Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Blood Adv ; 8(9): 2104-2117, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38498701

ABSTRACT

ABSTRACT: Venous thromboembolic events are significant contributors to morbidity and mortality in patients with stroke. Neutrophils are among the first cells in the blood to respond to stroke and are known to promote deep vein thrombosis (DVT). Integrin α9 is a transmembrane glycoprotein highly expressed on neutrophils and stabilizes neutrophil adhesion to activated endothelium via vascular cell adhesion molecule 1 (VCAM-1). Nevertheless, the causative role of neutrophil integrin α9 in poststroke DVT remains unknown. Here, we found higher neutrophil integrin α9 and plasma VCAM-1 levels in humans and mice with stroke. Using mice with embolic stroke, we observed enhanced DVT severity in a novel model of poststroke DVT. Neutrophil-specific integrin α9-deficient mice (α9fl/flMrp8Cre+/-) exhibited a significant reduction in poststroke DVT severity along with decreased neutrophils and citrullinated histone H3 in thrombi. Unbiased transcriptomics indicated that α9/VCAM-1 interactions induced pathways related to neutrophil inflammation, exocytosis, NF-κB signaling, and chemotaxis. Mechanistic studies revealed that integrin α9/VCAM-1 interactions mediate neutrophil adhesion at the venous shear rate, promote neutrophil hyperactivation, increase phosphorylation of extracellular signal-regulated kinase, and induce endothelial cell apoptosis. Using pharmacogenomic profiling, virtual screening, and in vitro assays, we identified macitentan as a potent inhibitor of integrin α9/VCAM-1 interactions and neutrophil adhesion to activated endothelial cells. Macitentan reduced DVT severity in control mice with and without stroke, but not in α9fl/flMrp8Cre+/- mice, suggesting that macitentan improves DVT outcomes by inhibiting neutrophil integrin α9. Collectively, we uncovered a previously unrecognized and critical pathway involving the α9/VCAM-1 axis in neutrophil hyperactivation and DVT.


Subject(s)
Integrins , Neutrophils , Stroke , Vascular Cell Adhesion Molecule-1 , Venous Thrombosis , Animals , Humans , Male , Mice , Cell Adhesion , Disease Models, Animal , Integrins/metabolism , Mice, Knockout , Neutrophil Activation , Neutrophils/metabolism , Stroke/metabolism , Stroke/etiology , Vascular Cell Adhesion Molecule-1/metabolism , Venous Thrombosis/metabolism , Venous Thrombosis/etiology
2.
J Am Heart Assoc ; 13(5): e032987, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38390810

ABSTRACT

Cardiovascular disease remains the leading cause of death worldwide, characterized by atherosclerotic activity within large and medium-sized arteries. Inflammation has been shown to be a primary driver of atherosclerotic plaque formation, with interleukin-1 (IL-1) having a principal role. This review focuses on the current state of knowledge of molecular mechanisms of IL-1 release from cells in atherosclerotic plaques. A more in-depth understanding of the process of IL-1's release into the vascular environment is necessary for the treatment of inflammatory disease processes, as the current selection of medicines being used primarily target IL-1 after it has been released. IL-1 is secreted by several heterogenous mechanisms, some of which are cell type-specific and could provide further specialized targets for therapeutic intervention. A major unmet challenge is to understand the mechanism before and leading to IL-1 release, especially by cells in atherosclerotic plaques, including endothelial cells, vascular smooth muscle cells, and macrophages. Data so far indicate a heterogeneity of IL-1 release mechanisms that vary according to cell type and are stimulus-dependent. Unraveling this complexity may reveal new targets to block excess vascular inflammation.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Plaque, Atherosclerotic/metabolism , Interleukin-1 , Endothelial Cells/metabolism , Atherosclerosis/metabolism , Inflammation
4.
Front Cardiovasc Med ; 10: 1190460, 2023.
Article in English | MEDLINE | ID: mdl-37539090

ABSTRACT

Introduction: Atherosclerosis is a progressive disease that develops in areas of disturbed flow (d-flow). Progressive atherosclerosis is characterized by bulky plaques rich in mesenchymal cells and high-grade inflammation that can rupture leading to sudden cardiac death or acute myocardial infarction. In response to d-flow, endothelial cells acquire a mesenchymal phenotype through endothelial-to-mesenchymal transition (EndMT). However, the signaling intermediaries that link d-flow to EndMT are incompletely understood. Methods and Results: In this study we found that in human atherosclerosis, cells expressing SNAI1 (Snail 1, EndMT transcription factor) were highly expressed within the endothelial cell (EC) layer and in the pre-necrotic areas in unstable lesions, whereas stable lesions did not show any SNAI1 positive cells, suggesting a role for EndMT in lesion instability. The interleukin-1 (IL-1), which signals through the type-I IL-1 receptor (IL-1R1), has been implicated in plaque instability and linked to EndMT formation in vitro. Interestingly, we observed an association between SNAI1 and IL-1R1 within ECs in the unstable lesions. To establish the causal relationship between EndMT and IL-1R1 expression, we next examined IL-1R1 levels in our Cre-lox endothelial-specific lineage tracing mice. IL-1R1 and Snail1 were highly expressed in ECs under atheroprone compared to athero-protective areas, and oscillatory shear stress (OSS) increased IL-1R1 protein and mRNA levels in vitro. Exposure of ECs to OSS resulted in loss of their EC markers and higher induction of EndMT markers. By contrast, genetic silencing of IL-1R1 significantly reduced the expression of EndMT markers and Snail1 nuclear translocation, suggesting a direct role for IL-1R1 in d-flow-induced EndMT. In vivo, re-analysis of scRNA-seq datasets in carotid artery exposed to d-flow confirmed the IL-1R1 upregulation among EndMT population, and in our partial carotid ligation model of d-flow, endothelial cell specific IL-1R1 KO significantly reduced SNAI1 expression. Discussion: Global inhibition of IL-1 signaling in atherosclerosis as a therapeutic target has recently been tested in the completed CANTOS trial, with promising results. However, the data on IL-1R1 signaling in different vascular cell-types are inconsistent. Herein, we show endothelial IL-1R1 as a novel mechanosensitive receptor that couples d-flow to IL-1 signaling in EndMT.

5.
Arterioscler Thromb Vasc Biol ; 42(9): 1113-1120, 2022 09.
Article in English | MEDLINE | ID: mdl-35861068

ABSTRACT

Over the last 2 years, the COVID-19 pandemic has stimulated the scientific community by starting a race to develop new vaccines and therapeutic approaches to fight this life-threatening illness. At the same time, the pandemic also evoked an urge for innovative communication strategies to maintain scientific networking and data sharing among investigators. Communication through audiovisual platforms has quickly become a unique tool to sustain scientific interaction, whereas social media has turned into an unmistakable pivotal environment for sharing scientific data and combating misinformation around SARS-CoV-2 infection, prevention, and therapy. Amid this challenging scenario, the scientific community organically established new roles, such as a social media ambassador, a conference-associated role to virtually promote breakthrough science while reconnecting investigators and forging new scientific networks via social media. Moreover, in response to the COVID-19 pandemic, it also became clear the critical need for the scientific community to support efforts to empower flexibility, creativity, and the inclusion of new forms of communication to advance science. Thus, the goal of this brief article is to provide a structured follow-up on the importance for researchers to occupy the internet to promote scientific findings and events, to combat science mistrust by stimulating communication among nonscientists to scientists, and to provide essential strategies for young and senior investigators on how to virtually expand their professional networks within and across research and clinical areas of the cardiovascular field.


Subject(s)
COVID-19 , Social Media , Communication , Humans , Pandemics , SARS-CoV-2
6.
Matrix Biol ; 103-104: 1-21, 2021 09.
Article in English | MEDLINE | ID: mdl-34537369

ABSTRACT

The multifunctional glycoprotein fibronectin influences several crucial cellular processes and contributes to multiple pathologies. While a link exists between fibronectin-associated pathologies and the receptor tyrosine kinase EphA2, the mechanism by which EphA2 promotes fibronectin matrix remodeling remains unknown. We previously demonstrated that EphA2 deletion reduces smooth muscle fibronectin deposition and blunts fibronectin deposition in atherosclerosis without influencing fibronectin expression. We now show that EphA2 expression is required for contractility-dependent elongation of tensin- and α5ß1 integrin-rich fibrillar adhesions that drive fibronectin fibrillogenesis. Mechanistically, EphA2 localizes to integrin adhesions where focal adhesion kinase mediates ligand-independent Y772 phosphorylation, and mutation of this site significantly blunts fibrillar adhesion length. EphA2 deficiency decreases smooth muscle cell contractility by enhancing p190RhoGAP activation and reducing RhoA activity, whereas stimulating RhoA signaling in EphA2 deficient cells rescues fibrillar adhesion elongation. Together, these data identify EphA2 as a novel regulator of fibrillar adhesion elongation and provide the first data identifying a role for EphA2 signaling in integrin adhesions.


Subject(s)
Fibronectins , Integrins , Cell Adhesion , Cytoskeleton , Fibronectins/genetics , Focal Adhesions , Integrin alpha5beta1 , Integrins/genetics , Signal Transduction , Tensins/genetics
7.
J Cardiovasc Pharmacol ; 78(5): e656-e661, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34328710

ABSTRACT

ABSTRACT: Infarct size is a major determinant of outcomes after acute myocardial infarction (AMI). Carbon monoxide-releasing molecules (CORMs), which deliver nanomolar concentrations of carbon monoxide to tissues, have been shown to reduce infarct size in rodents. We evaluated efficacy and safety of CORM-A1 to reduce infarct size in a clinically relevant porcine model of AMI. We induced AMI in Yorkshire White pigs by inflating a coronary angioplasty balloon to completely occlude the left anterior descending artery for 60 minutes, followed by deflation of the balloon to mimic reperfusion. Fifteen minutes after balloon occlusion, animals were given an infusion of 4.27 mM CORM-A1 (n = 7) or sodium borate control (n = 6) over 60 minutes. Infarct size, cardiac biomarkers, ejection fraction, and hepatic and renal function were compared amongst the groups. Immunohistochemical analyses were performed to compare inflammation, cell proliferation, and apoptosis between the groups. CORM-A1-treated animals had significant reduction in absolute infarct area (158 ± 16 vs. 510 ± 91 mm2, P < 0.001) and infarct area corrected for area at risk (24.8% ± 2.6% vs. 45.2% ± 4.0%, P < 0.0001). Biochemical markers of myocardial injury also tended to be lower and left ventricular function tended to recover better in the CORM-A1 treated group. There was no evidence of hepatic or renal toxicity with the doses used. The cardioprotective effects of CORM-A1 were associated with a significant reduction in cell proliferation and inflammation. CORM-A1 reduces infarct size and improves left ventricular remodeling and function in a porcine model of reperfused MI by a reduction in inflammation. These potential cardioprotective effects of CORMs warrant further translational investigations.


Subject(s)
Boranes/pharmacology , Carbon Monoxide/metabolism , Carbonates/pharmacology , Cardiovascular Agents/pharmacology , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Boranes/metabolism , Carbonates/metabolism , Cardiovascular Agents/metabolism , Caspase 3/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Interleukin-1beta/metabolism , Ki-67 Antigen/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Sus scrofa , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
8.
Front Cell Dev Biol ; 9: 688388, 2021.
Article in English | MEDLINE | ID: mdl-34124074

ABSTRACT

The Nck family of modular adaptor proteins, including Nck1 and Nck2, link phosphotyrosine signaling to changes in cytoskeletal dynamics and gene expression that critically modulate cellular phenotype. The Nck SH2 domain interacts with phosphotyrosine at dynamic signaling hubs, such as activated growth factor receptors and sites of cell adhesion. The Nck SH3 domains interact with signaling effectors containing proline-rich regions that mediate their activation by upstream kinases. In vascular biology, Nck1 and Nck2 play redundant roles in vascular development and postnatal angiogenesis. However, recent studies suggest that Nck1 and Nck2 differentially regulate cell phenotype in the adult vasculature. Domain-specific interactions likely mediate these isoform-selective effects, and these isolated domains may serve as therapeutic targets to limit specific protein-protein interactions. In this review, we highlight the function of the Nck adaptor proteins, the known differences in domain-selective interactions, and discuss the role of individual Nck isoforms in vascular remodeling and function.

9.
J Am Heart Assoc ; 9(11): e016099, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32468886

ABSTRACT

Background Alteration in hemodynamic shear stress at atheroprone sites promotes endothelial paracellular pore formation and permeability. The molecular mechanism remains unknown. Methods and Results We show that Nck (noncatalytic region of tyrosine kinase) deletion significantly ameliorates disturbed flow-induced permeability, and selective isoform depletion suggests distinct signaling mechanisms. Only Nck1 deletion significantly reduces disturbed flow-induced paracellular pore formation and permeability, whereas Nck2 depletion has no significant effects. Additionally, Nck1 re-expression, but not Nck2, restores disturbed flow-induced permeability in Nck1/2 knockout cells, confirming the noncompensating roles. In vivo, using the partial carotid ligation model of disturbed flow, Nck1 knockout prevented the increase in vascular permeability, as assessed by Evans blue and fluorescein isothiocyanate dextran extravasations and leakage of plasma fibrinogen into the vessel wall. Domain swap experiments mixing SH2 (phosphotyrosine binding) and SH3 (proline-rich binding) domains between Nck1 and Nck2 showed a dispensable role for SH2 domains but a critical role for the Nck1 SH3 domains in rescuing disturbed flow-induced endothelial permeability. Consistent with this, both Nck1 and Nck2 bind to platelet endothelial adhesion molecule-1 (SH2 dependent) in response to shear stress, but only Nck1 ablation interferes with shear stress-induced PAK2 (p21-activated kinase) membrane translocation and activation. A single point mutation into individual Nck1 SH3 domains suggests a role for the first domain of Nck1 in PAK recruitment to platelet endothelial cell adhesion molecule-1 and activation in response to shear stress. Conclusions This work provides the first evidence that Nck1 but not the highly similar Nck2 plays a distinct role in disturbed flow-induced vascular permeability by selective p21-activated kinase activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carotid Artery Diseases/enzymology , Endothelial Cells/enzymology , Mechanotransduction, Cellular , Oncogene Proteins/metabolism , p21-Activated Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Carotid Artery Diseases/pathology , Carotid Artery Diseases/physiopathology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Oncogene Proteins/genetics , Permeability , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Regional Blood Flow , Stress, Mechanical , p21-Activated Kinases/genetics , src Homology Domains
10.
J Clin Invest ; 130(8): 4331-4347, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32427580

ABSTRACT

Although the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS) established the role of treating inflammation in atherosclerosis, our understanding of endothelial activation at atherosclerosis-prone sites remains limited. Disturbed flow at atheroprone regions primes plaque inflammation by enhancing endothelial NF-κB signaling. Herein, we demonstrate a role for the Nck adaptor proteins in disturbed flow-induced endothelial activation. Although highly similar, only Nck1 deletion, but not Nck2 deletion, limited flow-induced NF-κB activation and proinflammatory gene expression. Nck1-knockout mice showed reduced endothelial activation and inflammation in both models, disturbed flow- and high fat diet-induced atherosclerosis, whereas Nck2 deletion did not. Bone marrow chimeras confirmed that vascular Nck1, but not hematopoietic Nck1, mediated this effect. Domain-swap experiments and point mutations identified the Nck1 SH2 domain and the first SH3 domain as critical for flow-induced endothelial activation. We further characterized Nck1's proinflammatory role by identifying interleukin 1 type I receptor kinase-1 (IRAK-1) as a Nck1-selective binding partner, demonstrating that IRAK-1 activation by disturbed flow required Nck1 in vitro and in vivo, showing endothelial Nck1 and IRAK-1 staining in early human atherosclerosis, and demonstrating that disturbed flow-induced endothelial activation required IRAK-1. Taken together, our data reveal a hitherto unknown link between Nck1 and IRAK-1 in atherogenic inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/metabolism , Oncogene Proteins/metabolism , Plaque, Atherosclerotic/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Line , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, Knockout , Oncogene Proteins/genetics , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Point Mutation , src Homology Domains
11.
Redox Biol ; 34: 101487, 2020 07.
Article in English | MEDLINE | ID: mdl-32173345

ABSTRACT

Endothelial nitric oxide (NO) is a critical mediator of vascular function and vascular remodeling. NO is produced by endothelial nitric oxide synthase (eNOS), which is activated by calcium (Ca2+)-dependent and Ca2+-independent pathways. Here, we report that neurogranin (Ng), which regulates Ca2+-calmodulin (CaM) signaling in the brain, is uniquely expressed in endothelial cells (EC) of human and mouse vasculature, and is also required for eNOS regulation. To test the role of Ng in eNOS activation, Ng knockdown in human aortic endothelial cells (HAEC) was performed using Ng SiRNA along with Ng knockout (Ng -/-) in mice. Depletion of Ng expression decreased eNOS activity in HAEC and NO production in mice. We show that Ng expression was decreased by short-term laminar flow and long-them oscillating flow shear stress, and that Ng siRNA with shear stress decreased eNOS expression as well as eNOS phosphorylation at S1177. We further reveled that lack of Ng expression decreases both AKT-dependent eNOS phosphorylation, NF-κB-mediated eNOS expression, and promotes endothelial activation. Our findings also indicate that Ng modulates Ca2+-dependent calcineurin (CaN) activity, which suppresses Ca2+-independent AKT-dependent eNOS signaling. Moreover, deletion of Ng in mice also reduced eNOS activity and caused endothelial dysfunction in flow-mediated dilation experiments. Our results demonstrate that Ng plays a crucial role in Ca2+-CaM-dependent eNOS regulation and contributes to vascular remodeling, which is important for the pathophysiology of cardiovascular disease.


Subject(s)
Neurogranin , Nitric Oxide Synthase Type III , Animals , Cells, Cultured , Endothelial Cells/metabolism , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation
12.
Arterioscler Thromb Vasc Biol ; 38(11): 2601-2614, 2018 11.
Article in English | MEDLINE | ID: mdl-30354234

ABSTRACT

Objective- Alterations in extracellular matrix quantity and composition contribute to atherosclerosis, with remodeling of the subendothelial basement membrane to an FN (fibronectin)-rich matrix preceding lesion development. Endothelial cell interactions with FN prime inflammatory responses to a variety of atherogenic stimuli; however, the mechanisms regulating early atherogenic FN accumulation remain unknown. We previously demonstrated that oxLDL (oxidized low-density lipoprotein) promotes endothelial proinflammatory gene expression by activating the integrin α5ß1, a classic mediator of FN fibrillogenesis. Approach and Results- We now show that oxLDL drives robust endothelial FN deposition and inhibiting α5ß1 (blocking antibodies, α5 knockout cells) completely inhibits oxLDL-induced FN deposition. Consistent with this, inducible endothelial-specific α5 integrin deletion in ApoE knockout mice significantly reduces atherosclerotic plaque formation, associated with reduced early atherogenic inflammation. Unlike TGFß (transforming growth factor ß)-induced FN deposition, oxLDL does not induce FN expression (mRNA, protein) or the endothelial-to-mesenchymal transition phenotype. In addition, we show that cell-derived and plasma-derived FN differentially affect endothelial function, with only cell-derived FN capable of supporting oxLDL-induced VCAM-1 (vascular cell adhesion molecule 1) expression, despite plasma FN deposition by oxLDL. The inclusion of alternative exon EIIIA (EDA) of FN (EIIIA) and alternative exon EIIIB (EDB) of FN (EIIIB) domains in cell-derived FN mediates this effect, as EIIIA/EIIIB knockout endothelial cells show diminished oxLDL-induced inflammation. Furthermore, our data suggest that EIIIA/EIIIB-positive cellular FN is required for maximal α5ß1 recruitment to focal adhesions and FN fibrillogenesis. Conclusions- Taken together, our data demonstrate that endothelial α5 integrins drive oxLDL-induced FN deposition and early atherogenic inflammation. Additionally, we show that α5ß1-dependent endothelial FN deposition mediates oxLDL-dependent endothelial inflammation and FN fibrillogenesis.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Carotid Artery Diseases/metabolism , Endothelial Cells/metabolism , Fibronectins/metabolism , Inflammation/metabolism , Integrin alpha5beta1/metabolism , Plaque, Atherosclerotic , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cadherins/genetics , Cadherins/metabolism , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Fibronectins/deficiency , Fibronectins/genetics , Focal Adhesions/metabolism , Focal Adhesions/pathology , Humans , Inflammation/genetics , Inflammation/pathology , Integrin alpha5beta1/deficiency , Integrin alpha5beta1/genetics , Lipoproteins, LDL/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Signal Transduction
13.
J Am Heart Assoc ; 7(13)2018 06 30.
Article in English | MEDLINE | ID: mdl-29960988

ABSTRACT

BACKGROUND: Hypertension is a complex condition and a common cardiovascular risk factor. Dietary docosahexaenoic acid (DHA) modulates atherosclerosis and hypertension, possibly via an inflammatory mechanism. IL-1 (interleukin 1) has an established role in atherosclerosis and inflammation, although whether IL-1 inhibition modulates blood pressure is unclear. METHODS AND RESULTS: Male apoE-/- (apolipoprotein E-null) mice were fed either a high fat diet or a high fat diet plus DHA (300 mg/kg per day) for 12 weeks. Blood pressure and cardiac function were assessed, and effects of DHA on wall shear stress and atherosclerosis were determined. DHA supplementation improved left ventricular function, reduced wall shear stress and oscillatory shear at ostia in the descending aorta, and significantly lowered blood pressure compared with controls (119.5±7 versus 159.7±3 mm Hg, P<0.001, n=4 per group). Analysis of atheroma following DHA feeding in mice demonstrated a 4-fold reduction in lesion burden in distal aortas and in brachiocephalic arteries (P<0.001, n=12 per group). In addition, DHA treatment selectively decreased plaque endothelial IL-1ß (P<0.01). CONCLUSIONS: Our findings revealed that raised blood pressure can be reduced by inhibiting IL-1 indirectly by administration of DHA in the diet through a mechanism that involves a reduction in wall shear stress and local expression of the proinflammatory cytokine IL-1ß.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Aorta/drug effects , Aortic Diseases/prevention & control , Arterial Pressure/drug effects , Atherosclerosis/prevention & control , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Hypertension/prevention & control , Interleukin-1beta/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Diet, High-Fat , Disease Models, Animal , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Signal Transduction/drug effects , Stress, Mechanical
14.
Arterioscler Thromb Vasc Biol ; 38(9): 2126-2136, 2018 09.
Article in English | MEDLINE | ID: mdl-30002061

ABSTRACT

Objective- Flow patterns differentially regulate endothelial cell phenotype, with laminar flow promoting vasodilation and disturbed flow promoting endothelial proinflammatory activation. CSE (cystathionine γ-lyase), a major source of hydrogen sulfide (H2S) in endothelial cells, critically regulates cardiovascular function, by both promoting vasodilation and reducing endothelial activation. Therefore, we sought to investigate the role of CSE in the endothelial response to flow. Approach and Results- Wild-type C57Bl/6J and CSE knockout ( CSE-/-) mice underwent partial carotid ligation to induce disturbed flow in the left carotid. In addition, endothelial cells isolated from wild-type and CSE -/- mice were exposed to either laminar or oscillatory flow, an in vitro model of disturbed flow. Interestingly, laminar flow significantly reduced CSE expression in vitro, and only disturbed flow regions show discernable CSE protein expression in vivo, correlating with enhanced H2S production in wild-type C57BL/6J but not CSE-/- mice. Lack of CSE limited disturbed flow-induced proinflammatory gene expression (ICAM-1[intercellular adhesion molecule 1], VCAM-1 [vascular cell adhesion molecular 1]) and monocyte infiltration and CSE-/- endothelial cells showed reduced NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation and proinflammatory gene expression in response to oscillatory flow in vitro. In addition, CSE-/- mice showed reduced inward remodeling after partial carotid ligation. CSE-/- mice showed elevated vascular nitrite levels (measure of nitric oxide [NO]) in the unligated carotids, suggesting an elevation in baseline NO production, and the NO scavenger 2-(4-carboxyphenyl)-4,5-dihydro-4,4,5,5-tetramethyl-1H-imidazolyl-1-oxy-3-oxide normalized the reduced inward remodeling, but not inflammation, of ligated carotids in CSE-/- mice. Conclusions- CSE expression in disturbed flow regions critically regulates both endothelial activation and flow-dependent vascular remodeling, in part through altered NO availability.


Subject(s)
Blood Flow Velocity , Cystathionine gamma-Lyase/metabolism , Endothelial Cells/physiology , Vascular Remodeling/physiology , Animals , Benzoates/metabolism , Biological Availability , Carotid Arteries/physiology , Cells, Cultured , Cystathionine gamma-Lyase/genetics , Gene Expression , Humans , Hydrogen Sulfide/metabolism , Imidazoles/metabolism , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Signal Transduction , Stress, Mechanical , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vasodilation
15.
Pulm Circ ; 8(1): 2045893217752328, 2018.
Article in English | MEDLINE | ID: mdl-29261014

ABSTRACT

Idiopathic pulmonary arterial hypertension (IPAH) is increasingly diagnosed in elderly patients who also have an increased risk of co-morbid atherosclerosis. Apolipoprotein E-deficient (ApoE-/-) mice develop atherosclerosis with severe PAH when fed a high-fat diet (HFD) and have increased levels of endothelin (ET)-1. ET-1 receptor antagonists (ERAs) are used for the treatment of PAH but less is known about whether ERAs are beneficial in atherosclerosis. We therefore examined whether treatment of HFD-ApoE-/- mice with macitentan, a dual ETA/ETB receptor antagonist, would have any effect on both atherosclerosis and PAH. ApoE-/- mice were fed chow or HFD for eight weeks. After four weeks of HFD, mice were randomized to a four-week treatment of macitentan by food (30 mg/kg/day dual ETA/ETB antagonist), or placebo groups. Echocardiography and closed-chest right heart catheterization were used to determine PAH phenotype and serum samples were collected for cytokine analysis. Thoracic aortas were harvested to assess vascular reactivity using wire myography, and histological analyses were performed on the brachiocephalic artery and aortic root to assess atherosclerotic burden. Macitentan treatment of HFD-fed ApoE-/- mice was associated with a beneficial effect on the PAH phenotype and led to an increase in endothelial-dependent relaxation in thoracic aortae. Macitentan treatment was also associated with a significant reduction in interleukin 6 (IL-6) concentration but there was no significant effect on atherosclerotic burden. Dual blockade of ETA/ETB receptors improves endothelial function and improves experimental PAH but had no significant effect on atherosclerosis.

16.
J Biol Chem ; 290(40): 24067-78, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26269588

ABSTRACT

The endothelium is critically involved in the pathogenesis of atherosclerosis by producing pro-inflammatory mediators, including IL-1ß. Coronary arteries from patients with ischemic heart disease express large amounts of IL-1ß in the endothelium. However, the mechanism by which endothelial cells (ECs) release IL-1ß remains to be elucidated. We investigated neutrophil elastase (NE), a potent serine protease detected in vulnerable areas of human carotid plaques, as a potential "trigger" for IL-1ß processing and release. This study tested the hypothesis that NE potentiates the processing and release of IL-1ß from human coronary endothelium. We found that NE cleaves the pro-isoform of IL-1ß in ECs and causes significant secretion of bioactive IL-1ß via extracellular vesicles. This release was attenuated significantly by inhibition of neutrophil elastase but not caspase-1. Transient increases in intracellular Ca(2+) levels were observed prior to secretion. Inside ECs, and after NE treatment only, IL-1ß was detected within LAMP-1-positive multivesicular bodies. The released vesicles contained bioactive IL-1ß. In vivo, in experimental atherosclerosis, NE was detected in mature atherosclerotic plaques, predominantly in the endothelium, alongside IL-1ß. This study reveals a novel mechanistic link between NE expression in atherosclerotic plaques and concomitant pro-inflammatory bioactive IL-1ß secretion from ECs. This could reveal additional potential anti-IL-1ß therapeutic targets and provide further insights into the inflammatory process by which vascular disease develops.


Subject(s)
Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation, Enzymologic , Interleukin-1beta/metabolism , Leukocyte Elastase/metabolism , Animals , Apolipoproteins E/genetics , Apoptosis , Calcium/metabolism , Cell Survival , Cells, Cultured , Endothelial Cells/enzymology , Humans , Immunohistochemistry , Inflammation/metabolism , Male , Mice , Mice, Transgenic , Microcirculation , Phosphorylation , Plaque, Atherosclerotic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...