires kilograms of ATP each day to support contractility for the circulation. Cardiac metabolism is omnivorous, utilizing multiple substrates and metabolic pathways to produce this energy. Cardiac development, metabolic tuning, and the response to ischemia are all regulated in part by the hypoxia-inducible factors (HIFs), central components of essential signaling pathways that respond to hypoxia. Here we review the actions of HIF1, HIF2, and HIF3 in the heart, from their roles in development and metabolism to their activity in regeneration and preconditioning strategies. We also discuss recent work on the role of HIFs in atherosclerosis, the precipitating cause of myocardial ischemia and the leading cause of death in the developed world.
Collapse
|
16
|
Sluiter TJ, van Buul JD, Huveneers S, Quax PHA, de Vries MR. Endothelial Barrier Function and Leukocyte Transmigration in Atherosclerosis. Biomedicines 2021; 9:328. [PMID: 33804952 PMCID: PMC8063931 DOI: 10.3390/biomedicines9040328] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelium is a highly specialized barrier that controls passage of fluids and migration of cells from the lumen into the vessel wall. Endothelial cells assist leukocytes to extravasate and despite the variety in the specific mechanisms utilized by different leukocytes to cross different vascular beds, there is a general principle of capture, rolling, slow rolling, arrest, crawling, and ultimately diapedesis via a paracellular or transcellular route. In atherosclerosis, the barrier function of the endothelium is impaired leading to uncontrolled leukocyte extravasation and vascular leakage. This is also observed in the neovessels that grow into the atherosclerotic plaque leading to intraplaque hemorrhage and plaque destabilization. This review focuses on the vascular endothelial barrier function and the interaction between endothelial cells and leukocytes during transmigration. We will discuss the role of endothelial dysfunction, transendothelial migration of leukocytes and plaque angiogenesis in atherosclerosis.
Collapse
Affiliation(s)
- Thijs J. Sluiter
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jaap D. van Buul
- Sanquin Research and Landsteiner Laboratory, Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Paul H. A. Quax
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Margreet R. de Vries
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
17
|
Wu T, Dang KR, Wang YF, Lyu BZ, Xu WQ, Dou GR, Zhou J, Hui YN, Du HJ. A modified laser-induced choroidal neovascularization animal model with intravitreal oxidized low-density lipoprotein. Int J Ophthalmol 2020; 13:1187-1194. [PMID: 32821671 DOI: 10.18240/ijo.2020.08.03] [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: 12/18/2019] [Accepted: 05/08/2020] [Indexed: 10/23/2022] Open
Abstract
AIM To investigate whether intravitreal injection of oxidized low-density lipoprotein (OxLDL) can promote laser-induced choroidal neovascularization (CNV) formation in mice and the mechanism involved, thereby to develop a better animal model. METHODS C57BL6/J mice were randomized into three groups. Immediately after CNV induction with 532 nm laser photocoagulation, 1.0 µL of OxLDL [100 µg/mL in phosphate-buffered saline (PBS)] was intravitreally injected, whereas PBS and the same volume low-density lipoprotein (LDL; 100 µg/mL in PBS) were injected into the vitreous as controls. Angiogenic and inflammatory cytokines were measured by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting (WB) after 5d, and CNV severity was analyzed by choroid flat mount and immunofluorescence staining after 1wk. In vitro, retinal pigment epithelial (RPE) cell line (ARPE19) were treated with OxLDL (LDL as control) for 8h. Angiogenic and inflammatory cytokine levels were measured. A specific inhibitor of lectin-like oxidized low-density lipoprotein receptor 1 (LOX1) was used to evaluate the role of LOX1 in this process. RESULTS At 7d after intravitreal injection of 1 µL (100 µg/mL) OxLDL, T15-labeled OxLDL was mainly deposited around the CNV area, and the F4/80-labeled macrophages, the CD31-labeled vascular endothelial cells number and CNV area were increased. Meanwhile, WB and qRT-PCR results showed that vascular endothelial growth factor (VEGF), CC chemokine receptor 2 (CCR2), interleukin-6 (IL-6), IL-1β, and matrix metalloproteinase 9 (MMP9) expressions were increased, which was supported by in vitro experiments in RPE cells. LOX1 inhibitors significantly reduced expressions of inflammatory factors IL-1β and VEGF. CONCLUSION A modified laser-induced CNV animal model is established with intravitreal injection of 1 µL (100 µg/mL) of OxLDL at 7d, which at least partially through LOX1. This animal model can be used as a simple model for studying the role of OxLDL in age-related macular degeneration.
Collapse
Affiliation(s)
- Tong Wu
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Kuan-Rong Dang
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Ya-Fen Wang
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Bao-Zhen Lyu
- Department of Anatomy and Histology and Embryology, Xi'an Health School, Xi'an 710032, Shaanxi Province, China
| | - Wen-Qin Xu
- The Orbital Disease Institute of the Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Guo-Rui Dou
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Jian Zhou
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Yan-Nian Hui
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Hong-Jun Du
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
18
|
Li HY, Yuan Y, Fu YH, Wang Y, Gao XY. Hypoxia-inducible factor-1α: A promising therapeutic target for vasculopathy in diabetic retinopathy. Pharmacol Res 2020; 159:104924. [PMID: 32464323 DOI: 10.1016/j.phrs.2020.104924] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 05/04/2020] [Accepted: 05/10/2020] [Indexed: 02/07/2023]
Abstract
Diabetic retinopathy (DR) is a serious condition that can cause blindness in diabetic patients. It is a neurovascular disease, but the pathogenesis leading to the onset of this disease is still not completely understood. However, hypoxia with subsequent neovascularization is a characteristic phenomenon observed with DR. Cellular response to hypoxia is mediated by the transcriptional regulator hypoxia-inducible factor (HIF). Long-term research has shown that one isotype of HIF, HIF-1α, may play a pivotal role under hypoxic conditions, and an increasing number of studies have shown that HIF-1α and its target genes contribute to retinal neovascularization. Therefore, targeting HIF-1α may lead to more effective DR treatments. This review describes the possible mechanisms of HIF-1α in neovascularization of DR. Furthermore, various inhibitors of HIF-1α that may have viable potential in the treatment of DR are also discussed.
Collapse
Affiliation(s)
- Hui-Yao Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yue Yuan
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yu-Hong Fu
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ying Wang
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xin-Yuan Gao
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
19
|
Macrophage metabolic reprogramming aggravates aortic dissection through the HIF1α-ADAM17 pathway ✰. EBioMedicine 2019; 49:291-304. [PMID: 31640947 PMCID: PMC6945268 DOI: 10.1016/j.ebiom.2019.09.041] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/17/2022] Open
Abstract
Background Aortic dissection is a severe inflammatory vascular disease with high mortality and limited therapeutic options. The hallmarks of aortic dissection comprise aortic inflammatory cell infiltration and elastic fiber disruption, highlighting the involvement of macrophage. Here a role for macrophage hypoxia-inducible factor 1-alpha (HIF-1α) in aortic dissection was uncovered. Methods Immunochemistry, immunofluorescence, western blot and qPCR were performed to test the change of macrophage HIF-1α in two kinds of aortic dissection models and human tissues. Metabolomics and Seahorse extracellular flux analysis were used to detect the metabolic state of macrophages involved in the development of aortic dissection. Chromatin Immunoprecipitation (ChIP), enzyme-linked immunosorbent assay (ELISA) and cytometric bead array (CBA) were employed for mechanistic studies. Findings Macrophages involved underwent distinct metabolic reprogramming, especially fumarate accumulation, thus inducing HIF-1α activation in the development of aortic dissection in human and mouse models. Mechanistic studies revealed that macrophage HIF-1α activation triggered vascular inflammation, extracellular matrix degradation and elastic plate breakage through increased a disintegrin and metallopeptidase domain 17 (ADAM17), identified as a novel target gene of HIF-1α. A HIF-1α specific inhibitor acriflavine elicited protective effects on aortic dissection dependent on macrophage HIF-1α. Interpretation This study reveals that macrophage metabolic reprogramming activates HIF-1α and subsequently promotes aortic dissection progression, suggesting that macrophage HIF-1α inhibition might be a potential therapeutic target for treating aortic dissection.
Collapse
|
20
|
Mury P, Chirico EN, Mura M, Millon A, Canet-Soulas E, Pialoux V. Oxidative Stress and Inflammation, Key Targets of Atherosclerotic Plaque Progression and Vulnerability: Potential Impact of Physical Activity. Sports Med 2019; 48:2725-2741. [PMID: 30302720 DOI: 10.1007/s40279-018-0996-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a complex cardiovascular disease, is a leading cause of mortality and morbidity worldwide. Oxidative stress and inflammation are both involved in the development of atherosclerotic plaque as they increase the biological processes associated with this pathology, such as endothelial dysfunction and macrophage recruitment and adhesion. Atherosclerotic plaque rupture leading to major ischemic events is the result of vulnerable plaque progression, which is a result of the detrimental effect of oxidative stress and inflammation on risk factors for atherosclerotic plaque rupture, such as intraplaque hemorrhage, neovascularization, and fibrous cap thickness. Thus, both are key targets for primary and secondary interventions. It is well recognized that chronic physical activity attenuates oxidative stress in healthy subjects via the improvement of antioxidant enzyme capacities and inflammation via the enhancement of anti-inflammatory molecules. Moreover, it was recently shown that chronic physical activity could decrease oxidative stress and inflammation in atherosclerotic patients. The aim of this review is to summarize the role of oxidative stress and inflammation in atherosclerosis and the results of therapeutic interventions targeting them in both preclinical and clinical studies. The effects of chronic physical activity on these two key processes are then reviewed in vulnerable atherosclerotic plaques in both coronary and carotid arteries.
Collapse
Affiliation(s)
- Pauline Mury
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Erica N Chirico
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Mathilde Mura
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Antoine Millon
- University of Lyon, University Claude Bernard Lyon 1, CarMeN Laboratory, INSERM U1060, Bron, France.,Department of Vascular Surgery, Edouard Herriot Hospital, Lyon, France
| | - Emmanuelle Canet-Soulas
- University of Lyon, University Claude Bernard Lyon 1, CarMeN Laboratory, INSERM U1060, Bron, France
| | - Vincent Pialoux
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France. .,Laboratory of Excellence GR-Ex, Paris, France. .,Institut Universitaire de France, Paris, France.
| |
Collapse
|
21
|
LI TT, WANG ZB, LI Y, CAO F, YANG BY, KUANG HX. The mechanisms of traditional Chinese medicine underlying the prevention and treatment of atherosclerosis. Chin J Nat Med 2019; 17:401-412. [DOI: 10.1016/s1875-5364(19)30048-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Indexed: 02/07/2023]
|
22
|
Emerging Roles of Redox-Mediated Angiogenesis and Oxidative Stress in Dermatoses. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2304018. [PMID: 31178954 PMCID: PMC6501144 DOI: 10.1155/2019/2304018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/14/2019] [Accepted: 03/14/2019] [Indexed: 12/24/2022]
Abstract
Angiogenesis is the process of new vessel formation, which sprouts from preexisting vessels. This process is highly complex and primarily involves several key steps, including stimulation of endothelial cells by growth factors, degradation of the extracellular matrix by proteolytic enzymes, migration and proliferation of endothelial cells, and capillary tube formation. Currently, it is considered that multiple cytokines play a vital role in this process, which consist of proangiogenic factors (e.g., vascular endothelial growth factor, fibroblast growth factors, and angiopoietins) and antiangiogenic factors (e.g., endostatin, thrombospondin, and angiostatin). Angiogenesis is essential for most physiological events, such as body growth and development, tissue repair, and wound healing. However, uncontrolled neovascularization may contribute to angiogenic disorders. In physiological conditions, the above promoters and inhibitors function in a coordinated way to induce and sustain angiogenesis within a limited period of time. Conversely, the imbalance between proangiogenic and antiangiogenic factors could cause pathological angiogenesis and trigger several diseases. With insights into the molecular mechanisms of angiogenesis, increasing reports have shown that a close relationship exists between angiogenesis and oxidative stress (OS) in both physiological and pathological conditions. OS, an imbalance between prooxidant and antioxidant systems, is a cause and consequence of many vascular complains and serves as one of the biomarkers for these diseases. Furthermore, emerging evidence supports that OS and angiogenesis play vital roles in many dermatoses, such as psoriasis, atopic dermatitis, and skin tumor. This review summarizes recent findings on the role of OS as a trigger of angiogenesis in skin disorders, highlights newly identified mechanisms, and introduces the antiangiogenic and antioxidant therapeutic strategies.
Collapse
|
23
|
Zhang M, Xu Y, Jiang L. Irisin attenuates oxidized low‐density lipoprotein impaired angiogenesis through AKT/mTOR/S6K1/Nrf2 pathway. J Cell Physiol 2019; 234:18951-18962. [PMID: 30942905 DOI: 10.1002/jcp.28535] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Min Zhang
- Division of Cardiology TongRen Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yinjie Xu
- Division of Cardiology TongRen Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Li Jiang
- Division of Cardiology TongRen Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
24
|
Jin F, Zheng X, Yang Y, Yao G, Ye L, Doeppner TR, Hermann DM, Wang H, Dai Y. Impairment of hypoxia-induced angiogenesis by LDL involves a HIF-centered signaling network linking inflammatory TNFα and angiogenic VEGF. Aging (Albany NY) 2019; 11:328-349. [PMID: 30659163 PMCID: PMC6366960 DOI: 10.18632/aging.101726] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023]
Abstract
Hypoxia inducible factors (HIFs) mediate angiogenesis via up-regulation of various pro-angiogenic factors (particularly VEGF) in response to hypoxia. Here, we report that hypoxia unexpectedly induced robust production of the pro-inflammatory factor TNFα by endothelial cells (ECs), suggesting an autocrine loop that in turn activated HIFs via an NF-κB-dependent process, resulting in production of VEGF and thereby promotion of angiogenesis. In contrast, low-density lipoprotein (LDL) prevented expression of HIFs in ECs exposed to either hypoxia or TNFα, while knockdown of either HIF-1α or HIF-2α strikingly attenuated hypoxia-induced production of VEGF by ECs as well as EC colony formation and tube formation. Significantly, LDL attenuated hypoxia-induced angiogenesis by disrupting the TNFα/NF-κB/HIF/VEGF signaling cascade via down-regulation of the TNF receptor TNF-R1, rather than TNFα itself, and multiple key components of both canonical and non-canonical NF-κB pathways. By doing so, LDL was able to either inhibit or down-regulate a wide spectrum of HIF-dependent pro-angiogenic downstream targets and signals. Together, these findings argue existence of a self-regulatory TNFα/NF-κB/HIF/VEGF signaling network in ECs, which mediates and fine-tones angiogenesis, at least in response to hypoxia. They also suggest that LDL impairs angiogenesis by disrupting this network, which might represent a novel mechanism underlying anti-angiogenic property of LDL.
Collapse
Affiliation(s)
- Fengyan Jin
- Department of Hematology, Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiangyu Zheng
- Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanping Yang
- Department of Hematology, Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Gang Yao
- Department of Neurology, the Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Long Ye
- Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Thorsten R. Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Dirk M. Hermann
- Department of Neurology, University Duisburg-Essen Medical School, Essen, Germany
| | - Haifeng Wang
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
25
|
Yang Y, Luo H, Zhou C, Zhang R, Liu S, Zhu X, Ke S, Liu H, Lu Z, Chen M. Regulation of capillary tubules and lipid formation in vascular endothelial cells and macrophages via extracellular vesicle-mediated microRNA-4306 transfer. J Int Med Res 2018; 47:453-469. [PMID: 30477383 PMCID: PMC6384455 DOI: 10.1177/0300060518809255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Objective This study aimed to examine regulation of capillary tubules and lipid formation in vascular endothelial cells and macrophages via extracellular vesicle-mediated microRNA (miRNA)-4306 transfer Methods Whole blood samples (12 mL) were collected from 53 patients, and miR-4306 levels in extracellular vesicles (EVs) were analyzed by reverse transcription-polymerase chain reaction. Human coronary artery vascular endothelial cells (HCAECs) and human monocyte-derived macrophages (HMDMs) were transfected with a scrambled oligonucleotide, an miR-4306 mimic, or an anti-miR-4306 inhibitor. The direct effect of miR-4306 on the target gene was analyzed by a dual-luciferase reporter assay. Results EV-contained miR-4306 released from HMDMs was significantly upregulated in coronary artery disease. Oxidized low-density lipoprotein (ox-LDL)-stimulated HMDM-derived EVs inhibited proliferation, migration, and angiogenesis abilities of HCAECs in vitro. However, ox-LDL-stimulated HCAEC-derived EVs enhanced lipid formation of HMDMs. The possible mechanism of these findings was partly due to EV-mediated miR-4306 upregulation of the Akt/nuclear factor kappa B signaling pathway. Conclusions Paracrine cellular crosstalk between HCAECs and HMDMs probably supports the pro-atherosclerotic effects of EVs under ox-LDL stress.
Collapse
Affiliation(s)
- Ying Yang
- 1 Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hui Luo
- 2 Department of Cardiothoracic Surgery, Nanchong Central Hospital, Nanchong, China
| | - Can Zhou
- 1 Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Rongyi Zhang
- 3 Department of Cardiology, Nanchong Central Hospital, Nanchong, China
| | - Si Liu
- 3 Department of Cardiology, Nanchong Central Hospital, Nanchong, China
| | - Xiao Zhu
- 1 Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Sha Ke
- 1 Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hui Liu
- 1 Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhan Lu
- 1 Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Mao Chen
- 4 Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Sedding DG, Boyle EC, Demandt JAF, Sluimer JC, Dutzmann J, Haverich A, Bauersachs J. Vasa Vasorum Angiogenesis: Key Player in the Initiation and Progression of Atherosclerosis and Potential Target for the Treatment of Cardiovascular Disease. Front Immunol 2018; 9:706. [PMID: 29719532 PMCID: PMC5913371 DOI: 10.3389/fimmu.2018.00706] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/22/2018] [Indexed: 01/08/2023] Open
Abstract
Plaque microvascularization and increased endothelial permeability are key players in the development of atherosclerosis, from the initial stages of plaque formation to the occurrence of acute cardiovascular events. First, endothelial dysfunction and increased permeability facilitate the entry of diverse inflammation-triggering molecules and particles such as low-density lipoproteins into the artery wall from the arterial lumen and vasa vasorum (VV). Recognition of entering particles by resident phagocytes in the vessel wall triggers a maladaptive inflammatory response that initiates the process of local plaque formation. The recruitment and accumulation of inflammatory cells and the subsequent release of several cytokines, especially from resident macrophages, stimulate the expansion of existing VV and the formation of new highly permeable microvessels. This, in turn, exacerbates the deposition of pro-inflammatory particles and results in the recruitment of even more inflammatory cells. The progressive accumulation of leukocytes in the intima, which trigger proliferation of smooth muscle cells in the media, results in vessel wall thickening and hypoxia, which further stimulates neoangiogenesis of VV. Ultimately, this highly inflammatory environment damages the fragile plaque microvasculature leading to intraplaque hemorrhage, plaque instability, and eventually, acute cardiovascular events. This review will focus on the pivotal roles of endothelial permeability, neoangiogenesis, and plaque microvascularization by VV during plaque initiation, progression, and rupture. Special emphasis will be given to the underlying molecular mechanisms and potential therapeutic strategies to selectively target these processes.
Collapse
Affiliation(s)
- Daniel G Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Erin C Boyle
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Jasper A F Demandt
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands.,BHF Centre for Cardiovascular Science, Edinburgh University, Edinburgh, United Kingdom
| | - Jochen Dutzmann
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
27
|
Bao MH, Li GY, Huang XS, Tang L, Dong LP, Li JM. Long Noncoding RNA LINC00657 Acting as a miR-590-3p Sponge to Facilitate Low Concentration Oxidized Low-Density Lipoprotein-Induced Angiogenesis. Mol Pharmacol 2018; 93:368-375. [PMID: 29436491 DOI: 10.1124/mol.117.110650] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/29/2018] [Indexed: 02/14/2025] Open
Abstract
Angiogenesis in atherosclerotic plaque promotes plaque growth, causes plaque hemorrhage, and violates plaque stability. LINC00657 is a long noncoding RNA highly conserved and abundantly expressed in vascular endothelial cells. The present study was designed to investigate the effects and mechanisms of LINC00675 on low concentrations of oxidized low-density lipoprotein (oxLDL)-induced angiogenesis. Cell proliferation, transwell, wound healing, and tube formation assays were conducted to detect the effects of low concentrations of oxLDL on angiogenesis; the results discovered that oxLDL promoted cell proliferation, migration, and tube formation. oxLDL also upregulated LINC00657 expression. Inhibition of LINC00657 by siRNA significantly suppressed oxLDL-induced endothelial cell proliferation, migration, and tube formation. Bioinformatic assay indicated six binding sites in the LINC00657 sequence to miR-590-3p. The upregulation of LINC00657 was related to the downregulation of miR-590-3p in oxLDL-treated endothelial cells; while downregulation of LINC00657 resulted in upregulation of miR-590-3p. The antiangiogenesis effects of si-LINC00657 were partly abrogated by miR-590-3p inhibitor. Further dual-luciferase assay found miR-590-3p inhibited the expression of hypoxia-inducible factor 1α (HIF-1α) by binding to the position of 689-696 in HIF-1α 3'-untranslated region directly. MiR-590-3p also inhibited the oxLDL-induced upregulation of HIF-1α, vascular endothelial growth factor (VEGF), matrix metalloproteinase-2 (MMP-2), and matrix metalloproteinase-9 (MMP-9). These results suggested that in oxLDL-treated endothelial cells, LINC00657 acted as a miR-590-3p sponge to attenuate the suppression of miR-590-3p on HIF-1α, and to promote angiogenesis through VEGF, MMP-2, and MMP-9. The present study provided new insight into the roles of LINC00657 and miR-590-3p in preventing oxLDL-induced angiogenesis and may provide a novel strategy for atherosclerosis treatment.
Collapse
Affiliation(s)
- Mei-Hua Bao
- Department of Anatomy, Histology and Embryology, Science Research Center, Institute of Neuroscience (M.-H.B., G.-Y.L., L.T., L.-P.D., J.-M.L.), and Department of Pharmacology (X.-S.H.), Changsha Medical University, Changsha, China
| | - Guang-Yi Li
- Department of Anatomy, Histology and Embryology, Science Research Center, Institute of Neuroscience (M.-H.B., G.-Y.L., L.T., L.-P.D., J.-M.L.), and Department of Pharmacology (X.-S.H.), Changsha Medical University, Changsha, China
| | - Xiao-Shan Huang
- Department of Anatomy, Histology and Embryology, Science Research Center, Institute of Neuroscience (M.-H.B., G.-Y.L., L.T., L.-P.D., J.-M.L.), and Department of Pharmacology (X.-S.H.), Changsha Medical University, Changsha, China
| | - Liang Tang
- Department of Anatomy, Histology and Embryology, Science Research Center, Institute of Neuroscience (M.-H.B., G.-Y.L., L.T., L.-P.D., J.-M.L.), and Department of Pharmacology (X.-S.H.), Changsha Medical University, Changsha, China
| | - Li-Ping Dong
- Department of Anatomy, Histology and Embryology, Science Research Center, Institute of Neuroscience (M.-H.B., G.-Y.L., L.T., L.-P.D., J.-M.L.), and Department of Pharmacology (X.-S.H.), Changsha Medical University, Changsha, China
| | - Jian-Ming Li
- Department of Anatomy, Histology and Embryology, Science Research Center, Institute of Neuroscience (M.-H.B., G.-Y.L., L.T., L.-P.D., J.-M.L.), and Department of Pharmacology (X.-S.H.), Changsha Medical University, Changsha, China
| |
Collapse
|
28
|
Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV, Orekhov AN. Role of lipids and intraplaque hypoxia in the formation of neovascularization in atherosclerosis. Ann Med 2017; 49:661-677. [PMID: 28797175 DOI: 10.1080/07853890.2017.1366041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
According to the current paradigm, chronic vascular inflammation plays a central role in the pathogenesis of atherosclerosis. The plaque progression is typically completed with rupture and subsequent acute cardiovascular complications. Previously, the role of adventitial vasa vasorum in atherogenesis was underestimated. However, investigators then revealed that vasa vasorum neovascularization can be observed when no clinical manifestation of atherosclerosis is present. Vasa vasorum is involved in various proatherogenic processes such as intimal accumulation of inflammatory leukocytes, intimal thickening, necrotic core formation, intraplaque haemorrhage, lesion rupture and atherothrombosis. Due to the destabilizing action of the intraplaque microenvironment, lesional vasa vasorum neovessels experience serious defects and abnormalities during development that leads to their immaturity, fragility and leakage. Indeed, intraplaque neovessels are a main cause of intraplaque haemorrhage. Visualization techniques showed that presence of neovascularization/haemorrhage can serve as a good indicator of lesion instability and higher risk of rupture. Vasa vasorum density is a strong predictor of acute cardiovascular events such as sudden death, myocardial infarction and stroke. At present, arterial vasa vasorum neovascularization is under intensive investigation along with development of therapeutic tools focused on the control of formation of vasa vasorum neovessels in order to prevent plaque haemorrhage/rupture and thromboembolism. KEY MESSAGE Neovascularization plays an important role in atherosclerosis, being involved in unstable plaque formation. Presence of neovascularization and haemorrhage indicates plaque instability and risk of rupture. Various imaging techniques are available to study neovascularization.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- a Department of Neurochemistry, Division of Basic and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia
| | - Alexandra A Melnichenko
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia
| | - Veronika A Myasoedova
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia
| | - Andrey V Grechko
- c Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology , Moscow , Russia
| | - Alexander N Orekhov
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia.,d Institute for Atherosclerosis Research, Skolkovo Innovative Center , Moscow , Russia
| |
Collapse
|
29
|
Alaarg A, Pérez-Medina C, Metselaar JM, Nahrendorf M, Fayad ZA, Storm G, Mulder WJM. Applying nanomedicine in maladaptive inflammation and angiogenesis. Adv Drug Deliv Rev 2017; 119:143-158. [PMID: 28506745 PMCID: PMC5682240 DOI: 10.1016/j.addr.2017.05.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/12/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022]
Abstract
Inflammation and angiogenesis drive the development and progression of multiple devastating diseases such as atherosclerosis, cancer, rheumatoid arthritis, and inflammatory bowel disease. Though these diseases have very different phenotypic consequences, they possess several common pathophysiological features in which monocyte recruitment, macrophage polarization, and enhanced vascular permeability play critical roles. Thus, developing rational targeting strategies tailored to the different stages of the journey of monocytes, from bone marrow to local lesions, and their extravasation from the vasculature in diseased tissues will advance nanomedicine. The integration of in vivo imaging uniquely allows studying nanoparticle kinetics, accumulation, clearance, and biological activity, at levels ranging from subcellular to an entire organism, and will shed light on the fate of intravenously administered nanomedicines. We anticipate that convergence of nanomedicines, biomedical engineering, and life sciences will help to advance clinically relevant therapeutics and diagnostic agents for patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Amr Alaarg
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Josbert M Metselaar
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Institute for Experimental Molecular Imaging, University Clinic, Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gert Storm
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
30
|
Wang Z, Wei Q, Han L, Cao K, Lan T, Xu Z, Wang Y, Gao Y, Xue J, Shan F, Feng J, Xie X. Tenascin-c renders a proangiogenic phenotype in macrophage via annexin II. J Cell Mol Med 2017; 22:429-438. [PMID: 28857429 PMCID: PMC5742692 DOI: 10.1111/jcmm.13332] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 06/21/2017] [Indexed: 12/31/2022] Open
Abstract
Tenascin-c is an extracellular matrix glycoprotein, the expression of which relates to the progression of atherosclerosis, myocardial infarction and heart failure. Annexin II acts as a cell surface receptor of tenascin-c. This study aimed to delineate the role of tenascin-c and annexin II in macrophages presented in atherosclerotic plaque. Animal models with atherosclerotic lesions were established using ApoE-KO mice fed with high-cholesterol diet. The expression of tenascin-c and annexin II in atherosclerotic lesions was determined by qRT-PCR, Western blot and immunohistochemistry analysis. Raw 264.7 macrophages and human primary macrophages were exposed to 5, 10 and 15 μg/ml tenascin-c for 12 hrs. Cell migration as well as the proangiogenic ability of macrophages was examined. Additionally, annexin II expression was delineated in raw 264.7 macrophages under normal condition (20% O2 ) for 12 hrs or hypoxic condition (1% O2 ) for 6-12 hrs. The expression of tenascin-c and annexin II was markedly augmented in lesion aorta. Tenascin-c positively regulated macrophage migration, which was dependent on the expression of annexin II in macrophages. VEGF release from macrophages and endothelial tube induction by macrophage were boosted by tenascin-c and attenuated by annexin II blocking. Furthermore, tenascin-c activated Akt/NF-κB and ERK signalling through annexin II. Lastly, hypoxia conditioning remarkably facilitates annexin II expression in macrophages through hypoxia-inducible factor (HIF)-1α but not HIF-2α. In conclusion, tenascin-c promoted macrophage migration and VEGF expression through annexin II, the expression of which was modulated by HIF-1α.
Collapse
Affiliation(s)
- Zhiyang Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Qi Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Liang Han
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Keqing Cao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Tianfeng Lan
- Institute of Integrated Medical Information, Xi'an, China
| | - Zhenjie Xu
- Institute of Integrated Medical Information, Xi'an, China
| | - Yingjuan Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Yuan Gao
- Department of Traditional Chinese Medicine, College of Life Science, Northwest University, Xi'an, China
| | - Jing Xue
- Department of Traditional Chinese Medicine, College of Life Science, Northwest University, Xi'an, China
| | - Fei Shan
- Department of Cardiovascular Surgery, Affiliated Hospital of Yan'an University, Yan'an, China
| | - Jun Feng
- Department of Vascular Surgery, the First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, China
| | - Xin Xie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China.,Institute of Integrated Medical Information, Xi'an, China
| |
Collapse
|
31
|
Yang Y, Luo NS, Ying R, Xie Y, Chen JY, Wang XQ, Gu ZJ, Mai JT, Liu WH, Wu MX, Chen ZT, Fang YB, Zhang HF, Zuo ZY, Wang JF, Chen YX. Macrophage-derived foam cells impair endothelial barrier function by inducing endothelial-mesenchymal transition via CCL-4. Int J Mol Med 2017; 40:558-568. [PMID: 28656247 PMCID: PMC5504989 DOI: 10.3892/ijmm.2017.3034] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 06/06/2017] [Indexed: 12/20/2022] Open
Abstract
Recently, endothelial-mesenchymal transition (EndMT) has been demonstrated to play an important role in the development of atherosclerosis, the molecular mechanisms of which remain unclear. In the present study, scanning electron microscopy directly revealed a widened endothelial space and immunohistofluorescence demonstrated that EndMT was increased in human aorta atherosclerotic plaques. M1 macrophage-derived foam cell (M1-FC) supernatants, but not M2 macrophage-derived foam cell (M2-FC) supernatants, induced EndMT. A protein array and enzyme-linked immunosorbent assay identified that the levels of several cytokines, including C-C motif chemokine ligand 4 (CCL-4) were increased in M1-FC supernatants, in which EndMT was promoted, accompanied by increased endothelial permeability and monocyte adhesion. Furthermore, anti-CCL-4 antibody abolished the effects of M1-FC supernatants on EndMT. At the same time, CCL-4 activated its receptor, C-C motif chemokine receptor-5 (CCR-5), and upregulated transforming growth factor-β (TGF-β) expression. Further experiments revealed that EndMT induced by CCL-4 was reversed by treatment with CCR-5 antagonist and the RNA-mediated knockdown of TGF-β. On the whole, the data of the present study suggest that M1-FCs induce EndMT by upregulating CCL-4, and increase endothelial permeability and monocyte adhesion. These data may help to elucidate the important role of EndMT in the development of atherosclerosis.
Collapse
Affiliation(s)
- Ying Yang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Nian-Sang Luo
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Ru Ying
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jia-Yuan Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiao-Qiao Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhen-Jie Gu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jing-Ting Mai
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Wen-Hao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Mao-Xiong Wu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhi-Teng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yong-Biao Fang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhi-Yi Zuo
- Laboratory of RNA and Major Disease of Heart and Brain, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yang-Xin Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
32
|
Wang L, Chen Q, Ke D, Li G. Ghrelin inhibits atherosclerotic plaque angiogenesis and promotes plaque stability in a rabbit atherosclerotic model. Peptides 2017; 90:17-26. [PMID: 28189525 DOI: 10.1016/j.peptides.2017.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/16/2017] [Accepted: 01/27/2017] [Indexed: 12/17/2022]
Abstract
Intraplaque angiogenesis associates with the instability of atherosclerotic plaques. In the present study, we investigated the effects of ghrelin on intraplaque angiogenesis and plaque instability in a rabbit model of atherosclerosis. The rabbits were randomly divided into three groups, namely, the control group, atherosclerotic model group, and ghrelin-treated group, with treatments lasting for 4 weeks. We found that the thickness ratio of the intima to media in rabbits of the ghrelin-treated group was significantly lower than that in rabbits of the atherosclerotic model group. The number of neovessels and the levels of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR2) decreased dramatically in rabbits of the ghrelin-treated group compared to those of the atherosclerotic model group. Ghrelin significantly decreased the plaque content of macrophages, matrix metalloproteinase (MMP)-2, and MMP-9, in a rabbit model of atherosclerosis. In addition, the level of the pro-inflammatory factor monocyte chemoattractant protein (MCP)-1 was significantly lower in rabbits of the ghrelin-treated group than in rabbits of the atherosclerotic model group. In summary, ghrelin can inhibit intraplaque angiogenesis and promote plaque stability by down-regulating VEGF and VEGFR2 expression, inhibiting the plaque content of macrophages, and reducing MCP-1 expression at an advanced stage of atherosclerosis in rabbits.
Collapse
Affiliation(s)
- Li Wang
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qingwei Chen
- Department of Geriatrics, The Second Affiliated Hospital, Chongqing University of Medical Sciences, Chongqing, 400010, China.
| | - Dazhi Ke
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Guiqiong Li
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
33
|
Cimmino G, Loffredo FS, Morello A, D'Elia S, De Palma R, Cirillo P, Golino P. Immune-Inflammatory Activation in Acute Coronary Syndromes: A Look into the Heart of Unstable Coronary Plaque. Curr Cardiol Rev 2017; 13:110-117. [PMID: 27758696 PMCID: PMC5452145 DOI: 10.2174/1573403x12666161014093812] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/24/2016] [Accepted: 10/06/2016] [Indexed: 12/21/2022] Open
Abstract
In the last twenty years, our comprehension of the molecular mechanisms involved in the formation, progression and complication of atherosclerotic plaque has advanced significantly and the main role of inflammation and immunity in this phenomenon is now largely accepted. Accumulating evidence highlight the crucial role of different inflammatory and immune cells, such as monocytes and T-lymphocytes, in the pathophysiology of atherosclerotic lesion, particularly in contributing to its com-plications, such as rupture or ulceration. According to the new terminology, “vulnerable plaque” identi-fies an inflamed atherosclerotic lesion that is particularly prone to rupture. Once disrupted, prothrom-botic material is exposed to the flowing blood, thus activating coagulation cascade and platelet aggrega-tion, ultimately leading to acute thrombus formation within the coronary vessel. To date this is the key event underlying the clinical manifestations of acute coronary syndromes (ACS). The degree of vessel occlusion (complete vs. incomplete) and the time of blood flow cessation will define the severity of clinical picture. This phenomenon seems to be the final effect of a complex inter-action between different local and systemic factors, involving the degree of inflammation, type of cells infiltration and the rheological characteristics of blood flow at the site of plaque rupture, thrombogenic substrates within the atherosclerotic lesion and different soluble mediators, already present or acutely released in the circulating blood. This article will review currently available data on the pathophysiology of ACS, emphasizing the immunological and inflammatory aspects of vulnerable plaque. We may pos-tulate that intraplaque antigens and local microenvironment will define the immune-inflammatory re-sponse and cells phenotype, thus determining the severity of clinical manifestations.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| | - Francesco S Loffredo
- Molecular Cardiology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Alberto Morello
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| | - Saverio D'Elia
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| | - Raffaele De Palma
- Department of Clinical and Experimental Medicine, Section of Immunology, Second University of Naples, Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Golino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, Second University of Naples, Naples, Italy
| |
Collapse
|
34
|
Integrin signaling in atherosclerosis. Cell Mol Life Sci 2017; 74:2263-2282. [PMID: 28246700 DOI: 10.1007/s00018-017-2490-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic lipid-driven inflammatory disease affecting large arteries, represents the primary cause of cardiovascular disease in the world. The local remodeling of the vessel intima during atherosclerosis involves the modulation of vascular cell phenotype, alteration of cell migration and proliferation, and propagation of local extracellular matrix remodeling. All of these responses represent targets of the integrin family of cell adhesion receptors. As such, alterations in integrin signaling affect multiple aspects of atherosclerosis, from the earliest induction of inflammation to the development of advanced fibrotic plaques. Integrin signaling has been shown to regulate endothelial phenotype, facilitate leukocyte homing, affect leukocyte function, and drive smooth muscle fibroproliferative remodeling. In addition, integrin signaling in platelets contributes to the thrombotic complications that typically drive the clinical manifestation of cardiovascular disease. In this review, we examine the current literature on integrin regulation of atherosclerotic plaque development and the suitability of integrins as potential therapeutic targets to limit cardiovascular disease and its complications.
Collapse
|
35
|
Camaré C, Pucelle M, Nègre-Salvayre A, Salvayre R. Angiogenesis in the atherosclerotic plaque. Redox Biol 2017; 12:18-34. [PMID: 28212521 PMCID: PMC5312547 DOI: 10.1016/j.redox.2017.01.007] [Citation(s) in RCA: 296] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a multifocal alteration of the vascular wall of medium and large arteries characterized by a local accumulation of cholesterol and non-resolving inflammation. Atherothrombotic complications are the leading cause of disability and mortality in western countries. Neovascularization in atherosclerotic lesions plays a major role in plaque growth and instability. The angiogenic process is mediated by classical angiogenic factors and by additional factors specific to atherosclerotic angiogenesis. In addition to its role in plaque progression, neovascularization may take part in plaque destabilization and thromboembolic events. Anti-angiogenic agents are effective to reduce atherosclerosis progression in various animal models. However, clinical trials with anti-angiogenic drugs, mainly anti-VEGF/VEGFR, used in anti-cancer therapy show cardiovascular adverse effects, and require additional investigations.
Collapse
Affiliation(s)
- Caroline Camaré
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France; Université Paul Sabatier Toulouse III, Faculty of Medicine, Biochemistry Departement, Toulouse, France; CHU Toulouse, Rangueil, 1 avenue Jean Poulhès, TSA 50032, 31059 Toulouse Cedex 9, France
| | - Mélanie Pucelle
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France
| | - Anne Nègre-Salvayre
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France.
| | - Robert Salvayre
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France; Université Paul Sabatier Toulouse III, Faculty of Medicine, Biochemistry Departement, Toulouse, France; CHU Toulouse, Rangueil, 1 avenue Jean Poulhès, TSA 50032, 31059 Toulouse Cedex 9, France.
| |
Collapse
|
36
|
Liberale L, Dallegri F, Montecucco F, Carbone F. Pathophysiological relevance of macrophage subsets in atherogenesis. Thromb Haemost 2017; 117:7-18. [PMID: 27683760 DOI: 10.1160/th16-08-0593] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
Macrophages are highly heterogeneous and plastic cells. They were shown to play a critical role in all stages of atherogenesis, from the initiation to the necrotic core formation and plaque rupture. Lesional macrophages primarily derive from blood monocyte, but local macrophage proliferation as well as differentiation from smooth muscle cells have also been described. Within atherosclerotic plaques, macrophages rapidly respond to changes in the microenvironment, shifting between pro- (M1) or anti-inflammatory (M2) functional phenotypes. Furthermore, different stimuli have been associated with differentiation of newly discovered M2 subtypes: IL-4/IL-13 (M2a), immune-complex (M2b), IL-10/glucocorticoids (M2c), and adenosine receptor agonist (M2d). More recently, additional intraplaque macrophage phenotypes were also recognized in response to CXCL4 (M4), oxidized phospholipids (Mox), haemoglobin/haptoglobin complexes (HA-mac/M(Hb)), and heme (Mhem). Such macrophage polarization was described as a progression among multiple phenotypes, which reflect the activity of different transcriptional factors and the cross-talk between intracellular signalling. Finally, the distribution of macrophage subsets within different plaque areas was markedly associated with cardiovascular (CV) vulnerability. The aim of this review is to update the current knowledge on the role of macrophage subsets in atherogenesis. In addition, the molecular mechanisms underlying macrophage phenotypic shift will be summarised and discussed. Finally, the role of intraplaque macrophages as predictors of CV events and the therapeutic potential of these cells will be discussed.
Collapse
Affiliation(s)
| | | | - Fabrizio Montecucco
- Fabrizio Montecucco, MD, PhD, Department of Internal Medicine, University of Genoa School of Medicine, 6 viale Benedetto XV, 16132 Genoa, Italy, Tel.: +39 010 353 8694, Fax: +39 010 353 8686, E-mail:
| | | |
Collapse
|
37
|
de Vries MR, Quax PHA. Plaque angiogenesis and its relation to inflammation and atherosclerotic plaque destabilization. Curr Opin Lipidol 2016; 27:499-506. [PMID: 27472406 DOI: 10.1097/mol.0000000000000339] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The review discusses the recent literature on plaque angiogenesis and its relation to inflammation and plaque destabilization. Furthermore, it discusses how plaque angiogenesis can be used to monitor atherosclerosis and serve as a therapeutic target. RECENT FINDINGS Histopathologic studies have shown a clear relationship between plaque angiogenesis, intraplaque hemorrhage (IPH), plaque vulnerability, and cardiovascular events. Hypoxia is a main driver of plaque angiogenesis and the mechanism behind angiogenesis is only partly known. IPH, as the result of immature neovessels, is associated with increased influx of inflammatory cells in the plaques. Experimental models displaying certain features of human atherosclerosis such as plaque angiogenesis or IPH are developed and can contribute to unraveling the mechanism behind plaque vulnerability. New imaging techniques are established, with which plaque angiogenesis and vulnerability can be detected. Furthermore, antiangiogenic therapies in atherosclerosis gain much attention. SUMMARY Plaque angiogenesis, IPH, and inflammation contribute to plaque vulnerability. Histopathologic and imaging studies together with specific experimental studies have provided insights in plaque angiogenesis and plaque vulnerability. However, more extensive knowledge on the underlying mechanism is required for establishing new therapies for patients at risk.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
38
|
Cao J, Li G, Wang M, Li H, Han Z. Protective effect of oleanolic acid on oxidized-low density lipoprotein induced endothelial cell apoptosis. Biosci Trends 2016; 9:315-24. [PMID: 26559024 DOI: 10.5582/bst.2015.01094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oleanolic acid (3β-hydroxyolean-12-en-28-oic acid, OA) is a naturally-occurring triterpenoid with various promising pharmacological properties. The present study was conducted to determine the protective effects of OA against oxidized low-density lipoprotein (ox-LDL) induced endothelial cell apoptosis and the possible underlying mechanisms. Our results showed that ox-LDL significantly decreased cell viability and induced apoptosis in human umbilical vein endothelial cells (HUVECs). OA in the co-treatment showed a protective effect against ox-LDL induced loss in cell viability and an increase in apoptosis, which was associated with the modulating effect of OA on ox-LDL induced hypoxia-inducible factor 1α(HIF-1α) expression. Moreover, our results showed that the modulating effect of OA against ox-LDL induced HIF-1α expression was obtained via inhibition of lipoprotein receptor 1 (LOX-1)/reactive oxygen species (ROS) signaling. Collectively, we suggested that the protective effect of OA against ox-LDL induced HUVEC apoptosis might, at least in part, be obtained via inhibition of the LOX-1/ROS/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Jianhua Cao
- Department of Pharmacy, the Third People's Hospital of Qingdao
| | | | | | | | | |
Collapse
|
39
|
Lin N, Simon MC. Hypoxia-inducible factors: key regulators of myeloid cells during inflammation. J Clin Invest 2016; 126:3661-3671. [PMID: 27599290 DOI: 10.1172/jci84426] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hypoxia is a prominent characteristic of many acute or chronic inflammatory diseases, and exerts significant influence on their progression. Macrophages and neutrophils are major cellular components of innate immunity and contribute not only to O2 deprivation at the site of inflammation, but also alter many of their functions in response to hypoxia to either facilitate or suppress inflammation. Hypoxia stabilizes HIF-αs in macrophages and neutrophils, and these O2-sensitive transcription factors are key regulators of inflammatory responses in myeloid cells. In this review, we will summarize our current understanding of the role of HIF-αs in shaping macrophage and neutrophil functions in the pathogenesis and progression of multiple inflammatory diseases.
Collapse
|
40
|
Abstract
The anoxemia theory proposes that an imbalance between the demand for and supply of oxygen in the arterial wall is a key factor in the development of atherosclerosis. There is now substantial evidence that there are regions within the atherosclerotic plaque in which profound hypoxia exists; this may fundamentally change the function, metabolism, and responses of many of the cell types found within the developing plaque and whether the plaque will evolve into a stable or unstable phenotype. Hypoxia is characterized in molecular terms by the stabilization of hypoxia-inducible factor (HIF) 1α, a subunit of the heterodimeric nuclear transcriptional factor HIF-1 and a master regulator of oxygen homeostasis. The expression of HIF-1 is localized to perivascular tissues, inflammatory macrophages, and smooth muscle cells adjacent to the necrotic core of atherosclerotic lesions and regulates several genes that are important to vascular function including vascular endothelial growth factor, nitric oxide synthase, endothelin-1, and erythropoietin. This review summarizes the effects of hypoxia on the functions of cells involved in atherogenesis and the evidence for its potential importance from experimental models and clinical studies.
Collapse
Affiliation(s)
- Gordon A A Ferns
- 1 Department of Medical Education, Brighton & Sussex Medical School, Brighton, United Kingdom
| | - Lamia Heikal
- 1 Department of Medical Education, Brighton & Sussex Medical School, Brighton, United Kingdom
| |
Collapse
|
41
|
Van der Veken B, De Meyer GR, Martinet W. Intraplaque neovascularization as a novel therapeutic target in advanced atherosclerosis. Expert Opin Ther Targets 2016; 20:1247-57. [PMID: 27148888 DOI: 10.1080/14728222.2016.1186650] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Atherosclerosis is a lipid-driven inflammatory process with a tremendously high mortality due to acute cardiac events. There is an emerging need for new therapies to stabilize atherosclerotic lesions. Growing evidence suggests that intraplaque (IP) neovascularisation and IP hemorrhages are important contributors to plaque instability. AREAS COVERED Neovascularization is a complex process that involves different growth factors and inflammatory mediators of which their individual significance in atherosclerosis remains poorly understood. This review discusses different aspects of IP neovascularization in atherosclerosis including the potential treatment opportunities to stabilize advanced plaques. Furthermore, we highlight the development of accurate and feasible in vivo imaging modalities for IP neovascularization to prevent acute events. EXPERT OPINION Although lack of a valuable animal model of IP neovascularization impeded the investigation of a causal and straightforward link between neovascularization and atherosclerosis, recent evidence shows that vein grafts in ApoE*3 Leiden mice as well as plaques in ApoE(-/-) Fbn1(C1039G+/-) mice are useful models for intraplaque neovessel research. Even though interference with vascular endothelial growth factor (VEGF) signalling has been widely investigated, new therapeutic opportunities have emerged. Cell metabolism, in particular glycolysis and fatty acid oxidation, appears to perform a crucial role in the development of IP neovessels and thereby serves as a promising target.
Collapse
Affiliation(s)
- Bieke Van der Veken
- a Laboratory of Physiopharmacology , University of Antwerp , Antwerp , Belgium
| | - Guido Ry De Meyer
- a Laboratory of Physiopharmacology , University of Antwerp , Antwerp , Belgium
| | - Wim Martinet
- a Laboratory of Physiopharmacology , University of Antwerp , Antwerp , Belgium
| |
Collapse
|
42
|
Camaré C, Augé N, Pucelle M, Saint-Lebes B, Grazide MH, Nègre-Salvayre A, Salvayre R. The neutral sphingomyelinase-2 is involved in angiogenic signaling triggered by oxidized LDL. Free Radic Biol Med 2016; 93:204-16. [PMID: 26855418 DOI: 10.1016/j.freeradbiomed.2016.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/31/2016] [Accepted: 02/02/2016] [Indexed: 12/31/2022]
Abstract
Capillaries of the external part of the normal arterial wall constitute the vasa vasorum network. In atherosclerotic lesions, neovascularization occurs in areas of intimal hyperplasia where it may promote plaque expansion, and intraplaque hemorrhage. Oxidized LDL that are present in atherosclerotic areas activate various angiogenic signaling pathways, including reactive oxygen species and the sphingosine kinase/sphingosine-1-phosphate pathway. We aimed to investigate whether oxidized LDL-induced angiogenesis requires neutral sphingomyelinase-2 activation and the neutral sphingomyelinase-2/sphingosine kinase-1 pathway. The role of neutral sphingomyelinase-2 in angiogenic signaling was investigated in Human Microvascular Endothelial Cells (HMEC-1) forming capillary tube on Matrigel and in vivo in the Matrigel plug assay in C57BL/6 mice and in the chicken chorioallantoic membrane model. Low concentration of human oxidized LDL elicits HMEC-1 capillary tube formation and neutral sphingomyelinase-2 activation, which were blocked by neutral sphingomyelinase-2 inhibitors, GW4869 and specific siRNA. This angiogenic effect was mimicked by low concentration of C6-Ceramide and was inhibited by sphingosine kinase-1 inhibitors. Upstream of neutral sphingomyelinase-2, oxidized LDL-induced activation required LOX-1, reactive oxygen species generation by NADPH oxidase and p38-MAPK activation. Inhibition of sphingosine kinase-1 blocked the angiogenic response and triggered HMEC-1 apoptosis. Low concentration of oxidized LDL was angiogenic in vivo, both in the Matrigel plug assay in mice and in the chorioallantoic membrane model, and was blocked by GW4869. In conclusion, low oxLDL concentration triggers sprouting angiogenesis that involves ROS-induced activation of the neutral sphingomyelinase-2/sphingosine kinase-1 pathway, and is effectively inhibited by GW4869.
Collapse
Affiliation(s)
- Caroline Camaré
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Faculty of Medicine, Biochemistry Department, Toulouse, France; CHU Toulouse, Rangueil, Toulouse, France
| | - Nathalie Augé
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Mélanie Pucelle
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Bertrand Saint-Lebes
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Faculty of Medicine, Biochemistry Department, Toulouse, France; CHU Toulouse, Rangueil, Toulouse, France
| | - Marie-Hélène Grazide
- University of Toulouse, Faculty of Medicine, Biochemistry Department, Toulouse, France
| | | | - Robert Salvayre
- Inserm UMR-1048, CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France; University of Toulouse, Faculty of Medicine, Biochemistry Department, Toulouse, France; CHU Toulouse, Rangueil, Toulouse, France.
| |
Collapse
|
43
|
Oh MJ, Zhang C, LeMaster E, Adamos C, Berdyshev E, Bogachkov Y, Kohler EE, Baruah J, Fang Y, Schraufnagel DE, Wary KK, Levitan I. Oxidized LDL signals through Rho-GTPase to induce endothelial cell stiffening and promote capillary formation. J Lipid Res 2016; 57:791-808. [PMID: 26989083 DOI: 10.1194/jlr.m062539] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Indexed: 12/26/2022] Open
Abstract
Endothelial biomechanics is emerging as a key factor in endothelial function. Here, we address the mechanisms of endothelial stiffening induced by oxidized LDL (oxLDL) and investigate the role of oxLDL in lumen formation. We show that oxLDL-induced endothelial stiffening is mediated by CD36-dependent activation of RhoA and its downstream target, Rho kinase (ROCK), via inhibition of myosin light-chain phosphatase (MLCP) and myosin light-chain (MLC)2 phosphorylation. The LC-MS/MS analysis identifies 7-ketocholesterol (7KC) as the major oxysterol in oxLDL. Similarly to oxLDL, 7KC induces RhoA activation, MLCP inhibition, and MLC2 phosphorylation resulting in endothelial stiffening. OxLDL also facilitates formation of endothelial branching networks in 3D collagen gels in vitro and induces increased formation of functional blood vessels in a Matrigel plug assay in vivo. Both effects are RhoA and ROCK dependent. An increase in lumen formation was also observed in response to pre-exposing the cells to 7KC, an oxysterol that induces endothelial stiffening, but not to 5α,6α epoxide that does not affect endothelial stiffness. Importantly, loading cells with cholesterol prevented oxLDL-induced RhoA activation and the downstream signaling cascade, and reversed oxLDL-induced lumen formation. In summary, we show that oxLDL-induced endothelial stiffening is mediated by the CD36/RhoA/ROCK/MLCP/MLC2 pathway and is associated with increased endothelial angiogenic activity.
Collapse
Affiliation(s)
- Myung-Jin Oh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Chongxu Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Elizabeth LeMaster
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Crystal Adamos
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Evgeny Berdyshev
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Yedida Bogachkov
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Erin E Kohler
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - Jugajyoti Baruah
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - Yun Fang
- Department of Medicine, University of Chicago, Chicago, IL
| | - Dean E Schraufnagel
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Kishore K Wary
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
44
|
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
|
45
|
Wang L, Chen Q, Li G, Ke D. Ghrelin ameliorates impaired angiogenesis of ischemic myocardium through GHSR1a-mediated AMPK/eNOS signal pathway in diabetic rats. Peptides 2015; 73:77-87. [PMID: 26364514 DOI: 10.1016/j.peptides.2015.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/21/2015] [Accepted: 09/08/2015] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Ghrelin, an endogenous ligand of the growth hormone secretagogue receptor (GHSR), has been found to stimulate angiogenesis in vivo and in vitro. However, the effect and the corresponding mechanisms of ghrelin on impaired myocardial angiogenesis in diabetic and myocardial infarction (MI) rat model are still unknown. METHODS In the present study, adult SD rats were randomly divided into 4 groups: control, DM, DM+ghrelin, DM+ghrelin+[D-Lys3]-GHRP-6 groups. DM was induced by streptozotocin (STZ) 60 mg/kg body weight. 12 weeks post STZ injection all groups were subjected to MI, which was induced by ligation left anterior descending artery (LAD). Ghrelin and [D-Lys3]-GHRP-6 were administered via intraperitoneal injection at the doses 200 μg/kg and 50mg/kg for 4 weeks, respectively. Left ventricular function, microvascular density (MVD), myocardial infarct size, the expression of hypoxia-inducible factor (HIF1α), vascular endothelial growth factor (VEGF), fetal liver kinase-1 (Flk-1) and fms-like tyrosine kinase-1 (Flt-1), AMPK and endothelial nitric oxide synthase (eNOS) phosphorylation were examined. RESULTS Compared with the DM group, left ventricular ejection fraction (LVEF), fractional shortening (FS), and MVD were increased, whereas myocardial infarct size decreased remarkably in DM+ghrelin group. For the mechanism study, we found that ghrelin promoted the HIF1α, VEGF, Flk-1 and Flt-1 expression, AMPK and eNOS phosphorylation in diabetic rats. However, the above biochemical events in ghrelin treated diabetic rats were completely inhibited by GHSR-1a blocker [D-Lys3]-GHRP-6. CONCLUSIONS These results suggest that administration of ghrelin ameliorates impaired angiogenesis in diabetic MI rats. And these beneficial effects derive from regulating GHSR1a-mediated AMPK/eNOS signal pathway by upregulating of HIF1α, VEGF and its receptors Flk-1, Flt-1 expressions.
Collapse
Affiliation(s)
- Li Wang
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qingwei Chen
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Guiqiong Li
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Dazhi Ke
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
46
|
Yao G, Zhang Q, Doeppner TR, Niu F, Li Q, Yang Y, Kuckelkorn U, Hagemann N, Li W, Hermann DM, Dai Y, Zhou W, Jin F. LDL suppresses angiogenesis through disruption of the HIF pathway via NF-κB inhibition which is reversed by the proteasome inhibitor BSc2118. Oncotarget 2015; 6:30251-30262. [PMID: 26388611 PMCID: PMC4745795 DOI: 10.18632/oncotarget.4943] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 09/04/2015] [Indexed: 02/05/2023] Open
Abstract
Since disturbance of angiogenesis predisposes to ischemic injuries, attempts to promote angiogenesis have been made to improve clinical outcomes of patients with many ischemic disorders. While hypoxia inducible factors (HIFs) stimulate vascular remodeling and angiogenesis, hyperlipidemia impairs angiogenesis in response to various pro-angiogenic factors. However, it remains uncertain how HIFs regulate angiogenesis under hyperlipidemia. Here, we report that exposure to low-density lipoprotein (LDL) suppressed in vitro angiogenesis of human brain microvascular endothelial cells. Whereas LDL exposure diminished expression of HIF-1α and HIF-2α induced by hypoxia, it inhibited DMOG- and TNFα-induced HIF-1α and HIF-2α expression in normoxia. Notably, in both hypoxia and normoxia, LDL markedly reduced expression of HIF-1β, a constitutively stable HIF subunit, an event associated with NF-κB inactivation. Moreover, knockdown of HIF-1β down-regulated HIF-1α and HIF-2α expression, in association with increased HIF-1α hydroxylation and 20S proteasome activity after LDL exposure. Significantly, the proteasome inhibitor BSc2118 prevented angiogenesis attenuation by LDL through restoring expression of HIFs. Together, these findings argue that HIF-1β might act as a novel cross-link between the HIF and NF-κB pathways in suppression of angiogenesis by LDL, while proteasome inhibitors might promote angiogenesis by reactivating this signaling cascade under hyperlipidemia.
Collapse
Affiliation(s)
- Gang Yao
- Cancer Center, the First Affiliated Hospital, Jilin University, Changchun, Jilin, China
- Department of Neurology, the Second Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | - Qi Zhang
- Cancer Center, the First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | | | - Feng Niu
- Cancer Center, the First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | - Qiaochuan Li
- Department of Hematology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Yanping Yang
- Cancer Center, the First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | - Ulrike Kuckelkorn
- Department of Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nina Hagemann
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Wei Li
- Cancer Center, the First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | - Dirk M. Hermann
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Yun Dai
- Department of Medicine, Virginia Commonwealth University, Massey Cancer Center, Richmond, Virginia, USA
| | - Wen Zhou
- Cancer Research Institute, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, National Health and Family Planning Commission, Changsha, Hunan, China
| | - Fengyan Jin
- Cancer Center, the First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| |
Collapse
|
47
|
Wang L, Li G, Chen Q, Ke D. Octanoylated ghrelin attenuates angiogenesis induced by oxLDL in human coronary artery endothelial cells via the GHSR1a-mediated NF-κB pathway. Metabolism 2015; 64:1262-71. [PMID: 26277200 DOI: 10.1016/j.metabol.2015.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 06/24/2015] [Accepted: 07/13/2015] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Low concentrations of oxidized low-density lipoprotein (oxLDL) promote the in vitro angiogenesis of endothelial cells and play an important role in plaque angiogenesis, which may cause plaque vulnerability and enhance the risk of intravascular thrombosis. The aim of this research was to investigate the effects of octanoylated ghrelin on oxLDL-induced angiogenesis and the underlying molecular mechanisms involved in this process. MATERIALS/METHODS Human coronary artery endothelial cells (HCAECs) were incubated with 5 μg/ml oxLDL and treated with various concentrations of octanoylated ghrelin (10(-9)-10(-6)M) with or without inhibitors for 24h. Cell proliferation, migration, and in vitro angiogenesis were analyzed by bromodeoxyuridine (BrdU) staining and BrdU enzyme-linked immunosorbent assay (ELISA), transwell assay, and tube formation on Matrigel, respectively. NF-κB (nuclear factor κB) expression was determined by Western-blot analysis. RESULTS Treatment with oxLDL at 5 μg/ml enhanced the proliferation, migration and tube formation of HCAECs. In contrast, pretreatment with octanoylated ghrelin significantly attenuated in vitro angiogenesis in oxLDL-induced HCAECs. In addition, Western blot analysis indicated that NF-κB expression was increased after oxLDL treatment, and that this effect was significantly reversed by pretreatment with octanoylated ghrelin. However, the NF-κB inhibitor PDTC or the GHSR1a inhibitor [D-Lys3]-GHRP-6 abolished the effects of octanoylated ghrelin on the inhibition of angiogenesis and NF-κB p65 expression induced by oxLDL. CONCLUSIONS These findings suggest that octanoylated ghrelin attenuates angiogenesis induced by oxLDL in HCAECs via the inhibition of GHSR1a-mediated NF-κB pathway. Furthermore, octanoylated ghrelin may promote the stability of vulnerable plaques by inhibiting plaque angiogenesis.
Collapse
Affiliation(s)
- Li Wang
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Guiqiong Li
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qingwei Chen
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Dazhi Ke
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
48
|
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
|
49
|
Chen KC, Liao YC, Wang JY, Lin YC, Chen CH, Juo SHH. Oxidized low-density lipoprotein is a common risk factor for cardiovascular diseases and gastroenterological cancers via epigenomical regulation of microRNA-210. Oncotarget 2015; 6:24105-24118. [PMID: 26254226 PMCID: PMC4695173 DOI: 10.18632/oncotarget.4152] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/21/2015] [Indexed: 01/11/2023] Open
Abstract
Hyperlipidemia, including the oxidized low-density lipoprotein (oxLDL) accumulation, is a risk and highly associated with the development of cancers and cardiovascular diseases. microRNA-210 (miR-210), a hypoxia-responsive microRNA regulated by HIF-1α, has been implicated in cancer and cardiovascular disease formation. Furthermore, Bioinformatics analysis revealed that the promoter of the miR-210 gene contains CpG-rich regions. It is unclear whether miR-210 expression could be epigenetically regulated in these disease progresses. The study aimed to explore the relationships between lipid and miR-210 in the context of cardiovascular disease and gastrointestinal cancer. We demonstrated oxLDL can decrease methylation in the miR-210 promoter to up-regulate miR-210. HIF-1α can bind to miR-210 promoter, but this HIF-1α binding site can be blocked by methylation. We showed that subjects of carotid atherosclerosis, stroke patients and cancer patients had hypomethylation in the miR-210 promoter, especially the HIF-1α binding site. Furthermore, miR-210 can directly inhibit sprouty-related EVH1 domain 2 (SPRED2) expressions, and SPRED2 reduces cell migration via ERK/c-Fos/MMPs pathways. Increased miR-210 and reduced SPRED2 levels were found in aorta of mice under high-fat diet and tumor tissues, which implied that miR-210 can be an underlying mechanism to explain oxLDL as a common risk factor for cardiovascular disease and gastrointestinal cancer.
Collapse
Affiliation(s)
- Ku-Chung Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chu Liao
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Neurology, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Jaw-Yuan Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Gastroenterology and General Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Ho Chen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Suh-Hang Hank Juo
- Department of Genome Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
50
|
Quesada IM, Lucero A, Amaya C, Meijles DN, Cifuentes ME, Pagano PJ, Castro C. Selective inactivation of NADPH oxidase 2 causes regression of vascularization and the size and stability of atherosclerotic plaques. Atherosclerosis 2015; 242:469-75. [PMID: 26298737 DOI: 10.1016/j.atherosclerosis.2015.08.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/23/2015] [Accepted: 08/06/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND A variety of NADPH oxidase (Nox) isoforms including Noxs 1, 2, 4 and 5 catalyze the formation of reactive oxygen species (ROS) in the vascular wall. The Nox2 isoform complex has arguably received the greatest attention in the progression of atherogenesis in animal models. Thus, in the current study we postulated that specific Nox2 oxidase inhibition could reverse or attenuate atherosclerosis in mice fed a high-fat diet. METHODS We evaluated the effect of isoform-selective Nox2 assembly inhibitor on the progression and vascularization of atheromatous plaques. Apolipoprotein E-deficient mice (ApoE-/-) were fed a high fat diet for two months and treated over 15 days with Nox2ds-tat or control sequence (scrambled); 10 mg/kg/day, i.p. Mice were sacrificed and superoxide production in arterial tissue was detected by cytochrome C reduction assay and dihydroethidium staining. Plaque development was evaluated and the angiogenic markers VEGF, HIF1-α and visfatin were quantified by real time qRT-PCR. MMP-9 protein release and gelatinolytic activity was determined as a marker for vascularization. RESULTS Nox2ds-tat inhibited Nox-derived superoxide determined by cytochrome C in carotid arteries (2.3 ± 0.1 vs 1.7 ± 0.1 O2(•-) nmol/min*mg protein; P < 0.01) and caused a significant regression in atherosclerotic plaques in aorta (66 ± 6 μm(2) vs 37 ± 1 μm(2); scrmb vs. Nox2ds-tat; P < 0.001). Increased VEGF, HIF-1α, MMP-9 and visfatin expression in arterial tissue in response to high-fat diet were significantly attenuated by Nox2ds-tat which in turn impaired both MMP-9 protein expression and activity. CONCLUSION Given these results, it is quite evident that selective Nox inhibitors can reverse vascular pathology arising with atherosclerosis.
Collapse
Affiliation(s)
- I M Quesada
- Vascular Biology Lab, Institute of Medicine and Experimental Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| | - A Lucero
- Vascular Biology Lab, Institute of Medicine and Experimental Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| | - C Amaya
- Cellular and Molecular Biology Lab, Institute of Histology and Embryology (IHEM) CONICET, Mendoza, Argentina
| | - D N Meijles
- Department of Pharmacology & Chemical Biology and Vascular Medicine Institute, 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - M E Cifuentes
- Department of Pharmacology & Chemical Biology and Vascular Medicine Institute, 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - P J Pagano
- Department of Pharmacology & Chemical Biology and Vascular Medicine Institute, 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - C Castro
- Vascular Biology Lab, Institute of Medicine and Experimental Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina.
| |
Collapse
|