1
|
Pilard M, Babran S, Martel C. Regulation of Platelet Function by HDL. Arterioscler Thromb Vasc Biol 2025. [PMID: 40207365 DOI: 10.1161/atvbaha.124.318260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Over the past decade, increasing the capacity of HDL (high-density lipoprotein) cholesterol to mediate macrophage reverse cholesterol transport has been a target of interest in the treatment of cardiovascular diseases (CVDs). However, clinical studies reporting the limited efficacy of HDL or its main apolipoprotein, APOA1, in reducing cardiovascular events have emerged. Although HDL cholesterol is unlikely to play a direct causal role in CVD, its inverse, albeit modest, association with CVD risk, consistently observed in large population studies, suggests it may influence alternative pathways beyond cholesterol metabolism. Given the diverse functions of HDL and its components, it is conceivable that its impact on CVD occurs through less direct mechanisms. A potential hypothesis is that HDL modulates platelet function, a crucial player in the initiation and progression of atherothrombosis, which may contribute to its observed relationship with CVD risk. In this review, we focus on how HDL and its components, with an emphasis on APOA1, interact with platelets (and their precursors or activation products) to modulate atherothrombotic responses.
Collapse
Affiliation(s)
- Marion Pilard
- Department of Medicine, Faculty of Medicine, Université de Montréal, Canada
- Montreal Heart Institute, Canada
| | - Sara Babran
- Department of Medicine, Faculty of Medicine, Université de Montréal, Canada
- Montreal Heart Institute, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Canada
- Montreal Heart Institute, Canada
| |
Collapse
|
2
|
Gonzalez M, Clayton S, Wauson E, Christian D, Tran QK. Promotion of nitric oxide production: mechanisms, strategies, and possibilities. Front Physiol 2025; 16:1545044. [PMID: 39917079 PMCID: PMC11799299 DOI: 10.3389/fphys.2025.1545044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
The discovery of nitric oxide (NO) and the role of endothelial cells (ECs) in its production has revolutionized medicine. NO can be produced by isoforms of NO synthases (NOS), including the neuronal (nNOS), inducible (iNOS), and endothelial isoforms (eNOS), and via the non-classical nitrate-nitrite-NO pathway. In particular, endothelium-derived NO, produced by eNOS, is essential for cardiovascular health. Endothelium-derived NO activates soluble guanylate cyclase (sGC) in vascular smooth muscle cells (VSMCs), elevating cyclic GMP (cGMP), causing vasodilation. Over the past four decades, the importance of this pathway in cardiovascular health has fueled the search for strategies to enhance NO bioavailability and/or preserve the outcomes of NO's actions. Currently approved approaches operate in three directions: 1) providing exogenous NO, 2) promoting sGC activity, and 3) preventing degradation of cGMP by inhibiting phosphodiesterase 5 activity. Despite clear benefits, these approaches face challenges such as the development of nitrate tolerance and endothelial dysfunction. This highlights the need for sustainable options that promote endogenous NO production. This review will focus on strategies to promote endogenous NO production. A detailed review of the mechanisms regulating eNOS activity will be first provided, followed by a review of strategies to promote endogenous NO production based on the levels of available preclinical and clinical evidence, and perspectives on future possibilities.
Collapse
Affiliation(s)
| | | | | | | | - Quang-Kim Tran
- Department of Physiology and Pharmacology, Des Moines University Medicine and Health Sciences, West Des Moines, IA, United States
| |
Collapse
|
3
|
Hou X, Zhu M, Zhu Z, Li Y, Chen X, Zhang X. Association between platelet-to-high-density lipoprotein cholesterol ratio and future stroke risk: a national cohort study based on CHARLS. Front Neurol 2024; 15:1479245. [PMID: 39606701 PMCID: PMC11599229 DOI: 10.3389/fneur.2024.1479245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Background According to recent research, there is a considerable correlation between the severity of coronary artery disease and the platelet-to-high-density lipoprotein cholesterol ratio (PHR), which suggests that PHR is a potentially valuable inflammatory biomarker. However, the body of current research offers insufficiently strong evidence to clarify the connection between PHR and the incidence of stroke. Therefore, this study aims to elucidate any potential associations between PHR and stroke risk. Methods This study employed data from the China Health and Retirement Longitudinal Study (CHARLS) covering the period from 2011 to 2018. It included 5,872 participants who did not have a history of stroke in 2011. These patients were separated into four groups according to their baseline PHR quartiles. The main goal of the study was to focus on stroke outcomes. Stroke was defined as an occurrence of a cerebrovascular accident confirmed by a physician. We employed Cox proportional hazards regression models to investigate the association between PHR and the likelihood of experiencing a stroke. Furthermore, we conducted restricted cubic spline regression analysis and subgroup analysis. Results The average follow-up period was 77.5 months, during which 390 participants experienced a stroke. In comparison to the lowest quartile group, participants in the highest quartile of PHR had a 49% increased risk of stroke (HR 1.49, 95% CI 1.13-1.96, p = 0.004). The adjusted multivariable Cox regression analysis maintained the statistical significance of this association (aHR 1.42, 95% CI 1.06-1.90, p = 0.019). After adjustment, a positive linear relationship between stroke risk and PHR was identified through restricted cubic spline regression analysis (nonlinear p > 0.05). Additionally, the impact of stroke was consistent across a variety of subgroups, as evidenced by subgroup analysis. Conclusion Our study indicates that higher PHR levels are significantly associated with an increased risk of stroke and that these levels can be used to identify groups that are at high risk of stroke.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaohong Zhang
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
4
|
Engin A. Endothelial Dysfunction in Obesity and Therapeutic Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:489-538. [PMID: 39287863 DOI: 10.1007/978-3-031-63657-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Parallel to the increasing prevalence of obesity in the world, the mortality from cardiovascular disease has also increased. Low-grade chronic inflammation in obesity disrupts vascular homeostasis, and the dysregulation of adipocyte-derived endocrine and paracrine effects contributes to endothelial dysfunction. Besides the adipose tissue inflammation, decreased nitric oxide (NO)-bioavailability, insulin resistance (IR), and oxidized low-density lipoproteins (oxLDLs) are the main factors contributing to endothelial dysfunction in obesity and the development of cardiorenal metabolic syndrome. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in the profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Higher stiffness parameter β, increased oxidative stress, upregulation of pro-inflammatory cytokines, and nicotinamide adenine dinucleotide phosphate (NADP) oxidase in PVAT turn the macrophages into pro-atherogenic phenotypes by oxLDL-induced adipocyte-derived exosome-macrophage crosstalk and contribute to the endothelial dysfunction. In clinical practice, carotid ultrasound, higher leptin levels correlate with irisin over-secretion by human visceral and subcutaneous adipose tissues, and remnant cholesterol (RC) levels predict atherosclerotic disease in obesity. As a novel therapeutic strategy for cardiovascular protection, liraglutide improves vascular dysfunction by modulating a cyclic adenosine monophosphate (cAMP)-independent protein kinase A (PKA)-AMP-activated protein kinase (AMPK) pathway in PVAT in obese individuals. Because the renin-angiotensin-aldosterone system (RAAS) activity, hyperinsulinemia, and the resultant IR play key roles in the progression of cardiovascular disease in obesity, RAAS-targeted therapies contribute to improving endothelial dysfunction. By contrast, arginase reciprocally inhibits NO formation and promotes oxidative stress. Thus, targeting arginase activity as a key mediator in endothelial dysfunction has therapeutic potential in obesity-related vascular comorbidities. Obesity-related endothelial dysfunction plays a pivotal role in the progression of type 2 diabetes (T2D). The peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone (thiazolidinedione), is a popular drug for treating diabetes; however, it leads to increased cardiovascular risk. Selective sodium-glucose co-transporter-2 (SGLT-2) inhibitor empagliflozin (EMPA) significantly improves endothelial dysfunction and mortality occurring through redox-dependent mechanisms. Although endothelial dysfunction and oxidative stress are alleviated by either metformin or EMPA, currently used drugs to treat obesity-related diabetes neither possess the same anti-inflammatory potential nor simultaneously target endothelial cell dysfunction and obesity equally. While therapeutic interventions with glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide or bariatric surgery reverse regenerative cell exhaustion, support vascular repair mechanisms, and improve cardiometabolic risk in individuals with T2D and obesity, the GLP-1 analog exendin-4 attenuates endothelial endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
5
|
Corona DG, Vena W, Pizzocaro A, Rastrelli G, Sparano C, Sforza A, Vignozzi L, Maggi M. Metabolic syndrome and erectile dysfunction: a systematic review and meta-analysis study. J Endocrinol Invest 2023; 46:2195-2211. [PMID: 37515706 DOI: 10.1007/s40618-023-02136-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/09/2023] [Indexed: 07/31/2023]
Abstract
PURPOSE The clinical significance of metabolic syndrome (MetS) versus its single components in erectile dysfunction (ED) is conflicting. Thus, the purpose is to analyze the available evidence on the relationship between MetS-along with its components-and ED. METHODS All prospective and retrospective observational studies reporting information on ED and MetS were included. In addition, we here reanalyzed preclinical and clinical data obtained from a previously published animal model of MetS and from a consecutive series of more than 2697 men (mean age: 52.7 ± 12), respectively. RESULTS Data derived from this meta-analysis showed that MetS was associated with an up to fourfold increased risk of ED when either unadjusted or adjusted data were considered. Meta-regression analysis, performed using unadjusted statistics, showed that the MetS-related risk of ED was closely associated with all the MetS components. These associations were confirmed when unadjusted analyses from clinical models were considered. However, fully adjusted data showed that MetS-associated ED was more often due to morbidities included (or not) in the algorithm than to the MetS diagnostic category itself. MetS is also associated with low testosterone, but its contribution to MetS-associated ED-as derived from preclinical and clinical models-although independent, is marginal. CONCLUSIONS The results of our analysis suggest that MetS is a useless diagnostic category for studying ED. However, treating the individual MetS components is important, because they play a pivotal role in determining ED.
Collapse
Affiliation(s)
- D G Corona
- Endocrinology Unit, Azienda AUSL Bologna, Largo Nigrisoli 2, 40133, Bologna, Italy.
| | - W Vena
- Unit of Endocrinology, Diabetology and Medical Andrology, IRCSS, Humanitas Research Hospital, Rozzano, Milan, Italy
| | - A Pizzocaro
- Unit of Endocrinology, Diabetology and Medical Andrology, IRCSS, Humanitas Research Hospital, Rozzano, Milan, Italy
| | - G Rastrelli
- Andrology, Women's Endocrinology and Gender Incongruence Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - C Sparano
- Endocrinology Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - A Sforza
- Endocrinology Unit, Azienda AUSL Bologna, Largo Nigrisoli 2, 40133, Bologna, Italy
| | - L Vignozzi
- Andrology, Women's Endocrinology and Gender Incongruence Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - M Maggi
- Endocrinology Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
6
|
Effects of Antirheumatic Treatment on Cell Cholesterol Efflux and Loading Capacity of Serum Lipoproteins in Spondylarthropathies. J Clin Med 2022; 11:jcm11247330. [PMID: 36555946 PMCID: PMC9780876 DOI: 10.3390/jcm11247330] [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: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Spondyloarthropathies (SpA) are associated with increased cardiovascular risk. Among possible mechanisms is the dysfunction of serum lipoproteins in regulating cell cholesterol homeostasis. Cholesterol efflux capacity (CEC)-the atheroprotective ability of HDL (high density lipoproteins) to accept cholesterol from macrophages-might predict cardiovascular disease independently of HDL-cholesterol levels. We aimed at evaluating modifications of CEC and of the atherogenic cholesterol loading capacity (CLC) of serum lipoproteins in psoriatic arthritis (PsA) and ankylosing spondylitis (AS) following anti-rheumatic treatment. A total of 62 SpA patients (37 PsA and 25 AS) were evaluated before and after treatment with tumor necrosis factor inhibitor and/or methotrexate. CEC and CLC were measured by radioisotopic and fluorometric techniques, respectively. Endothelial function was assessed by finger plethysmography (Endopat). In the whole SpA group, total and HDL-cholesterol increased after treatment, while lipoprotein(a) decreased and CLC was unchanged. Treatment was associated with increased Scavenger Receptor class B type I (SR-BI)-mediated CEC in the AS group. SR-BI- and ABCG1-mediated CEC were negatively associated with inflammatory parameters and positively related to coffee consumption. SR-BI CEC and CLC were positively and negatively associated with endothelial function, respectively. Our pilot study suggests that anti-rheumatic treatment is associated with favorable modulation of lipoprotein quality and function in SpA, particularly in AS, in spite of the induced increase in total cholesterol levels. If confirmed in a larger population, this might represent an atheroprotective benefit beyond what is reflected by conventional serum lipid profile.
Collapse
|
7
|
Liu JD, Gong R, Zhang SY, Zhou ZP, Wu YQ. Beneficial effects of high-density lipoprotein (HDL) on stent biocompatibility and the potential value of HDL infusion therapy following percutaneous coronary intervention. Medicine (Baltimore) 2022; 101:e31724. [PMID: 36397406 PMCID: PMC9666103 DOI: 10.1097/md.0000000000031724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Several epidemiological studies have shown a clear inverse relationship between serum levels of high-density lipoprotein cholesterol (HDL-C) and the risk of atherosclerotic cardiovascular disease (ASCVD), even at low-density lipoprotein cholesterol levels below 70 mg/dL. There is much evidence from basic and clinical studies that higher HDL-C levels are beneficial, whereas lower HDL-C levels are detrimental. Thus, HDL is widely recognized as an essential anti-atherogenic factor that plays a protective role against the development of ASCVD. Percutaneous coronary intervention is an increasingly common treatment choice to improve myocardial perfusion in patients with ASCVD. Although drug-eluting stents have substantially overcome the limitations of conventional bare-metal stents, there are still problems with stent biocompatibility, including delayed re-endothelialization and neoatherosclerosis, which cause stent thrombosis and in-stent restenosis. According to numerous studies, HDL not only protects against the development of atherosclerosis, but also has many anti-inflammatory and vasoprotective properties. Therefore, the use of HDL as a therapeutic target has been met with great interest. Although oral medications have not shown promise, the developed HDL infusions have been tested in clinical trials and have demonstrated viability and reproducibility in increasing the cholesterol efflux capacity and decreasing plasma markers of inflammation. The aim of the present study was to review the effect of HDL on stent biocompatibility in ASCVD patients following implantation and discuss a novel therapeutic direction of HDL infusion therapy that may be a promising candidate as an adjunctive therapy to improve stent biocompatibility following percutaneous coronary intervention.
Collapse
Affiliation(s)
- Jian-Di Liu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ren Gong
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shi-Yuan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhi-Peng Zhou
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yan-Qing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- * Correspondence: Yan-Qing Wu, Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, Jiangxi 330006, China (e-mail: )
| |
Collapse
|
8
|
Oxidised Low-Density Lipoprotein-Induced Platelet Hyperactivity—Receptors and Signalling Mechanisms. Int J Mol Sci 2022; 23:ijms23169199. [PMID: 36012465 PMCID: PMC9409144 DOI: 10.3390/ijms23169199] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Dyslipidaemia leads to proatherogenic oxidative lipid stress that promotes vascular inflammation and thrombosis, the pathologies that underpin myocardial infarction, stroke, and deep vein thrombosis. These prothrombotic states are driven, at least in part, by platelet hyperactivity, and they are concurrent with the appearancxe of oxidatively modified low-density lipoproteins (LDL) in the circulation. Modified LDL are heterogenous in nature but, in a general sense, constitute a prototype circulating transporter for a plethora of oxidised lipid epitopes that act as danger-associated molecular patterns. It is well-established that oxidatively modified LDL promote platelet activation and arterial thrombosis through a number of constitutively expressed scavenger receptors, which transduce atherogenic lipid stress to a complex array of proactivatory signalling pathways in the platelets. Stimulation of these signalling events underlie the ability of modified LDL to induce platelet activation and blunt platelet inhibitory pathways, as well as promote platelet-mediated coagulation. Accumulating evidence from patients at risk of arterial thrombosis and experimental animal models of disease suggest that oxidised LDL represents a tangible link between the dyslipidaemic environment and increased platelet activation. The aim of this review is to summarise recent advances in our understanding of the pro-thrombotic signalling events induced in platelets by modified LDL ligation, describe the contribution of individual platelet scavenger receptors, and highlight potential future challenges of targeting these pathways.
Collapse
|
9
|
Indirect mediators of systemic health outcomes following nanoparticle inhalation exposure. Pharmacol Ther 2022; 235:108120. [PMID: 35085604 PMCID: PMC9189040 DOI: 10.1016/j.pharmthera.2022.108120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023]
Abstract
The growing field of nanoscience has shed light on the wide diversity of natural and anthropogenic sources of nano-scale particulates, raising concern as to their impacts on human health. Inhalation is the most robust route of entry, with nanoparticles (NPs) evading mucociliary clearance and depositing deep into the alveolar region. Yet, impacts from inhaled NPs are evident far outside the lung, particularly on the cardiovascular system and highly vascularized organs like the brain. Peripheral effects are partly explained by the translocation of some NPs from the lung into the circulation; however, other NPs largely confined to the lung are still accompanied by systemic outcomes. Omic research has only just begun to inform on the complex myriad of molecules released from the lung to the blood as byproducts of pulmonary pathology. These indirect mediators are diverse in their molecular make-up and activity in the periphery. The present review examines systemic outcomes attributed to pulmonary NP exposure and what is known about indirect pathological mediators released from the lung into the circulation. Further focus was directed to outcomes in the brain, a highly vascularized region susceptible to acute and longer-term outcomes. Findings here support the need for big-data toxicological studies to understand what drives these health outcomes and better predict, circumvent, and treat the potential health impacts arising from NP exposure scenarios.
Collapse
|
10
|
Shearston K, Tan JTM, Cochran BJ, Rye KA. Inhibition of Vascular Inflammation by Apolipoprotein A-IV. Front Cardiovasc Med 2022; 9:901408. [PMID: 35845068 PMCID: PMC9279673 DOI: 10.3389/fcvm.2022.901408] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
Background Apolipoprotein (apo) A-IV, the third most abundant apolipoprotein in human high density lipoproteins (HDLs), inhibits intestinal and systemic inflammation. This study asks if apoA-IV also inhibits acute vascular inflammation. Methods Inflammation was induced in New Zealand White rabbits by placing a non-occlusive silastic collar around the common carotid artery. A single 1 mg/kg intravenous infusion of lipid-free apoA-IV or saline (control) was administered to the animals 24 h before collar insertion. The animals were euthanised 24 h post-collar insertion. Human coronary artery cells (HCAECs) were pre-incubated with reconstituted HDLs containing apoA-IV complexed with phosphatidylcholine, (A-IV)rHDLs, then activated by incubation with tumour necrosis factor (TNF)-α. Cell surface vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) in the TNF-α-activated HCAECs was quantified by flow cytometry. VCAM-1, ICAM-1 and 3β-hydroxysteroid-Δ24 reductase (DHCR24) mRNA levels were quantified by real time PCR. Results Apolipoprotein ApoA-IV treatment significantly decreased collar-induced endothelial expression of VCAM-1, ICAM-1 and neutrophil infiltration into the arterial intima by 67.6 ± 9.9% (p < 0.01), 75.4 ± 6.9% (p < 0.01) and 74.4 ± 8.5% (p < 0.05), respectively. It also increased endothelial expression of DHCR24 by 2.6-fold (p < 0.05). Pre-incubation of HCAECs with (A-IV)rHDLs prior to stimulation with TNF-α inhibited VCAM-1 and ICAM-1 protein levels by 62.2 ± 12.1% and 33.7 ± 5.7%, respectively. VCAM-1 and ICAM-1 mRNA levels were decreased by 55.8 ± 7.2% and 49.6 ± 7.9%, respectively, while DHCR24 mRNA expression increased by threefold. Transfection of HCAECs with DHCR24 siRNA attenuated the anti-inflammatory effects of (A-IV)rHDLs. Pre-incubation of TNF-α-activated HCAECs with (A-IV)rHDLs also inhibited nuclear translocation of the p65 subunit of nuclear factor-κB (NF-κB), and decreased IκBα phosphorylation. Conclusion These results indicate that apoA-IV inhibits vascular inflammation in vitro and in vivo by inhibiting NF-κB activation in a DHCR24-dependent manner.
Collapse
Affiliation(s)
- Kate Shearston
- Lipid Research Group, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Joanne T. M. Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Blake J. Cochran
- Lipid Research Group, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- *Correspondence: Kerry-Anne Rye,
| |
Collapse
|
11
|
Strahlhofer-Augsten M, Schliefsteiner C, Cvitic S, George M, Lang-Olip I, Hirschmugl B, Marsche G, Lang U, Novakovic B, Saffery R, Desoye G, Wadsack C. The Distinct Role of the HDL Receptor SR-BI in Cholesterol Homeostasis of Human Placental Arterial and Venous Endothelial Cells. Int J Mol Sci 2022; 23:ijms23105364. [PMID: 35628180 PMCID: PMC9141204 DOI: 10.3390/ijms23105364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022] Open
Abstract
As opposed to adults, high-density lipoprotein (HDL) is the main cholesterol carrying lipoprotein in fetal circulation. The major HDL receptor, scavenger receptor class B type I (SR-BI), contributes to local cholesterol homeostasis. Arterial endothelial cells (ECA) from human placenta are enriched with cholesterol compared to venous endothelial cells (ECV). Moreover, umbilical venous and arterial plasma cholesterol levels differ markedly. We tested the hypothesis that the uptake of HDL-cholesteryl esters differs between ECA and ECV because of the differential expression of SR-BI. We aimed to identify the key regulators underlying these differences and the functional consequences. Immunohistochemistry was used for visualization of SR-BI in situ. ECA and ECV were isolated from the chorionic plate of human placenta and used for RT-qPCR, Western Blot, and HDL uptake assays with 3H- and 125I-labeled HDL. DNA was extracted for the methylation profiling of the SR-BI promoter. SR-BI regulation was studied by exposing ECA and ECV to differential oxygen concentrations or shear stress. Our results show elevated SR-BI expression and protein abundance in ECA compared to ECV in situ and in vitro. Immunohistochemistry demonstrated that SR-BI is mainly expressed on the apical side of placental endothelial cells in situ, allowing interaction with mature HDL circulating in the fetal blood. This was functionally linked to a higher increase of selective cholesterol ester uptake from fetal HDL in ECA than in ECV, and resulted in increased cholesterol availability in ECA. SR-BI expression on ECV tended to decrease with shear stress, which, together with heterogeneous immunostaining, suggests that SR-BI expression is locally regulated in the placental vasculature. In addition, hypomethylation of several CpG sites within the SR-BI promoter region might contribute to differential expression of SR-BI between chorionic arteries and veins. Therefore, SR-BI contributes to a local cholesterol homeostasis in ECA and ECV of the human feto-placental vasculature.
Collapse
Affiliation(s)
- Manuela Strahlhofer-Augsten
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- BioBank Graz, Medical University of Graz, 8036 Graz, Austria
| | - Carolin Schliefsteiner
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Silvija Cvitic
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Meekha George
- Otto Loewi Research Center, Division of Pathophysiology and Immunology, Medical University of Graz, 8010 Graz, Austria;
| | - Ingrid Lang-Olip
- Gottfried Schatz Research Center, Divison of Cell Biology, Histology and Embryology, Medical University of Graz, 8036 Graz, Austria;
| | - Birgit Hirschmugl
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria;
| | - Uwe Lang
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Boris Novakovic
- Molecular Immunity, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (B.N.); (R.S.)
| | - Richard Saffery
- Molecular Immunity, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (B.N.); (R.S.)
| | - Gernot Desoye
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Christian Wadsack
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- Correspondence:
| |
Collapse
|
12
|
Ma S, Zhang M, Qu H, Cheng Y, Du S, Fan J, Yao Q, Zhang X, Chen M, Zhang N, Shi K, Huang Y, Zhan S. Combination of High-Density Lipoprotein Cholesterol and Lipoprotein(a) as a Predictor of Collateral Circulation in Patients With Severe Unilateral Internal Carotid Artery Stenosis or Occlusion. J Clin Neurol 2022; 18:14-23. [PMID: 35021272 PMCID: PMC8762497 DOI: 10.3988/jcn.2022.18.1.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Collateral circulation is considered an important factor affecting the risk of stroke, but the factors that affect collateral circulation remain unclear. This study was performed to identify the factors associated with collateral circulation, especially blood lipids. METHODS The study involved patients who had undergone digital subtraction angiography and were confirmed as having severe unilateral stenosis or occlusion of the internal carotid artery (ICA). We classified the collateral circulation status of each patient as good (Grade 3 or 4) or poor (Grade 0, 1, or 2) according to the grading system of the American Society of Interventional and Therapeutic Neuroradiology/American Society of Interventional Radiology. We collected data on patients' characteristics and identified the factors that affect collateral circulation. RESULTS This study included 212 patients. The multivariate logistic regression analysis showed that the high-density lipoprotein cholesterol (HDL-C) concentration and a complete anterior half of the circle of Willis were independent protective factors for good collateral circulation, whereas elevated lipoprotein(a) [Lp(a)] and serum creatinine concentrations were independent risk factors for good collateral circulation. The area under the receiver operating characteristics curve (AUC) was 0.68 (95% confidence interval [CI], 0.61-0.76) for HDL-C and 0.69 (95% CI, 0.62-0.76) for Lp(a). A binary logistic regression model analysis of the joint factor of HDL-C and Lp(a) yielded an AUC of 0.77 (95% CI, 0.71-0.84). CONCLUSIONS In patients with severe unilateral ICA stenosis or occlusion, the combination of HDL-C and Lp(a) is a useful predictor of collateral circulation.
Collapse
Affiliation(s)
- Shuyin Ma
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meijuan Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huiyang Qu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuxuan Cheng
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuang Du
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiaxin Fan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qingling Yao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaodong Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengying Chen
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Nan Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kaili Shi
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yizhou Huang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuqin Zhan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
13
|
HDL and Endothelial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:27-47. [DOI: 10.1007/978-981-19-1592-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
14
|
Kotlyarov S. Diversity of Lipid Function in Atherogenesis: A Focus on Endothelial Mechanobiology. Int J Mol Sci 2021; 22:11545. [PMID: 34768974 PMCID: PMC8584259 DOI: 10.3390/ijms222111545] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is one of the most important problems in modern medicine. Its high prevalence and social significance determine the need for a better understanding of the mechanisms of the disease's development and progression. Lipid metabolism and its disorders are one of the key links in the pathogenesis of atherosclerosis. Lipids are involved in many processes, including those related to the mechanoreception of endothelial cells. The multifaceted role of lipids in endothelial mechanobiology and mechanisms of atherogenesis are discussed in this review. Endothelium is involved in ensuring adequate vascular hemodynamics, and changes in blood flow characteristics are detected by endothelial cells and affect their structure and function.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
15
|
Xu ZH, Xu HX, Jiang S, Xu QF, Ding K, Zhang DX, Guan Y, Zhao ST. Effect of high-density lipoprotein on penile erection: A cross-sectional study. Andrologia 2021; 53:e13979. [PMID: 33774838 DOI: 10.1111/and.13979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/27/2020] [Accepted: 01/04/2021] [Indexed: 01/15/2023] Open
Abstract
Previous studies have shown that elevated levels of high-density lipoprotein (HDL) could inhibit penile erection, but the relationship between HDL and the erection of the penile tip or base has not been extensively researched. We investigated the effects of HDL on erection of the penile tip and base through a cross-sectional study of 113 patients with erectile dysfunction, using a cut-off score of ≤21 on the International Index of Erectile Function-5. The following patient data were collected: nocturnal penile tumescence; blood pressure; platelet count; platelet distribution width; mean platelet volume; plateletcrit; and levels of serum glucose, total cholesterol, triglyceride, HDL, and low-density lipoprotein. Univariate and multivariate analyses were used to assess the association between HDL levels and the erection of the penile tip and base. We confirmed that HDL had a beneficial effect on penile erectile function. We also found that when the HDL level exceeded the normal range, the change in HDL had a significant effect on the penile base. In addition, our study did not find any relationship between platelet parameters and erection of the penile tip or penile base.
Collapse
Affiliation(s)
- Zhi-He Xu
- School of Medicine, Shandong University, Jinan, China.,Department of Urology, Shandong Provincial Hospital, Jinan, China.,Department of Urology, The Second Hospital of Shandong University, Jinan, China
| | | | - Shan Jiang
- School of Medicine, Shandong University, Jinan, China.,Department of Urology, Shandong Provincial Hospital, Jinan, China
| | - Qin-Feng Xu
- School of Medicine, Shandong University, Jinan, China.,Department of Urology, Shandong Provincial Hospital, Jinan, China
| | - Kun Ding
- School of Medicine, Shandong University, Jinan, China.,Department of Urology, Shandong Provincial Hospital, Jinan, China
| | - Dong-Xiang Zhang
- School of Medicine, Shandong University, Jinan, China.,Department of Urology, Shandong Provincial Hospital, Jinan, China
| | - Yong Guan
- School of Medicine, Shandong University, Jinan, China.,Department of Urology, Shandong Provincial Hospital, Jinan, China
| | | |
Collapse
|
16
|
Effects of lipoproteins on endothelial cells and macrophages function and its possible implications on fetal adverse outcomes associated to maternal hypercholesterolemia during pregnancy. Placenta 2021; 106:79-87. [PMID: 33706211 DOI: 10.1016/j.placenta.2021.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/03/2021] [Accepted: 02/25/2021] [Indexed: 11/23/2022]
Abstract
Hypercholesterolemia is one of the main risk factors associated with atherosclerosis and cardiovascular disease, the leading cause of death worldwide. During pregnancy, maternal hypercholesterolemia develops, and it can occur in a physiological (MPH) or supraphysiological (MSPH) manner, where MSPH is associated with endothelial dysfunction and early atherosclerotic lesions in the fetoplacental vasculature. In the pathogenesis of atherosclerosis, endothelial activation and endothelial dysfunction, characterized by an imbalance in the bioavailability of nitric oxide, contribute to the early stages of this disease. Macrophages conversion to foam cells, cholesterol efflux from these cells and its differentiation into a pro- or anti-inflammatory phenotype are also important processes that contribute to atherosclerosis. In adults it has been reported that native and modified HDL and LDL play an important role in endothelial and macrophage function. In this review it is proposed that fetal lipoproteins could be also relevant factors involved in the detrimental vascular effects described in MSPH. Changes in the composition and function of neonatal lipoproteins compared to adults has been reported and, although in MSPH pregnancies the fetal lipid profile does not differ from MPH, differences in the lipidomic profiles of umbilical venous blood have been reported, which could have implications in the vascular function. In this review we summarize the available information regarding the effects of lipoproteins on endothelial and macrophage function, emphasizing its possible implications on fetal adverse outcomes associated to maternal hypercholesterolemia during pregnancy.
Collapse
|
17
|
Potje SR, Paula TDC, Paulo M, Bendhack LM. The Role of Glycocalyx and Caveolae in Vascular Homeostasis and Diseases. Front Physiol 2021; 11:620840. [PMID: 33519523 PMCID: PMC7838704 DOI: 10.3389/fphys.2020.620840] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
This review highlights recent findings about the role that endothelial glycocalyx and caveolae play in vascular homeostasis. We describe the structure, synthesis, and function of glycocalyx and caveolae in vascular cells under physiological and pathophysiological conditions. Special focus will be given in glycocalyx and caveolae that are associated with impaired production of nitric oxide (NO) and generation of reactive oxygen species (ROS). Such alterations could contribute to the development of cardiovascular diseases, such as atherosclerosis, and hypertension.
Collapse
Affiliation(s)
- Simone Regina Potje
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Tiago Dal-Cin Paula
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Michele Paulo
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Lusiane Maria Bendhack
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
18
|
High-Density Lipoprotein-Targeted Therapies for Heart Failure. Biomedicines 2020; 8:biomedicines8120620. [PMID: 33339429 PMCID: PMC7767106 DOI: 10.3390/biomedicines8120620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 02/08/2023] Open
Abstract
The main and common constituents of high-density lipoproteins (HDLs) are apolipoprotein A-I, cholesterol, and phospholipids. Biochemical heterogeneity of HDL particles is based on the variable presence of one or more representatives of at least 180 proteins, 200 lipid species, and 20 micro RNAs. HDLs are circulating multimolecular platforms that perform divergent functions whereby the potential of HDL-targeted interventions for treatment of heart failure can be postulated based on its pleiotropic effects. Several murine studies have shown that HDLs exert effects on the myocardium, which are completely independent of any impact on coronary arteries. Overall, HDL-targeted therapies exert a direct positive lusitropic effect on the myocardium, inhibit the development of cardiac hypertrophy, suppress interstitial and perivascular myocardial fibrosis, increase capillary density in the myocardium, and prevent the occurrence of heart failure. In four distinct murine models, HDL-targeted interventions were shown to be a successful treatment for both pre-existing heart failure with reduced ejection fraction (HFrEF) and pre-existing heart failure with preserved ejection fraction (HFrEF). Until now, the effect of HDL-targeted interventions has not been evaluated in randomized clinical trials in heart failure patients. As HFpEF represents an important unmet therapeutic need, this is likely the preferred therapeutic domain for clinical translation.
Collapse
|
19
|
Raz BD, Dimitry C, Andrea SS. The uptake mechanism and intracellular fate of Paraoxonase-1 in endothelial cells. Free Radic Biol Med 2020; 153:26-33. [PMID: 32244050 DOI: 10.1016/j.freeradbiomed.2020.03.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022]
Abstract
Paraoxonase-1 (PON1) is a high-density lipoprotein (HDL)-associated lactonase that plays a significant role in the anti-atherosclerotic activity of HDL. However, several studies have shown that PON1 localizes in cells, where it operates independently of HDL. Previously, we showed that PON1 localizes in endothelial cells (ECs), and impairs vasodilation mediated by the endothelium-derived hyperpolarizing factor (EDHF) 5,6-δ-DHTL. However, the internalization pathway of PON1 into ECs, and the intracellular fate of PON1 are unknown. Therefore, the present study aimed to elucidate the uptake mechanism, intracellular trafficking and the function of PON1 in ECs. We conducted a series of inhibition experiments of fluorescently labeled recombinant PON1 (rePON1) in ECs, followed by FACS analyses. We found that rePON1 binds the EC membrane via specific binding sites located in lipid-rafts/caveolae microdomains that are shared with HDL, and internalized through dynamin-dependent endocytosis. Qualitative assessments of the intracellular trafficking of rePON1, using confocal z-stack images, showed colocalization of the labeled rePON1 with early and late endosome/lysosome markers. Accordingly, a "pulse-chase" incubation of rePON1, followed by lactonase activity measurement in EC lysate, revealed that rePON1 retains its lactonase activity after binding to the cells. However, this activity decreases over time. Finally, induction of endothelial dysfunction with high glucose, angiotensin II, or palmitic acid increased rePON1 uptake by ECs. In conclusion, these results indicate that free PON1 interacts with ECs via binding sites located in lipid-rafts/caveolae, where it is enzymatically active and regulates endothelial functions. However, once internalized, PON1 is degraded. Additionally, alteration in endothelial function affects PON1 uptake by ECs.
Collapse
Affiliation(s)
- Ben-David Raz
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | | | - Szuchman-Sapir Andrea
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel.
| |
Collapse
|
20
|
It takes more than two to tango: mechanosignaling of the endothelial surface. Pflugers Arch 2020; 472:419-433. [PMID: 32239285 PMCID: PMC7165135 DOI: 10.1007/s00424-020-02369-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
The endothelial surface is a highly flexible signaling hub which is able to sense the hemodynamic forces of the streaming blood. The subsequent mechanosignaling is basically mediated by specific structures, like the endothelial glycocalyx building the top surface layer of endothelial cells as well as mechanosensitive ion channels within the endothelial plasma membrane. The mechanical properties of the endothelial cell surface are characterized by the dynamics of cytoskeletal proteins and play a key role in the process of signal transmission from the outside (lumen of the blood vessel) to the interior of the cell. Thus, the cell mechanics directly interact with the function of mechanosensitive structures and ion channels. To precisely maintain the vascular tone, a coordinated functional interdependency between endothelial cells and vascular smooth muscle cells is necessary. This is given by the fact that mechanosensitive ion channels are expressed in both cell types and that signals are transmitted via autocrine/paracrine mechanisms from layer to layer. Thus, the outer layer of the endothelial cells can be seen as important functional mechanosensitive and reactive cellular compartment. This review aims to describe the known mechanosensitive structures of the vessel building a bridge between the important role of physiological mechanosignaling and the proper vascular function. Since mutations and dysfunction of mechanosensitive proteins are linked to vascular pathologies such as hypertension, they play a potent role in the field of channelopathies and mechanomedicine.
Collapse
|
21
|
The Role and Function of HDL in Patients with Chronic Kidney Disease and the Risk of Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21020601. [PMID: 31963445 PMCID: PMC7014265 DOI: 10.3390/ijms21020601] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health problem with steadily increasing occurrence. Significantly elevated cardiovascular morbidity and mortality have been observed in CKD. Cardiovascular diseases are the most important and frequent cause of death of CKD patients globally. The presence of CKD is related to disturbances in lipoprotein metabolism whose consequences are dyslipidemia and the accumulation of atherogenic particles. CKD not only fuels the reduction of high-density lipoprotein (HDL) cholesterol concentration, but also it modifies the composition of this lipoprotein. The key role of HDL is the participation in reverse cholesterol transport from peripheral tissues to the liver. Moreover, HDL prevents the oxidation of low-density lipoprotein (LDL) cholesterol by reactive oxygen species (ROS) and protects against the adverse effects of oxidized LDL (ox-LDL) on the endothelium. Numerous studies have demonstrated the ability of HDL to promote the production of nitric oxide (NO) by endothelial cells (ECs) and to exert antiapoptotic and anti-inflammatory effects. Increasing evidence suggests that in patients with chronic inflammatory disorders, HDLs may lose important antiatherosclerotic properties and become dysfunctional. So far, no therapeutic strategy to raise HDL, or alter the ratio of HDL subfractions, has been successful in slowing the progression of CKD or reducing cardiovascular disease in patients either with or without CKD.
Collapse
|
22
|
Pirillo A, Catapano AL, Norata GD. Biological Consequences of Dysfunctional HDL. Curr Med Chem 2019; 26:1644-1664. [PMID: 29848265 DOI: 10.2174/0929867325666180530110543] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/25/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022]
Abstract
Epidemiological studies have suggested an inverse correlation between high-density lipoprotein (HDL) cholesterol levels and the risk of cardiovascular disease. HDLs promote reverse cholesterol transport (RCT) and possess several putative atheroprotective functions, associated to the anti-inflammatory, anti-thrombotic and anti-oxidant properties as well as to the ability to support endothelial physiology. The assumption that increasing HDL-C levels would be beneficial on cardiovascular disease (CVD), however, has been questioned as, in most clinical trials, HDL-C-raising therapies did not result in improved cardiovascular outcomes. These findings, together with the observations from Mendelian randomization studies showing that polymorphisms mainly or solely associated with increased HDL-C levels did not decrease the risk of myocardial infarction, shift the focus from HDL-C levels toward HDL functional properties. Indeed, HDL from atherosclerotic patients not only exhibit impaired atheroprotective functions but also acquire pro-atherogenic properties and are referred to as "dysfunctional" HDL; this occurs even in the presence of normal or elevated HDL-C levels. Pharmacological approaches aimed at restoring HDL functions may therefore impact more significantly on CVD outcome than drugs used so far to increase HDL-C levels. The aim of this review is to discuss the pathological conditions leading to the formation of dysfunctional HDL and their role in atherosclerosis and beyond.
Collapse
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.,IRCCS Multimedica, Milan, Italy
| | - Alberico Luigi Catapano
- IRCCS Multimedica, Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Danilo Norata
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia
| |
Collapse
|
23
|
Huang L, Chambliss KL, Gao X, Yuhanna IS, Behling-Kelly E, Bergaya S, Ahmed M, Michaely P, Luby-Phelps K, Darehshouri A, Xu L, Fisher EA, Ge WP, Mineo C, Shaul PW. SR-B1 drives endothelial cell LDL transcytosis via DOCK4 to promote atherosclerosis. Nature 2019; 569:565-569. [PMID: 31019307 PMCID: PMC6631346 DOI: 10.1038/s41586-019-1140-4] [Citation(s) in RCA: 224] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 03/25/2019] [Indexed: 01/15/2023]
Abstract
Atherosclerosis, which underlies life-threatening cardiovascular disorders including myocardial infarction and stroke1, is initiated by low density lipoprotein cholesterol (LDL) passage into the artery wall and engulfment by macrophages, leading to foam cell formation and lesion development2, 2, 3, 3. How circulating LDL enters the artery wall to instigate atherosclerosis is unknown. Here we show in mice that scavenger receptor, class B type 1 (SR-B1) in endothelial cells mediates LDL delivery into arteries and its accumulation by artery wall macrophages, thereby promoting atherosclerosis. LDL particles are colocalized with SR-B1 in endothelial cell intracellular vesicles in vivo, and LDL transcytosis across endothelial monolayers requires its direct binding to SR-B1 and an 8 amino acid cytoplasmic domain of the receptor that recruits the guanine nucleotide exchange factor dedicator of cytokinesis 4 (DOCK4)4. DOCK4 promotes SR-B1 internalization and LDL transport by coupling LDL binding to SR-B1 with Rac1 activation. SR-B1 and DOCK4 expression are increased in atherosclerosis-prone regions of the mouse aorta prior to lesion formation, and in human atherosclerotic versus normal arteries. These findings challenge the long-held concept that atherogenesis involves passive LDL movement across a compromised endothelial barrier. Interventions inhibiting endothelial delivery of LDL into the artery wall may represent a new therapeutic category in the battle against cardiovascular disease.
Collapse
Affiliation(s)
- Linzhang Huang
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ken L Chambliss
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaofei Gao
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ivan S Yuhanna
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Erica Behling-Kelly
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Sonia Bergaya
- Department of Medicine, New York University School of Medicine, New York, NY, USA.,Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY, USA.,Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, USA
| | - Mohamed Ahmed
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Peter Michaely
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kate Luby-Phelps
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anza Darehshouri
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward A Fisher
- Department of Medicine, New York University School of Medicine, New York, NY, USA.,Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY, USA.,Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, USA
| | - Woo-Ping Ge
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
24
|
Miki T, Miyoshi T, Kotani K, Kohno K, Asonuma H, Sakuragi S, Koyama Y, Nakamura K, Ito H. Decrease in oxidized high-density lipoprotein is associated with slowed progression of coronary artery calcification: Subanalysis of a prospective multicenter study. Atherosclerosis 2019; 283:1-6. [DOI: 10.1016/j.atherosclerosis.2019.01.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/15/2019] [Accepted: 01/22/2019] [Indexed: 12/27/2022]
|
25
|
Marin-Palma D, Sirois CM, Urcuqui-Inchima S, Hernandez JC. Inflammatory status and severity of disease in dengue patients are associated with lipoprotein alterations. PLoS One 2019; 14:e0214245. [PMID: 30901375 PMCID: PMC6430398 DOI: 10.1371/journal.pone.0214245] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/09/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The triggering of severe dengue has been associated with an exacerbated inflammatory process characterized by the production of pro-inflammatory cytokines such as IL-1β/IL-18, which are the product of inflammasome activation. Furthermore, alteration in the levels of high-density (HDL) and low-density lipoproteins (LDL) has been observed; and HDL are known to have immunomodulatory properties, including the regulation of inflammasomes. While HDL would be expected to counteract hyperactivation of the inflammasome, the relationship between HDL and dengue severity, has not previously been explored. METHODOLOGY We conducted a cross-sectional study of 30 patients with dengue and 39 healthy controls matched by sex and age. Lipid profile and levels of C-reactive protein were quantified. Serum levels of IL-1β, IL-6, IL-10, IL-18, and TNF-α, were assessed by ELISA. Expression of inflammasome-related genes in PBMC was quantified by qPCR. RESULTS Dengue patients presented an alteration in the parameters of the lipid profile, with a significant decrease in HDL levels, which was more pronounced in dengue patients with warning signs. Moreover, a decrease in the expression of the inflammasome-related genes NLRP1, NLRC4, caspase-1, IL-1β and IL-18 was observed, as well as an increase in serum levels of C-reactive protein and IL-10 in dengue patients versus healthy donors. Significant positive correlations between LDL levels and the relative expression of NLRP3, NLRC4, IL-1β and IL-18, were found. CONCLUSION The results suggest that there is a relationship between the alteration of LDL and HDL with the imbalance in the inflammatory response, which could be associated with the severity of dengue.
Collapse
Affiliation(s)
- Damariz Marin-Palma
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellin, Colombia
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Cherilyn M. Sirois
- Department of Biology & Chemistry, Springfield College, Springfield, MA, United States of America
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Juan C. Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellin, Colombia
- * E-mail:
| |
Collapse
|
26
|
Marín-Palma D, Castro GA, Cardona-Arias JA, Urcuqui-Inchima S, Hernandez JC. Lower High-Density Lipoproteins Levels During Human Immunodeficiency Virus Type 1 Infection Are Associated With Increased Inflammatory Markers and Disease Progression. Front Immunol 2018; 9:1350. [PMID: 29963050 PMCID: PMC6010517 DOI: 10.3389/fimmu.2018.01350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/31/2018] [Indexed: 01/14/2023] Open
Abstract
Introduction High-density lipoproteins (HDL) are responsible for the efflux and transport of cholesterol from peripheral tissues to the liver. In addition, HDL can modulate various immunological mechanisms, including the inflammatory response. Inflammasomes are multiprotein complexes that have been reported to be activated during human immunodeficiency virus type 1 (HIV-1) infection, thus contributing to immune hyperactivation, which is the main pathogenic mechanism of HIV-1 progression. However, the relationship between HDL and inflammasomes in the context of HIV-1 infection is unclear. Therefore, this research aims to explore the association between HDL and the components of the inflammatory response during HIV-1 infection. Methodology A cross-sectional study, including 36 HIV-1-infected individuals without antiretroviral treatment and 36 healthy controls matched by sex and age, was conducted. Viral load, CD4+ T-cell counts, serum HDL, and C-reactive protein (CRP) were quantified. Serum cytokine levels, including IL-1β, IL-6, and IL-18, were assessed by ELISA. The inflammasome-related genes in peripheral blood mononuclear cells were determined by quantitative real-time PCR. Results HIV-1-infected individuals showed a significant decrease in HDL levels, particularly those subjects with higher viral load and lower CD4+ T-cell counts. Moreover, upregulation of inflammasome-related genes (NLRP3, AIM2, ASC, IL-1β, and IL-18) was observed, notably in those HIV-1-infected individuals with higher viral loads (above 5,000 copies/mL). Serum levels of IL-6 and CRP were also elevated in HIV-1-infected individuals. Significant negative correlations between HDL and the mRNA of NLRP3, AIM2, ASC, IL-1β, and IL-18, as well as viral load and CRP were observed in HIV-1-infected individuals. Likewise, a significant positive correlation between HDL and CD4+ T-cell counts was found. Conclusion In summary, our results indicate that HDL might modulate the expression of several key components of the inflammasomes during HIV-1 infection, suggesting a novel role of HDL in modifying the inflammatory state and consequently, the progression of HIV-1 infection.
Collapse
Affiliation(s)
- Damariz Marín-Palma
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia.,Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Gustavo A Castro
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Jaiberth A Cardona-Arias
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia.,Escuela de Microbiología, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Juan C Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| |
Collapse
|
27
|
Experimental challenges regarding the in vitro investigation of the nanoparticle-biocorona in disease states. Toxicol In Vitro 2018; 51:40-49. [PMID: 29738787 DOI: 10.1016/j.tiv.2018.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/11/2018] [Accepted: 05/03/2018] [Indexed: 11/20/2022]
Abstract
Toxicological evaluation of nanoparticles (NPs) requires the utilization of in vitro techniques due to their number and diverse properties. Cell culture systems are often lacking in their ability to perform comparative toxicity assessment due to dosimetry issues and capacity to simulate in vivo environments. Upon encountering a physiological environment, NPs become coated with biomolecules forming a biocorona (BC), influencing function, biodistribution, and toxicity. Disease-induced alterations in the biological milieu can alter BC formation. This study evaluates the role of low-density lipoprotein (LDL) in altering macrophage responses to iron oxide (Fe3O4) NPs. BCs were formed by incubating Fe3O4 NPs in serum-free media, or 10% fetal bovine serum with or without LDL present. Following exposures to a normalized dose (25 μg/mL), macrophage association of Fe3O4 NPs with a LDL-BC was enhanced. TNF-α mRNA expression and protein levels were differentially induced due to BCs. Cell surface expression of SR-B1 was reduced following all Fe3O4 NPs exposures, while only NPs with an LDL-BC enhanced mitochondrial membrane potential. These findings suggest that elevations in LDL may contribute to distinct BC formation thereby influencing NP-cellular interactions and response. Further, our study highlights challenges that may arise during the in vitro evaluation of disease-related variations in the NP-BC.
Collapse
|
28
|
Marín D, Taborda NA, Urcuqui S, Hernandez JC. Inflamación y respuesta inmune innata: participación de las lipoproteínas de alta densidad. IATREIA 2017. [DOI: 10.17533/udea.iatreia.v30n4a06] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
29
|
Fung KY, Wang C, Nyegaard S, Heit B, Fairn GD, Lee WL. SR-BI Mediated Transcytosis of HDL in Brain Microvascular Endothelial Cells Is Independent of Caveolin, Clathrin, and PDZK1. Front Physiol 2017; 8:841. [PMID: 29163190 PMCID: PMC5670330 DOI: 10.3389/fphys.2017.00841] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/09/2017] [Indexed: 01/19/2023] Open
Abstract
The vascular endothelium supplying the brain exhibits very low paracellular and transcellular permeability and is a major constituent of the blood-brain barrier. High-density lipoprotein (HDL) crosses the blood-brain barrier by transcytosis, but technical limitations have made it difficult to elucidate its regulation. Using a combination of spinning-disc confocal and total internal reflection fluorescence microscopy, we examined the uptake and transcytosis of HDL by human primary brain microvascular endothelial cell monolayers. Using these approaches, we report that HDL internalization requires dynamin but not clathrin heavy chain and that its internalization and transcytosis are saturable. Internalized HDL partially co-localized with the scavenger receptor BI (SR-BI) and knockdown of SR-BI significantly attenuated HDL internalization. However, we observed that the adaptor protein PDZK1—which is critical to HDL-SR-BI signaling in other tissues—is not required for HDL uptake in these cells. Additionally, while these cells express caveolin, the abundance of caveolae in this tissue is negligible and we find that SR-BI and caveolin do not co-fractionate. Furthermore, direct silencing of caveolin-1 had no impact on the uptake of HDL. Finally, inhibition of endothelial nitric oxide synthase increased HDL internalization while increasing nitric oxide levels had no impact. Together, these data indicate that SR-BI-mediated transcytosis in brain microvascular endothelial cells is distinct from uptake and signaling pathways described for this receptor in other cell types.
Collapse
Affiliation(s)
- Karen Y Fung
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Changsen Wang
- Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Steffen Nyegaard
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Bryan Heit
- Department of Microbiology and Immunology, Centre for Human Immunology, University of Western Ontario, London, ON, Canada
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Surgery, University of Toronto, ON, Canada
| | - Warren L Lee
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Medicine and Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| |
Collapse
|
30
|
Abdo AI, Rayner BS, van Reyk DM, Hawkins CL. Low-density lipoprotein modified by myeloperoxidase oxidants induces endothelial dysfunction. Redox Biol 2017; 13:623-632. [PMID: 28818791 PMCID: PMC5558469 DOI: 10.1016/j.redox.2017.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/04/2017] [Indexed: 12/19/2022] Open
Abstract
Low-density lipoprotein (LDL) modified by hypochlorous acid (HOCl) produced by myeloperoxidase (MPO) is present in atherosclerotic lesions, where it is implicated in the propagation of inflammation and acceleration of lesion development by multiple pathways, including the induction of endothelial dysfunction. Thiocyanate (SCN-) ions are utilised by MPO to produce the oxidant hypothiocyanous acid (HOSCN), which reacts with LDL in a different manner to HOCl. Whilst the reactivity of HOCl-modified LDL has been previously studied, the role of HOSCN in the modification of LDL in vivo is poorly defined, although emerging evidence suggests that these particles have distinct biological properties. This is important because elevated plasma SCN- is linked with both the propagation and prevention of atherosclerosis. In this study, we demonstrate that both HOSCN- and HOCl-modified LDL inhibit endothelium-mediated vasorelaxation ex vivo in rat aortic ring segments. In vitro experiments with human coronary artery endothelial cells show that HOSCN-modified LDL decreases in the production of nitric oxide (NO•) and induces the loss of endothelial nitric oxide synthase (eNOS) activity. This occurs to a similar extent to that seen with HOCl-modified LDL. In each case, these effects are related to eNOS uncoupling, rather than altered expression, phosphorylation or cellular localisation. Together, these data provide new insights into role of MPO and LDL modification in the induction of endothelial dysfunction, which has implications for both the therapeutic use of SCN- within the setting of atherosclerosis and for smokers, who have elevated plasma levels of SCN-, and are more at risk of developing cardiovascular disease.
Myeloperoxidase produces HOCl and HOSCN that modify LDL in a distinct manner. HOSCN- and HOCl-LDL inhibit endothelium-mediated vasorelaxation in aortic rings ex vivo. HOSCN- and HOCl-LDL decrease endothelial production of nitric oxide in vitro. Decreased eNOS activity is seen, which associated with enzyme uncoupling. HOSCN- and HOCl-LDL induce colocalisation of eNOS and caveolin 1.
Collapse
Affiliation(s)
- Adrian I Abdo
- The Heart Research Institute, 7 Eliza St, Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Benjamin S Rayner
- The Heart Research Institute, 7 Eliza St, Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - David M van Reyk
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Clare L Hawkins
- The Heart Research Institute, 7 Eliza St, Newtown, NSW 2042, Australia; Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia; Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Blegdamsvej 3, Copenhagen N 2200, Denmark.
| |
Collapse
|
31
|
Boyce G, Button E, Soo S, Wellington C. The pleiotropic vasoprotective functions of high density lipoproteins (HDL). J Biomed Res 2017; 32:164. [PMID: 28550271 PMCID: PMC6265396 DOI: 10.7555/jbr.31.20160103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/23/2016] [Indexed: 12/19/2022] Open
Abstract
The pleiotropic functions of circulating high density lipoprotein (HDL) on peripheral vascular health are well established. HDL plays a pivotal role in reverse cholesterol transport and is also known to suppress inflammation, endothelial activation and apoptosis in peripheral vessels. Although not expressed in the central nervous system, HDL has nevertheless emerged as a potential resilience factor for dementia in multiple epidemiological studies. Animal model data specifically support a role for HDL in attenuating the accumulation of β-amyloid within cerebral vessels concomitant with reduced neuroinflammation and improved cognitive performance. As the vascular contributions to dementia are increasingly appreciated, this review seeks to summarize recent literature focused on the vasoprotective properties of HDL that may extend to cerebral vessels, discuss potential roles of HDL in dementia relative to brain-derived lipoproteins, identify gaps in current knowledge, and highlight new opportunities for research and discovery.
Collapse
Affiliation(s)
- Guilaine Boyce
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Emily Button
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sonja Soo
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Cheryl Wellington
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
32
|
Abstract
On the basis of studies that extend back to the early 1900s, regression and stabilization of atherosclerosis in humans has progressed from being a concept to one that is achievable. Successful attempts at regression generally applied robust measures to improve plasma lipoprotein profiles. Possible mechanisms responsible for lesion shrinkage include decreased retention of atherogenic apolipoprotein B within the arterial wall, efflux of cholesterol and other toxic lipids from plaques, emigration of lesional foam cells out of the arterial wall, and influx of healthy phagocytes that remove necrotic debris as well as other components of the plaque. Currently available clinical agents, however, still fail to stop most cardiovascular events. For years, HDL has been considered the 'good cholesterol.' Clinical intervention studies to causally link plasma HDL-C levels to decreased progression or to the regression of atherosclerotic plaques are relatively few because of the lack of therapeutic agents that can selectively and potently increase HDL-C. The negative results of studies that were carried out have led to uncertainty as to the role that HDL plays in atherosclerosis. It is becoming clearer, however, that HDL function rather than quantity is most crucial and, therefore, discovery of agents that enhance the quality of HDL should be the goal.
Collapse
|
33
|
Abstract
Chronic inflammatory state in obesity causes dysregulation of the endocrine and paracrine actions of adipocyte-derived factors, which disrupt vascular homeostasis and contribute to endothelial vasodilator dysfunction and subsequent hypertension. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Adipose tissue inflammation, nitric oxide (NO)-bioavailability, insulin resistance and oxidized low-density lipoprotein (oxLDL) are main participating factors in endothelial dysfunction of obesity. In this chapter, disruption of inter-endothelial junctions between endothelial cells, significant increase in the production of reactive oxygen species (ROS), inflammation mediators, which are originated from inflamed endothelial cells, the balance between NO synthesis and ROS , insulin signaling and NO production, and decrease in L-arginine/endogenous asymmetric dimethyl-L-arginine (ADMA) ratio are discussed in connection with endothelial dysfunction in obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
34
|
See Hoe LE, May LT, Headrick JP, Peart JN. Sarcolemmal dependence of cardiac protection and stress-resistance: roles in aged or diseased hearts. Br J Pharmacol 2016; 173:2966-91. [PMID: 27439627 DOI: 10.1111/bph.13552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/25/2022] Open
Abstract
Disruption of the sarcolemmal membrane is a defining feature of oncotic death in cardiac ischaemia-reperfusion (I-R), and its molecular makeup not only fundamentally governs this process but also affects multiple determinants of both myocardial I-R injury and responsiveness to cardioprotective stimuli. Beyond the influences of membrane lipids on the cytoprotective (and death) receptors intimately embedded within this bilayer, myocardial ionic homeostasis, substrate metabolism, intercellular communication and electrical conduction are all sensitive to sarcolemmal makeup, and critical to outcomes from I-R. As will be outlined in this review, these crucial sarcolemmal dependencies may underlie not only the negative effects of age and common co-morbidities on myocardial ischaemic tolerance but also the on-going challenge of implementing efficacious cardioprotection in patients suffering accidental or surgically induced I-R. We review evidence for the involvement of sarcolemmal makeup changes in the impairment of stress-resistance and cardioprotection observed with ageing and highly prevalent co-morbid conditions including diabetes and hypercholesterolaemia. A greater understanding of membrane changes with age/disease, and the inter-dependences of ischaemic tolerance and cardioprotection on sarcolemmal makeup, can facilitate the development of strategies to preserve membrane integrity and cell viability, and advance the challenging goal of implementing efficacious 'cardioprotection' in clinically relevant patient cohorts. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.
Collapse
Affiliation(s)
- Louise E See Hoe
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.,Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Queensland, Australia
| | - Lauren T May
- Monash Institute of Pharmaceutical Sciences, Monash University, Clayton, VIC, Australia
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.
| |
Collapse
|
35
|
Lipoproteins as modulators of atherothrombosis: From endothelial function to primary and secondary coagulation. Vascul Pharmacol 2016; 82:1-10. [DOI: 10.1016/j.vph.2015.10.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 12/20/2022]
|
36
|
Shannahan JH, Fritz KS, Raghavendra AJ, Podila R, Persaud I, Brown JM. From the Cover: Disease-Induced Disparities in Formation of the Nanoparticle-Biocorona and the Toxicological Consequences. Toxicol Sci 2016; 152:406-16. [PMID: 27255384 DOI: 10.1093/toxsci/kfw097] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Nanoparticle (NP) association with macromolecules in a physiological environment forms a biocorona (BC), which alters NP distribution, activity, and toxicity. While BC formation is dependent on NP physicochemical properties, little information exists on the influence of the physiological environment. Obese individuals and those with cardiovascular disease exist with altered serum chemistry, which is expected to influence BC formation and NP toxicity. We hypothesize that a BC formed on NPs following incubation in hyperlipidemic serum will result in altered NP-BC protein content, cellular association, and toxicity compared to normal serum conditions. We utilized Fe3O4 NPs, which are being developed as MRI contrast and tumor targeting agents to test our hypothesis. We used rat aortic endothelial cells (RAECs) within a dynamic flow in vitro exposure system to more accurately depict the in vivo environment. A BC was formed on 20nm PVP-suspended Fe3O4 NPs following incubation in water, 10% normal or hyperlipidemic rat serum. Addition of BCs resulted in increased hydrodynamic size and decreased surface charge. More cholesterol associated with Fe3O4 NPs after incubation in hyperlipidemic as compared with normal serum. Using quantitative proteomics, we identified unique differences in BC protein components between the 2 serum types. Under flow conditions, formation of a BC from both serum types reduced RAECs association of Fe3O4 NPs. Addition of BCs was found to exacerbate RAECs inflammatory gene responses to Fe3O4 NPs (Fe3O4-hyperlipidemic > Fe3O4-normal > Fe3O4) including increased expression of IL-6, TNF-α, Cxcl-2, VCAM-1, and ICAM-1. Overall, these findings demonstrate that disease-induced variations in physiological environments have a significant impact NP-BC formation, cellular association, and cell response.
Collapse
Affiliation(s)
- Jonathan H Shannahan
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Kristofer S Fritz
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Achyut J Raghavendra
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina, 29634; Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina, 29625
| | - Ramakrishna Podila
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina, 29634; Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina, 29625
| | - Indushekar Persaud
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Jared M Brown
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045;
| |
Collapse
|
37
|
Westerterp M, Tsuchiya K, Tattersall IW, Fotakis P, Bochem AE, Molusky MM, Ntonga V, Abramowicz S, Parks JS, Welch CL, Kitajewski J, Accili D, Tall AR. Deficiency of ATP-Binding Cassette Transporters A1 and G1 in Endothelial Cells Accelerates Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2016; 36:1328-37. [PMID: 27199450 DOI: 10.1161/atvbaha.115.306670] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 05/10/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Plasma high-density lipoproteins have several putative antiatherogenic effects, including preservation of endothelial functions. This is thought to be mediated, in part, by the ability of high-density lipoproteins to promote cholesterol efflux from endothelial cells (ECs). The ATP-binding cassette transporters A1 and G1 (ABCA1 and ABCG1) interact with high-density lipoproteins to promote cholesterol efflux from ECs. To determine the impact of endothelial cholesterol efflux pathways on atherogenesis, we prepared mice with endothelium-specific knockout of Abca1 and Abcg1. APPROACH AND RESULTS Generation of mice with EC-ABCA1 and ABCG1 deficiency required crossbreeding Abca1(fl/fl)Abcg1(fl/fl)Ldlr(-/-) mice with the Tie2Cre strain, followed by irradiation and transplantation of Abca1(fl/fl)Abcg1(fl/fl) bone marrow to abrogate the effects of macrophage ABCA1 and ABCG1 deficiency induced by Tie2Cre. After 20 to 22 weeks of Western-type diet, both single EC-Abca1 and Abcg1 deficiency increased atherosclerosis in the aortic root and whole aorta. Combined EC-Abca1/g1 deficiency caused a significant further increase in lesion area at both sites. EC-Abca1/g1 deficiency dramatically enhanced macrophage lipid accumulation in the branches of the aorta that are exposed to disturbed blood flow, decreased aortic endothelial NO synthase activity, and increased monocyte infiltration into the atherosclerotic plaque. Abca1/g1 deficiency enhanced lipopolysaccharide-induced inflammatory gene expression in mouse aortic ECs, which was recapitulated by ABCG1 deficiency in human aortic ECs. CONCLUSIONS These studies provide direct evidence that endothelial cholesterol efflux pathways mediated by ABCA1 and ABCG1 are nonredundant and atheroprotective, reflecting preservation of endothelial NO synthase activity and suppression of endothelial inflammation, especially in regions of disturbed arterial blood flow.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1/deficiency
- ATP Binding Cassette Transporter 1/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 1/deficiency
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/physiopathology
- Bone Marrow Transplantation
- Cholesterol/metabolism
- Diet, High-Fat
- Disease Models, Animal
- Disease Progression
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Genetic Predisposition to Disease
- Inflammation Mediators/metabolism
- Macrophages/metabolism
- Male
- Mice, Knockout
- Monocytes/metabolism
- Neovascularization, Physiologic
- Nitric Oxide Synthase Type III/metabolism
- Phenotype
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Regional Blood Flow
- Retinal Neovascularization/genetics
- Retinal Neovascularization/metabolism
- Time Factors
- Tissue Culture Techniques
- Whole-Body Irradiation
Collapse
Affiliation(s)
- Marit Westerterp
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.).
| | - Kyoichiro Tsuchiya
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Ian W Tattersall
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Panagiotis Fotakis
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Andrea E Bochem
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Matthew M Molusky
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Vusisizwe Ntonga
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Sandra Abramowicz
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - John S Parks
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Carrie L Welch
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Jan Kitajewski
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Domenico Accili
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Alan R Tall
- From the Division of Molecular Medicine, Department of Medicine (M.W., P.F., A.E.B., M.M.M., V.N., S.A., C.L.W., A.R.T.), Naomi Berrie Diabetes Center (K.T., D.A.), and Department of Pathology, Obstetrics, and Gynaecology (I.W.T., J.K.), Columbia University, New York, NY; Section on Molecular Genetics, Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands (M.W.); Department of Diabetes, Endocrinology, and Metabolism, Medical Hospital of Tokyo Medical and Dental University, Tokyo, Japan (K.T.); and Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| |
Collapse
|
38
|
Transcriptional and Posttranslational Regulation of eNOS in the Endothelium. ADVANCES IN PHARMACOLOGY 2016; 77:29-64. [PMID: 27451094 DOI: 10.1016/bs.apha.2016.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is a highly reactive free radical gas and these unique properties have been adapted for a surprising number of biological roles. In neurons, NO functions as a neurotransmitter; in immune cells, NO contributes to host defense; and in endothelial cells, NO is a major regulator of blood vessel homeostasis. In the vasculature, NO is synthesized on demand by a specific enzyme, endothelial nitric oxide synthase (eNOS) that is uniquely expressed in the endothelial cells that form the interface between the circulating blood and the various tissues of the body. NO regulates endothelial and blood vessel function via two distinct pathways, the activation of soluble guanylate cyclase and cGMP-dependent signaling and the S-nitrosylation of proteins with reactive thiols (S-nitrosylation). The chemical properties of NO also serve to reduce oxidation and regulate mitochondrial function. Reduced synthesis and/or compromised biological activity of NO precede the development of cardiovascular disease and this has generated a high level of interest in the mechanisms controlling the synthesis and fate of NO in the endothelium. The amount of NO produced results from the expression level of eNOS, which is regulated at the transcriptional and posttranscriptional levels as well as the acute posttranslational regulation of eNOS. The goal of this chapter is to highlight and integrate past and current knowledge of the mechanisms regulating eNOS expression in the endothelium and the posttranslational mechanisms regulating eNOS activity in both health and disease.
Collapse
|
39
|
Aragon M, Erdely A, Bishop L, Salmen R, Weaver J, Liu J, Hall P, Eye T, Kodali V, Zeidler-Erdely P, Stafflinger JE, Ottens AK, Campen MJ. MMP-9-Dependent Serum-Borne Bioactivity Caused by Multiwalled Carbon Nanotube Exposure Induces Vascular Dysfunction via the CD36 Scavenger Receptor. Toxicol Sci 2016; 150:488-98. [PMID: 26801584 PMCID: PMC4966280 DOI: 10.1093/toxsci/kfw015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inhalation of multiwalled carbon nanotubes (MWCNT) causes systemic effects including vascular inflammation, endothelial dysfunction, and acute phase protein expression. MWCNTs translocate only minimally beyond the lungs, thus cardiovascular effects thereof may be caused by generation of secondary biomolecular factors from MWCNT-pulmonary interactions that spill over into the systemic circulation. Therefore, we hypothesized that induced matrix metalloproteinase-9 (MMP-9) is a generator of factors that, in turn, drive vascular effects through ligand-receptor interactions with the multiligand pattern recognition receptor, CD36. To test this, wildtype (WT; C57BL/6) and MMP-9(-/-)mice were exposed to varying doses (10 or 40 µg) of MWCNTs via oropharyngeal aspiration and serum was collected at 4 and 24 h postexposure. Endothelial cells treated with serum from MWCNT-exposed WT mice exhibited significantly reduced nitric oxide (NO) generation, as measured by electron paramagnetic resonance, an effect that was independent of NO scavenging. Serum from MWCNT-exposed WT mice inhibited acetylcholine (ACh)-mediated relaxation of aortic rings at both time points. Absence of CD36 on the aortic rings (obtained from CD36-deficient mice) abolished the serum-induced impairment of vasorelaxation. MWCNT exposure induced MMP-9 protein levels in both bronchoalveolar lavage and whole lung lysates. Serum from MMP-9(-/-)mice exposed to MWCNT did not diminish the magnitude of vasorelaxation in naïve WT aortic rings, although a modest right shift of the ACh dose-response curve was observed in both MWCNT dose groups relative to controls. In conclusion, pulmonary exposure to MWCNT leads to elevated MMP-9 levels and MMP-9-dependent generation of circulating bioactive factors that promote endothelial dysfunction and decreased NO bioavailability via interaction with vascular CD36.
Collapse
Affiliation(s)
- Mario Aragon
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| | - Aaron Erdely
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Lindsey Bishop
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Rebecca Salmen
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - John Weaver
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| | - Jim Liu
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| | - Pamela Hall
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| | - Tracy Eye
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Vamsi Kodali
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Patti Zeidler-Erdely
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Jillian E Stafflinger
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Matthew J Campen
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| |
Collapse
|
40
|
Ousmaal MEF, Martínez MC, Andriantsitohaina R, Chabane K, Gaceb A, Mameri S, Giaimis J, Baz A. Increased monocyte/neutrophil and pro-coagulant microparticle levels and overexpression of aortic endothelial caveolin-1β in dyslipidemic sand rat, Psammomys obesus. J Diabetes Complications 2016; 30:21-9. [PMID: 26597597 DOI: 10.1016/j.jdiacomp.2015.09.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/28/2015] [Accepted: 09/30/2015] [Indexed: 01/05/2023]
Abstract
AIMS To compare the effects of a high-energy diet (HED) with those of a low-energy diet (LED) on biochemical parameters, microparticle (MP) subpopulations and endothelial caveolin-1 (cav-1) protein expression in Psammomys obesus (P. obesus). METHODS After 12weeks of feeding with either the HED or LED, fasting plasma glucose and lipid parameters were measured using an enzymatic colorimetric kit while serum insulin concentration was determined with radioimmunoassay kits. MP subpopulations and cav-1 protein expression were quantified using flow cytometry and western blot analysis, respectively. RESULTS We observed that the HED caused a marked increase in lipid parameters, even in normoglycemic P. obesus. The total number of circulating MPs and the numbers of platelet-, leukocyte-, and erythrocyte-derived MPs were unaltered in the HED group. However, the HED induced increases in the numbers of monocytes/neutrophils and procoagulant MPs and a decrease in the endothelial MP levels. Cav-1β protein expression and reactive oxygen species production were increased in the vascular endothelium of HED-treated P. obesus. CONCLUSION From these findings, it is indicated that the HED exerts deleterious effects on the vascular system by increasing the monocyte/neutrophil and procoagulant MP levels, which may lead to cav-1β protein overexpression in dyslipidemic P. obesus.
Collapse
Affiliation(s)
- Mohamed El Fadel Ousmaal
- Laboratory of Biology and Organism Physiology, University of Sciences and Technology Houari Boumediene (USTHB), Algiers, Algeria; Laboratory of Biology and Animal Physiology, ENS Kouba, Algiers, Algeria.
| | - M Carmen Martínez
- INSERM U1063- Stress Oxydant et Pathologies Métaboliques, Université d'Angers, France.
| | | | - Kahina Chabane
- Laboratory of Biology and Animal Physiology, ENS Kouba, Algiers, Algeria.
| | - Abderahim Gaceb
- INSERM U1063- Stress Oxydant et Pathologies Métaboliques, Université d'Angers, France.
| | - Saâdia Mameri
- Laboratory of Anatomopathology, Mustapha Bacha Hospital, Algiers, Algeria.
| | - Jean Giaimis
- UMR Qualisud- Faculty of Pharmacy, University of Montpellier I, Montpellier, France.
| | - Ahsene Baz
- Laboratory of Biology and Animal Physiology, ENS Kouba, Algiers, Algeria.
| |
Collapse
|
41
|
Shannahan JH, Bai W, Brown JM. Implications of scavenger receptors in the safe development of nanotherapeutics. ACTA ACUST UNITED AC 2015; 2:e811. [PMID: 26005702 DOI: 10.14800/rci.811] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nanomaterials (NMs) are being utilized in a variety of biomedical applications including drug delivery, diagnostics, and therapeutic targeting. These applications are made possible due to the unique physicochemical properties that are exhibited at the nanoscale. To ensure safe development of NMs for clinical use, it is necessary to understand their interactions with cells and specifically cell surface receptors, which will facilitate either their toxicity and/or clinical function. Recently our research and others have investigated the role of scavenger receptors in mediating NM-cell interactions and responses. Scavenger receptors are expressed by a variety of cell types that are first to encounter NMs during clinical use such as macrophages and endothelial cells. Scavenger receptors are recognized to facilitate uptake of a wide variety of ligands ranging from foreign substances to endogenous lipids/proteins. While interaction of NMs with scavenger receptors may allow therapeutic targeting in some instances, it also presents a challenge for the stealth delivery of NMs and avoidance of the scavenging capability of this class of receptors. Due to their role in facilitating immune responses, scavenger receptor-mediated inflammation is also of concern following NM delivery. The research highlighted in this brief review intends to summarize our current understanding regarding the consequences of NM-scavenger receptor interactions.
Collapse
Affiliation(s)
- Jonathan H Shannahan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Wei Bai
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Jared M Brown
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| |
Collapse
|
42
|
Dhananjayan R, Koundinya KSS, Malati T, Kutala VK. Endothelial Dysfunction in Type 2 Diabetes Mellitus. Indian J Clin Biochem 2015; 31:372-9. [PMID: 27605734 DOI: 10.1007/s12291-015-0516-y] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 07/29/2015] [Indexed: 12/13/2022]
Abstract
Endothelial dysfunction is an imbalance in the production of vasodilator factors and when this balance is disrupted, it predisposes the vasculature towards pro-thrombotic and pro-atherogenic effects. This results in vasoconstriction, leukocyte adherence, platelet activation, mitogenesis, pro-oxidation, impaired coagulation and nitric oxide production, vascular inflammation, atherosclerosis and thrombosis. Endothelial dysfunction is focussed as it is a potential contributor to the pathogenesis of vascular disease in diabetes mellitus. Under physiological conditions, there is a balanced release of endothelial-derived relaxing and contracting factors, but this delicate balance is altered in diabetes mellitus and atherosclerosis, thereby contributing to further progression of vascular and end-organ damage. This review focuses on endothelial dysfunction in atherosclerosis, insulin resistance, metabolic syndrome, oxidative stress associated with diabetes mellitus, markers and genetics that are implicated in endothelial dysfunction.
Collapse
Affiliation(s)
- R Dhananjayan
- Department of Biochemistry, ACS Medical College & Hospital, Velappanchavadi, Chennai, Tamil Nadu India
| | | | - T Malati
- Department of Biochemistry, Nizam's Institute of Medical Sciences, Hyderabad, Telangana India
| | - Vijay Kumar Kutala
- Department of Clinical Pharmacology and Therapeutics (Biochemistry), Nizam's Institute of Medical Sciences, Hyderabad, Telangana India
| |
Collapse
|
43
|
Rizzo V. The Role of Caveolae and Caveolins in Atherogenesis. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Sanon VP, Sawaki D, Mjaatvedt CH, Jourdan‐Le Saux C. Myocardial Tissue Caveolae. Compr Physiol 2015; 5:871-86. [DOI: 10.1002/cphy.c140050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
45
|
Riwanto M, Rohrer L, von Eckardstein A, Landmesser U. Dysfunctional HDL: from structure-function-relationships to biomarkers. Handb Exp Pharmacol 2015; 224:337-366. [PMID: 25522994 DOI: 10.1007/978-3-319-09665-0_10] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Reduced plasma levels of HDL-C are associated with an increased risk of CAD and myocardial infarction, as shown in various prospective population studies. However, recent clinical trials on lipid-modifying drugs that increase plasma levels of HDL-C have not shown significant clinical benefit. Notably, in some recent clinical studies, there is no clear association of higher HDL-C levels with a reduced risk of cardiovascular events observed in patients with existing CAD. These observations have prompted researchers to shift from a cholesterol-centric view of HDL towards assessing the function and composition of HDL particles. Of importance, experimental and translational studies have further demonstrated various potential antiatherogenic effects of HDL. HDL has been proposed to promote macrophage reverse cholesterol transport and to protect endothelial cell functions by prevention of oxidation of LDL and its adverse endothelial effects. Furthermore, HDL from healthy subjects can directly stimulate endothelial cell production of nitric oxide and exert anti-inflammatory and antiapoptotic effects. Of note, increasing evidence suggests that the vascular effects of HDL can be highly heterogeneous and HDL may lose important anti-atherosclerotic properties and turn dysfunctional in patients with chronic inflammatory disorders. A greater understanding of mechanisms of action of HDL and its altered vascular effects is therefore critical within the context of HDL-targeted therapies.
Collapse
Affiliation(s)
- Meliana Riwanto
- Cardiology, University Heart Center, University Hospital Zurich and Center of Molecular Cardiology, University of Zurich, Rämistrasse 100, CH 8091, Zurich, Switzerland
| | | | | | | |
Collapse
|
46
|
Mancini A, Serrano-Díaz J, Nava E, D'Alessandro AM, Alonso GL, Carmona M, Llorens S. Crocetin, a Carotenoid Derived from Saffron ( Crocus sativus L.), Improves Acetylcholine-Induced Vascular Relaxation in Hypertension. J Vasc Res 2014; 51:393-404. [DOI: 10.1159/000368930] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/30/2014] [Indexed: 11/19/2022] Open
|
47
|
Shannahan JH, Podila R, Aldossari AA, Emerson H, Powell BA, Ke PC, Rao AM, Brown JM. Formation of a protein corona on silver nanoparticles mediates cellular toxicity via scavenger receptors. Toxicol Sci 2014; 143:136-46. [PMID: 25326241 DOI: 10.1093/toxsci/kfu217] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Addition of a protein corona (PC) or protein adsorption layer on the surface of nanomaterials following their introduction into physiological environments may modify their activity, bio-distribution, cellular uptake, clearance, and toxicity. We hypothesize that silver nanoparticles (AgNPs) will associate with proteins common to human serum and cell culture media forming a PC that will impact cell activation and cytotoxicity. Furthermore, the role of scavenger receptor BI (SR-BI) in mediating this toxicity was evaluated. Citrate-suspended 20 nm AgNPs were incubated with human serum albumin (HSA), bovine serum albumin (BSA), high-density lipoprotein (HDL), or water (control) to form a PC. AgNPs associated with each protein (HSA, BSA, and HDL) forming PCs as assessed by electron microscopy, hyperspectral analysis, ζ-potential, and hydrodynamic size. Addition of the PC decreased uptake of AgNPs by rat lung epithelial and rat aortic endothelial cells. Hyperspectral analysis demonstrated a loss of the AgNP PC following internalization. Cells demonstrated concentration-dependent cytotoxicity following exposure to AgNPs with or without PCs (0, 6.25, 12.5, 25 or 50 μg/ml). All PC-coated AgNPs were found to activate cells by inducing IL-6 mRNA expression. A small molecule SR-BI inhibitor was utilized to determine the role of SR-BI in the observed effects. Pretreatment with the SR-BI inhibitor decreased internalization of AgNPs with or without PCs, and reduced both cytotoxicity and IL-6 mRNA expression. This study characterizes the formation of a PC on AgNPs and demonstrates its influence on cytotoxicity and cell activation through a cell surface receptor.
Collapse
Affiliation(s)
- Jonathan H Shannahan
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625
| | - Ramakrishna Podila
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625 *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625
| | - Abdullah A Aldossari
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625
| | - Hilary Emerson
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625
| | - Brian A Powell
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625
| | - Pu Chun Ke
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625
| | - Apparao M Rao
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625 *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625
| | - Jared M Brown
- *Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, South Carolina 29625, and Department of Environmental Engineering and Earth Sciences, Clemson University, Anderson, South Carolina 29625
| |
Collapse
|
48
|
Tran-Dinh A, Diallo D, Delbosc S, Varela-Perez LM, Dang QB, Lapergue B, Burillo E, Michel JB, Levoye A, Martin-Ventura JL, Meilhac O. HDL and endothelial protection. Br J Pharmacol 2014; 169:493-511. [PMID: 23488589 DOI: 10.1111/bph.12174] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/07/2013] [Accepted: 02/24/2013] [Indexed: 12/23/2022] Open
Abstract
High-density lipoproteins (HDLs) represent a family of particles characterized by the presence of apolipoprotein A-I (apoA-I) and by their ability to transport cholesterol from peripheral tissues back to the liver. In addition to this function, HDLs display pleiotropic effects including antioxidant, anti-apoptotic, anti-inflammatory, anti-thrombotic or anti-proteolytic properties that account for their protective action on endothelial cells. Vasodilatation via production of nitric oxide is also a hallmark of HDL action on endothelial cells. Endothelial cells express receptors for apoA-I and HDLs that mediate intracellular signalling and potentially participate in the internalization of these particles. In this review, we will detail the different effects of HDLs on the endothelium in normal and pathological conditions with a particular focus on the potential use of HDL therapy to restore endothelial function and integrity.
Collapse
|
49
|
Oxidation-induced loss of the ability of HDL to counteract the inhibitory effect of oxidized LDL on vasorelaxation. Heart Vessels 2014; 30:845-9. [PMID: 25031153 DOI: 10.1007/s00380-014-0543-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 06/27/2014] [Indexed: 02/02/2023]
Abstract
Several current diseases are associated with an increase in the oxidation of HDL, which is likely to impair their functionality. Our aim was to identify whether oxidation could change the protective effect of HDL against the deleterious effect on vasoreactivity induced by oxidative stress. HDL from healthy subjects were oxidized in vitro by Cu(2+), and the ability of oxidized HDL to counteract the inhibitory effect of oxidized LDL on acetylcholine-induced vasodilation was tested on isolated rabbit aorta rings. Oxidation of HDL was evidenced by the increase in the 7-oxysterols/cholesterol ratio (3.20 ± 1.12 vs 0.02 ± 0.01 % in native HDL, p < 0.05). Oxidized LDL inhibited endothelium-dependent vasodilation (E max = 50.2 ± 5.0 vs 92.5 ± 1.7 % for incubation in Kreb's buffer, p < 0.05) and native HDL counteracted this inhibition (E max = 72.4 ± 4.8 vs 50.2 ± 5.0 % p < 0.05). At the opposite, oxidized HDL had no effect on oxidized LDL-induced inhibition on endothelium-dependent vasorelaxation (E max = 53.7 ± 4.8 vs 50.2 ± 5.0 %, NS). HDL oxidation is associated with a decreased ability of HDL to remove 7-oxysterols from oxidized LDL. In conclusion, these results show that oxidation of HDL induces the loss of their protective effect against endothelial dysfunction, which could promote atherosclerosis in diseases associated with increased oxidative stress.
Collapse
|
50
|
Ramadoss J, Pastore MB, Magness RR. Endothelial caveolar subcellular domain regulation of endothelial nitric oxide synthase. Clin Exp Pharmacol Physiol 2014; 40:753-64. [PMID: 23745825 DOI: 10.1111/1440-1681.12136] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 05/29/2013] [Accepted: 05/31/2013] [Indexed: 12/12/2022]
Abstract
Complex regulatory processes alter the activity of endothelial nitric oxide synthase (eNOS) leading to nitric oxide (NO) production by endothelial cells under various physiological states. These complex processes require specific subcellular eNOS partitioning between plasma membrane caveolar domains and non-caveolar compartments. Translocation of eNOS from the plasma membrane to intracellular compartments is important for eNOS activation and subsequent NO biosynthesis. We present data reviewing and interpreting information regarding: (i) the coupling of endothelial plasma membrane receptor systems in the caveolar structure relative to eNOS trafficking; (ii) how eNOS trafficking relates to specific protein-protein interactions for inactivation and activation of eNOS; and (iii) how these complex mechanisms confer specific subcellular location relative to eNOS multisite phosphorylation and signalling. Dysfunction in the regulation of eNOS activation may contribute to several disease states, in particular gestational endothelial abnormalities (pre-eclampsia, gestational diabetes etc.), that have life-long deleterious health consequences that predispose the offspring to develop hypertensive disease, Type 2 diabetes and adiposity.
Collapse
Affiliation(s)
- Jayanth Ramadoss
- Department of Obstetrics and Gynaecology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | |
Collapse
|