Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters











Publication year range
1.
Sci Rep ; 11(1): 5894, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33723357

ABSTRACT

The aetiology and progression of hypertension involves various endogenous systems, such as the renin angiotensin system, the sympathetic nervous system, and endothelial dysfunction. Recent data suggest that vascular inflammation may also play a key role in the pathogenesis of hypertension. This study sought to determine whether high intraluminal pressure results in vascular inflammation. Leukocyte adhesion was assessed in rat carotid arteries exposed to 1 h of high intraluminal pressure. The effect of intraluminal pressure on signaling mechanisms including reactive oxygen species production (ROS), arginase expression, and NFĸB translocation was monitored. 1 h exposure to high intraluminal pressure (120 mmHg) resulted in increased leukocyte adhesion and inflammatory gene expression in rat carotid arteries. High intraluminal pressure also resulted in a downstream signaling cascade of ROS production, arginase expression, and NFĸB translocation. This process was found to be angiotensin II-independent and mediated by the mechanosensor caveolae, as caveolin-1 (Cav1)-deficient endothelial cells and mice were protected from pressure-induced vascular inflammatory signaling and leukocyte adhesion. Cav1 deficiency also resulted in a reduction in pressure-induced glomerular macrophage infiltration in vivo. These findings demonstrate Cav1 is an important mechanosensor in pressure-induced vascular and renal inflammation.


Subject(s)
Blood Vessels/metabolism , Blood Vessels/pathology , Caveolin 1/metabolism , Inflammation/metabolism , Inflammation/pathology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure , Caveolae/metabolism , Cell Adhesion , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/ultrastructure , Hypertension/pathology , Kidney/pathology , Leukocytes/pathology , Macrophages/pathology , Mice, Inbred C57BL , Models, Biological , NF-kappa B/metabolism , Norepinephrine , Rats , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/metabolism
2.
Int J Mol Sci ; 20(17)2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31480335

ABSTRACT

Cyclo-oxygenase (COX) inhibitors are among the most commonly used drugs in the western world for their anti-inflammatory and analgesic effects. However, they are also well-known to increase the risk of coronary events. This area is of renewed significance given alarming new evidence suggesting this effect can occur even with acute usage. This contrasts with the well-established usage of aspirin as a mainstay for cardiovascular prophylaxis, as well as overwhelming evidence that COX inhibition induces vasodilation and is protective for vascular function. Here, we present an updated review of the preclinical and clinical literature regarding the cardiotoxicity of COX inhibitors. While studies to date have focussed on the role of COX in influencing renal and vascular function, we suggest an interaction between prostanoids and T cells may be a novel factor, mediating elevated cardiovascular disease risk with NSAID use.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cardiovascular Diseases/chemically induced , Cyclooxygenase Inhibitors/adverse effects , Animals , Cardiovascular Diseases/immunology , Cardiovascular Diseases/physiopathology , Humans , Prostaglandins/metabolism , Risk Factors , T-Lymphocytes/drug effects
3.
Int J Mol Sci ; 20(12)2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31200567

ABSTRACT

It is now becomingly increasingly evident that the functions of the mammalian Y chromosome are not circumscribed to the induction of male sex. While animal studies have shown variations in the Y are strongly accountable for blood pressure (BP), this is yet to be confirmed in humans. We have recently shown modulation of adaptive immunity to be a significant mechanism underpinning Y-chromosome-dependent differences in BP in consomic strains. This is paralleled by studies in man showing Y chromosome haplogroup is a significant predictor for coronary artery disease through influencing pathways of immunity. Furthermore, recent studies in mice and humans have shown that Y chromosome lineage determines susceptibility to autoimmune disease. Here we review the evidence in animals and humans that Y chromosome lineage influences hypertension and cardiovascular disease risk, with a novel focus on pathways of immunity as a significant pathway involved.


Subject(s)
Blood Pressure/genetics , Cardiovascular Diseases/genetics , Immunity, Innate/genetics , Y Chromosome/genetics , Animals , Cardiovascular Diseases/immunology , Humans
4.
Front Immunol ; 10: 380, 2019.
Article in English | MEDLINE | ID: mdl-30899260

ABSTRACT

Elevated serum amyloid A (SAA) levels may promote endothelial dysfunction, which is linked to cardiovascular and renal pathologies. We investigated the effect of SAA on vascular and renal function in apolipoprotein E-deficient (ApoE-/-) mice. Male ApoE-/- mice received vehicle (control), low-level lipopolysaccharide (LPS), or recombinant human SAA by i.p. injection every third day for 2 weeks. Heart, aorta and kidney were harvested between 3 days and 18 weeks after treatment. SAA administration increased vascular cell adhesion molecule (VCAM)-1 expression and circulating monocyte chemotactic protein (MCP)-1 and decreased aortic cyclic guanosine monophosphate (cGMP), consistent with SAA inhibiting nitric oxide bioactivity. In addition, binding of labeled leukocytes to excised aorta increased as monitored using an ex vivo leukocyte adhesion assay. Renal injury was evident 4 weeks after commencement of SAA treatment, manifesting as increased plasma urea, urinary protein, oxidized lipids, urinary kidney injury molecule (KIM)-1 and multiple cytokines and chemokines in kidney tissue, relative to controls. Phosphorylation of nuclear-factor-kappa-beta (NFκB-p-P65), tissue factor (TF), and macrophage recruitment increased in kidneys from ApoE-/- mice 4 weeks after SAA treatment, confirming that SAA elicited a pro-inflammatory and pro-thrombotic phenotype. These data indicate that SAA impairs endothelial and renal function in ApoE-/- mice in the absence of a high-fat diet.


Subject(s)
Blood Vessels/metabolism , Kidney Diseases/etiology , Kidney Diseases/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Apolipoproteins E/deficiency , Biomarkers , Blood Vessels/pathology , Blood Vessels/physiopathology , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Endothelial Cells/metabolism , Immunohistochemistry , Inflammation Mediators/metabolism , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney Function Tests , Lipids/blood , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Peroxidase/metabolism , Serum Amyloid A Protein/metabolism
5.
FASEB J ; 33(1): 1510-1521, 2019 01.
Article in English | MEDLINE | ID: mdl-30156911

ABSTRACT

Despite its well-known antithrombotic properties, the effect of aspirin on blood pressure (BP) and hypertension pathology is unclear. The hugely varying doses used clinically have contributed to this confusion, with high-dose aspirin still commonly used due to concerns about the efficacy of low-dose aspirin. Because prostaglandins have been shown to both promote and inhibit T-cell activation, we also explored the immunomodulatory properties of aspirin in hypertension. Although the common preclinical high dose of 100 mg/kg/d improved vascular dysfunction and cardiac hypertrophy, this effect was accompanied by indices of elevated adaptive immunity, renal T-cell infiltration, renal fibrosis, and BP elevation in stroke-prone spontaneously hypertensive rats and in angiotensin II-induced hypertensive mice. The cardioprotective effects of aspirin were conserved with a lower dose (10 mg/kg/d) while circumventing heightened adaptive immunity and elevated BP. We also show that low-dose aspirin improves renal fibrosis. Differential inhibition of the COX-2 isoform may underlie the disparate effects of the 2 doses. Our results demonstrate the efficacy of low-dose aspirin in treating a vast array of cardiovascular parameters and suggest modulation of adaptive immunity as a novel mechanism underlying adverse cardiovascular profiles associated with COX-2 inhibitors. Clinical studies should identify the dose of aspirin that achieves maximal cardioprotection with a new awareness that higher doses of aspirin could trigger undesired autoimmunity in hypertensive individuals. This work also warrants an evaluation of high-dose aspirin and COX-2 inhibitor therapy in sufferers of inflammatory conditions who are already at increased risk for cardiovascular disease.-Khan, S. I., Shihata, W. A., Andrews, K. L., Lee, M. K. S., Moore, X.-L., Jefferis, A.-M., Vinh, A., Gaspari, T., Dragoljevic, D., Jennings, G. L., Murphy, A. J., Chin-Dusting, J. P. F. Effects of high- and low-dose aspirin on adaptive immunity and hypertension in the stroke-prone spontaneously hypertensive rat.


Subject(s)
Adaptive Immunity/drug effects , Aspirin/pharmacology , Hypertension/drug therapy , Stroke/complications , Stroke/immunology , Angiotensin II/pharmacology , Animals , Aspirin/administration & dosage , Aspirin/therapeutic use , Biomarkers/blood , Blood Pressure/drug effects , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/physiopathology , Cardiomegaly/drug therapy , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cytokines/blood , Disease Susceptibility , Dose-Response Relationship, Drug , Epoprostenol/biosynthesis , Hypertension/chemically induced , Kidney/drug effects , Kidney/enzymology , Kidney/pathology , Mice , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Real-Time Polymerase Chain Reaction , Systole , T-Lymphocytes/immunology , Thromboxanes/blood
6.
Haematologica ; 104(3): 456-467, 2019 03.
Article in English | MEDLINE | ID: mdl-30361420

ABSTRACT

Hypertension is a major, independent risk factor for atherosclerotic cardiovascular disease. However, this pathology can arise through multiple pathways, which could influence vascular disease through distinct mechanisms. An overactive sympathetic nervous system is a dominant pathway that can precipitate in elevated blood pressure. We aimed to determine how the sympathetic nervous system directly promotes atherosclerosis in the setting of hypertension. We used a mouse model of sympathetic nervous system-driven hypertension on the atherosclerotic-prone apolipoprotein E-deficient background. When mice were placed on a western type diet for 16 weeks, we showed the evolution of unstable atherosclerotic lesions. Fortuitously, the changes in lesion composition were independent of endothelial dysfunction, allowing for the discovery of alternative mechanisms. With the use of flow cytometry and bone marrow imaging, we found that sympathetic activation caused deterioration of the hematopoietic stem and progenitor cell niche in the bone marrow, promoting the liberation of these cells into the circulation and extramedullary hematopoiesis in the spleen. Specifically, sympathetic activation reduced the abundance of key hematopoietic stem and progenitor cell niche cells, sinusoidal endothelial cells and osteoblasts. Additionally, sympathetic bone marrow activity prompted neutrophils to secrete proteases to cleave the hematopoietic stem and progenitor cell surface receptor CXCR4. All these effects could be reversed using the ß-blocker propranolol during the feeding period. These findings suggest that elevated blood pressure driven by the sympathetic nervous system can influence mechanisms that modulate the hematopoietic system to promote atherosclerosis and contribute to cardiovascular events.


Subject(s)
Atherosclerosis/blood , Atherosclerosis/etiology , Hematopoiesis , Hypertension/complications , Hypertension/etiology , Sympathetic Nervous System/physiopathology , Animals , Atherosclerosis/pathology , Autonomic Nerve Block , Biomarkers , Biopsy , Bone Marrow/metabolism , Bone Marrow/pathology , Disease Models, Animal , Disease Susceptibility , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Myelopoiesis , Phenotype , Signal Transduction/drug effects , Stem Cell Niche
7.
Eur Heart J ; 39(23): 2158-2167, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29905812

ABSTRACT

Aim: Rheumatoid arthritis (RA) is associated with an approximately two-fold elevated risk of cardiovascular (CV)-related mortality. Patients with RA present with systemic inflammation including raised circulating myeloid cells, but fail to display traditional CV risk-factors, particularly dyslipidaemia. We aimed to explore if increased circulating myeloid cells is associated with impaired atherosclerotic lesion regression or altered progression in RA. Methods and results: Using flow cytometry, we noted prominent monocytosis, neutrophilia, and thrombocytosis in two mouse models of RA. This was due to enhanced proliferation of the haematopoietic stem and progenitor cells (HSPCs) in the bone marrow and the spleen. HSPCs expansion was associated with an increase in the cholesterol content, due to a down-regulation of cholesterol efflux genes, Apoe, Abca1, and Abcg1. The HSPCs also had enhanced expression of key myeloid promoting growth factor receptors. Systemic inflammation was found to cause defective cellular cholesterol metabolism. Increased myeloid cells in mice with RA were associated with a significant impairment in lesion regression, even though cholesterol levels were equivalent to non-arthritic mice. Lesions from arthritic mice exhibited a less stable phenotype as demonstrated by increased immune cell infiltration, lipid accumulation, and decreased collagen formation. In a progression model, we noted monocytosis, enhanced monocytes recruitment to lesions, and increased plaque macrophages. This was reversed with administration of reconstituted high-density lipoprotein (rHDL). Furthermore, RA patients have expanded CD16+ monocyte subsets and a down-regulation of ABCA1 and ABCG1. Conclusion: Rheumatoid arthritis impairs atherosclerotic regression and alters progression, which is associated with an expansion of myeloid cells and disturbed cellular cholesterol handling, independent of plasma cholesterol levels. Infusion of rHDL prevented enhanced myelopoiesis and monocyte entry into lesions. Targeting cellular cholesterol defects in people with RA, even if plasma cholesterol is within the normal range, may limit vascular disease.


Subject(s)
Arthritis, Rheumatoid/metabolism , Atherosclerosis/metabolism , Cholesterol/metabolism , Hematopoietic Stem Cells/metabolism , Monocytes/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Adult , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Arthritis, Rheumatoid/immunology , Atherosclerosis/genetics , Atherosclerosis/immunology , Disease Models, Animal , Down-Regulation , Female , Hematopoiesis, Extramedullary/immunology , Humans , Leukocytosis , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Mice , Middle Aged , Monocytes/immunology , Myelopoiesis/immunology , Neutrophils , RNA, Messenger/metabolism , Thrombocytosis
8.
FASEB J ; 32(5): 2747-2756, 2018 05.
Article in English | MEDLINE | ID: mdl-29301944

ABSTRACT

The essential role of the Y chromosome in male sex determination has largely overshadowed the possibility that it may exert other biologic roles. Here, we show that Y-chromosome lineage is a strong determinant of perivascular and renal T-cell infiltration in the stroke-prone spontaneously hypertensive rat, which, in turn, may influence vascular function and blood pressure (BP). We also show, for the first time to our knowledge, that augmented perivascular T-cell levels can directly instigate vascular dysfunction, and that the production of reactive oxygen species that stimulate cyclo-oxygenase underlies this. We thus provide strong evidence for the consideration of Y-chromosome lineage in the diagnosis and treatment of male hypertension, and point to the modulation of cardiovascular organ T-cell infiltration as a possible mechanism that underpins Y- chromosome regulation of BP.-Khan, S. I., Andrews, K. L., Jackson, K. L., Memon, B., Jefferis, A.-M., Lee, M. K. S., Diep, H., Wei, Z., Drummond, G. R., Head, G. A., Jennings, G. L., Murphy, A. J., Vinh, A., Sampson, A. K., Chin-Dusting, J. P. F. Y-chromosome lineage determines cardiovascular organ T-cell infiltration in the stroke-prone spontaneously hypertensive rat.


Subject(s)
Blood Pressure , Hypertension/metabolism , Hypertension/physiopathology , T-Lymphocytes/metabolism , Y Chromosome/metabolism , Animals , Hypertension/genetics , Male , Rats , Rats, Inbred SHR , Rats, Transgenic , T-Lymphocytes/pathology , Y Chromosome/genetics
9.
Haematologica ; 103(4): 597-606, 2018 04.
Article in English | MEDLINE | ID: mdl-29371326

ABSTRACT

Obesity enhances the risk of developing myelodysplastic syndromes. However, the effect of obesity on survival is unclear. Obese people present with monocytosis due to inflammatory signals emanating from obese adipose tissue. We hypothesized that obesity-induced myelopoiesis would promote the transition of myelodysplastic syndrome to acute myeloid leukemia and accelerate mortality in obesity. Obese Ob/Ob mice or their lean littermate controls received a bone marrow transplant from NUP98-HOXD13 transgenic mice, a model of myelodysplastic syndrome. The metabolic parameters of the mice were examined throughout the course of the study, as were blood leukocytes. Myeloid cells were analyzed in the bone, spleen, liver and adipose tissue by flow cytometry halfway through the disease progression and at the endpoint. Survival curves were also calculated. Contrary to our hypothesis, transplantation of NUP98-HOXD13 bone marrow into obese recipient mice significantly increased survival time compared with lean recipient controls. While monocyte skewing was exacerbated in obese mice receiving NUP98-HOXD13 bone marrow, transformation to acute myeloid leukemia was not enhanced. Increased survival of obese mice was associated with a preservation of fat mass as well as increased myeloid cell deposition within the adipose tissue, and a concomitant reduction in detrimental myeloid cell accumulation within other organs. The study herein revealed that obesity increases survival in animals with myelodysplastic syndrome. This may be due to the greater fat mass of Ob/Ob mice, which acts as a sink for myeloid cells, preventing their accumulation in other key organs, such as the liver.


Subject(s)
Myelodysplastic Syndromes/mortality , Obesity , Animals , Bone Marrow/chemistry , Bone Marrow Transplantation , Disease Models, Animal , Homeodomain Proteins , Leptin/deficiency , Leukemia, Myeloid, Acute/etiology , Mice , Mice, Transgenic , Myelodysplastic Syndromes/pathology , Myeloid Cells , Nuclear Pore Complex Proteins , Survival Rate , Transcription Factors
10.
Clin Sci (Lond) ; 132(1): 131-143, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29162746

ABSTRACT

Vascular dysfunction is a hallmark of hypertension and the strongest risk factor to date for coronary artery disease. As Y chromosome lineage has emerged as one of the strongest genetic predictors of cardiovascular disease risk to date, we investigated if Y chromosome lineage modulated this important facet in the stroke-prone spontaneously hypertensive rat (SHRSP) using consomic strains. Here, we show that vascular dysfunction in the SHRSP is attributable to differential cyclooxygenase (COX) activity with nitric oxide (NO) levels playing a less significant role. Measurement of prostacyclin, the most abundant product of COX in the vasculature, confirmed the augmented COX activity in the SHRSP aorta. This was accompanied by functional impairment of the vasodilatory prostacyclin (IP) receptor, while inhibition of the thromboxane (TP) receptor significantly ameliorated vascular dysfunction in the SHRSP, suggesting this is the downstream target responsible for constrictor prostanoid activity. Importantly, Y chromosome lineage was shown to modulate vascular function in the SHRSP through influencing COX activity, prostacyclin levels and IP dysfunction. Vascular dysfunction in the renal and intrarenal arteries was also found to be prostanoid and Y chromosome dependent. Interestingly, despite no apparent differences in agonist-stimulated NO levels, basal NO levels were compromised in the SHRSP aorta, which was also Y chromosome dependent. Thus, in contrast with the widely held view that COX inhibition is deleterious for the vasculature due to inhibition of the vasodilator prostacyclin, we show that COX inhibition abolishes vascular dysfunction in three distinct vascular beds, with IP dysfunction likely being a key mechanism underlying this effect. We also delineate a novel role for Y chromosome lineage in regulating vascular function through modulation of COX and basal NO levels.


Subject(s)
Aorta/physiopathology , Hypertension/physiopathology , Prostaglandins/metabolism , Stroke/physiopathology , Y Chromosome , Acetylcholine/pharmacology , Animals , Aorta/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Hypertension/genetics , Male , Nitric Oxide/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Stroke/genetics , Vasodilation/drug effects , Vasodilator Agents/pharmacology
11.
Int J Mol Sci ; 20(1)2018 Dec 28.
Article in English | MEDLINE | ID: mdl-30597899

ABSTRACT

The acute phase protein serum amyloid A (SAA) is associated with endothelial dysfunction and early-stage atherogenesis. Stimulation of vascular cells with SAA increases gene expression of pro-inflammation cytokines and tissue factor (TF). Activation of the transcription factor, nuclear factor kappa-B (NFκB), may be central to SAA-mediated endothelial cell inflammation, dysfunction and pro-thrombotic responses, while targeting NFκB with a pharmacologic inhibitor, BAY11-7082, may mitigate SAA activity. Human carotid artery endothelial cells (HCtAEC) were pre-incubated (1.5 h) with 10 µM BAY11-7082 or vehicle (control) followed by SAA (10 µg/mL; 4.5 h). Under these conditions gene expression for TF and Tumor Necrosis Factor (TNF) increased in SAA-treated HCtAEC and pre-treatment with BAY11-7082 significantly (TNF) and marginally (TF) reduced mRNA expression. Intracellular TNF and interleukin 6 (IL-6) protein also increased in HCtAEC supplemented with SAA and this expression was inhibited by BAY11-7082. Supplemented BAY11-7082 also significantly decreased SAA-mediated leukocyte adhesion to apolipoprotein E-deficient mouse aorta in ex vivo vascular flow studies. In vascular function studies, isolated aortic rings pre-treated with BAY11-7082 prior to incubation with SAA showed improved endothelium-dependent vasorelaxation and increased vascular cyclic guanosine monophosphate (cGMP) content. Together these data suggest that inhibition of NFκB activation may protect endothelial function by inhibiting the pro-inflammatory and pro-thrombotic activities of SAA.


Subject(s)
Aorta/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Leukocytes/metabolism , NF-kappa B/metabolism , Serum Amyloid A Protein/metabolism , Animals , Aorta/pathology , Atherosclerosis/etiology , Atherosclerosis/metabolism , Biomarkers , Cell Adhesion , Gene Expression Regulation , Humans , Immunohistochemistry , Inflammation Mediators , Leukocytes/immunology , Rats
12.
Front Pharmacol ; 8: 567, 2017.
Article in English | MEDLINE | ID: mdl-28970796

ABSTRACT

Fibrosis is a process of dysfunctional wound repair, described by a failure of tissue regeneration and excessive deposition of extracellular matrix, resulting in tissue scarring and subsequent organ deterioration. There are a broad range of stimuli that may trigger, and exacerbate the process of fibrosis, which can contribute to the growing rates of morbidity and mortality. Whilst the process of fibrosis is widely described and understood, there are no current standard treatments that can reduce or reverse the process effectively, likely due to the continuing knowledge gaps surrounding the cellular mechanisms involved. Several cellular targets have been implicated in the regulation of the fibrotic process including membrane domains, ion channels and more recently mechanosensors, specifically caveolae, particularly since these latter contain various signaling components, such as members of the TGFß and MAPK/ERK signaling pathways, all of which are key players in the process of fibrosis. This review explores the anti-fibrotic influences of the caveola, and in particular the key underpinning protein, caveolin-1, and its potential as a novel therapeutic target.

13.
Atherosclerosis ; 265: 47-53, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28858686

ABSTRACT

BACKGROUND AND AIMS: Monocyte levels predict cardiovascular outcomes and play a causal role in atherogenesis. Monocytes can be produced in the spleen and track to the atherosclerotic lesion in significant numbers. The cholinergic system has been shown to have anti-inflammatory actions in the spleen. We aimed to explore whether therapeutic stimulation of the nicotinic acetylcholine receptor alpha 7 (nAChRα7) can suppress atherogenesis. METHODS: Apoe-/- mice were placed on a Western-type diet and treated with bi-daily injections of the nAChRα7 agonist GTS-21 or vehicle every 2-3 days for 8 weeks. RESULTS: GTS-21 caused a reduction in atherosclerosis in the aortic arch and proximal aorta. This also resulted in less plaque macrophages. Moreover, GTS-21 reduced the abundance of blood monocytes, which was caused by inhibition of inflammatory cytokines and extramedullary hematopoiesis in the spleen, along with splenic monocytes. CONCLUSIONS: Stimulation of nAChRα7 with GTS-21 reduced atherosclerosis, which was associated with dampened splenic myelopoiesis.


Subject(s)
Aorta, Thoracic/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Benzylidene Compounds/pharmacology , Myelopoiesis/drug effects , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Spleen/drug effects , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Diet, Western , Disease Models, Animal , Male , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Spleen/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism
14.
Arterioscler Thromb Vasc Biol ; 37(7): 1301-1306, 2017 07.
Article in English | MEDLINE | ID: mdl-28522696

ABSTRACT

OBJECTIVE: Apolipoprotein A-I (apoA-I) mimetic peptides have antiatherogenic properties of high-density lipoprotein in vitro and have been shown to inhibit atherosclerosis in vivo. It is unclear, however, if each in vitro antiatherogenic property of these peptides translates to a corresponding activity in vivo, and if so, which of these contributes most to reduce atherosclerosis. APPROACH AND RESULTS: The effect of 7 apoA-I mimetic peptides, which were developed to selectively reproduce a specific component of the antiatherogenic properties of apoA-I, on the development of atherosclerosis was investigated in apolipoprotein E-deficient mice fed a high-fat diet for 4 or 12 weeks. The peptides include those that selectively upregulate cholesterol efflux, or are anti-inflammatory, or have antioxidation properties. All the peptides studied effectively inhibited the in vivo development of atherosclerosis in this model to the same extent. However, none of the peptides had the same selective effect in vivo as they had exhibited in vitro. None of the tested peptides affected plasma lipoprotein profile; capacity of plasma to support cholesterol efflux was increased modestly and similarly for all peptides. CONCLUSIONS: There is a discordance between the selective in vitro and in vivo functional properties of apoA-I mimetic peptides, and the in vivo antiatherosclerotic effect of apoA-I-mimetic peptides is independent of their in vitro functional profile. Comparing the properties of apoA-I mimetic peptides in plasma rather than in the lipid-free state is better for predicting their in vivo effects on atherosclerosis.


Subject(s)
Aortic Diseases/prevention & control , Apolipoprotein A-I/pharmacology , Atherosclerosis/prevention & control , Hypolipidemic Agents/pharmacology , Macrophages/drug effects , Peptide Fragments/pharmacology , Animals , Aortic Diseases/blood , Aortic Diseases/pathology , Apolipoprotein A-I/pharmacokinetics , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/pathology , Biological Mimicry , Biomarkers/blood , Cholesterol/blood , Diet, High-Fat , Disease Models, Animal , Genetic Predisposition to Disease , Lipoproteins/blood , Macrophages/metabolism , Male , Mice , Mice, Knockout , Peptide Fragments/pharmacokinetics , Phenotype , Plaque, Atherosclerotic , RAW 264.7 Cells , Tissue Distribution
15.
J Clin Invest ; 127(6): 2133-2147, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28504650

ABSTRACT

Platelets play a critical role in atherogenesis and thrombosis-mediated myocardial ischemia, processes that are accelerated in diabetes. Whether hyperglycemia promotes platelet production and whether enhanced platelet production contributes to enhanced atherothrombosis remains unknown. Here we found that in response to hyperglycemia, neutrophil-derived S100 calcium-binding proteins A8/A9 (S100A8/A9) interact with the receptor for advanced glycation end products (RAGE) on hepatic Kupffer cells, resulting in increased production of IL-6, a pleiotropic cytokine that is implicated in inflammatory thrombocytosis. IL-6 acts on hepatocytes to enhance the production of thrombopoietin, which in turn interacts with its cognate receptor c-MPL on megakaryocytes and bone marrow progenitor cells to promote their expansion and proliferation, resulting in reticulated thrombocytosis. Lowering blood glucose using a sodium-glucose cotransporter 2 inhibitor (dapagliflozin), depleting neutrophils or Kupffer cells, or inhibiting S100A8/A9 binding to RAGE (using paquinimod), all reduced diabetes-induced thrombocytosis. Inhibiting S100A8/A9 also decreased atherogenesis in diabetic mice. Finally, we found that patients with type 2 diabetes have reticulated thrombocytosis that correlates with glycated hemoglobin as well as increased plasma S100A8/A9 levels. These studies provide insights into the mechanisms that regulate platelet production and may aid in the development of strategies to improve on current antiplatelet therapies and to reduce cardiovascular disease risk in diabetes.


Subject(s)
Atherosclerosis/immunology , Calgranulin A/physiology , Calgranulin B/physiology , Diabetes Mellitus, Experimental/immunology , Neutrophils/metabolism , Thrombocytosis/immunology , Animals , Atherosclerosis/metabolism , Blood Platelets/physiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Thrombocytosis/metabolism
17.
Pharmacol Res ; 116: 77-86, 2017 02.
Article in English | MEDLINE | ID: mdl-27986554

ABSTRACT

OBJECTIVE: To determine whether a clinically-utilised IL-1 receptor antagonist, anakinra, reduces renal inflammation, structural damage and blood pressure (BP) in mice with established hypertension. METHODS: Hypertension was induced in male mice by uninephrectomy, deoxycorticosterone acetate (2.4mg/d,s.c.) and replacement of drinking water with saline (1K/DOCA/salt). Control mice received uninephrectomy, a placebo pellet and normal drinking water. 10days post-surgery, mice commenced treatment with anakinra (75mg/kg/d, i.p.) or vehicle (0.9% saline, i.p.) for 11days. Systolic BP was measured by tail cuff while qPCR, immunohistochemistry and flow cytometry were used to measure inflammatory markers, collagen and immune cell infiltration in the kidneys. RESULTS: By 10days post-surgery, 1K/DOCA/salt-treated mice displayed elevated systolic BP (148.3±2.4mmHg) compared to control mice (121.7±2.7mmHg; n=18, P<0.0001). The intervention with anakinra reduced BP in 1K/DOCA/salt-treated mice by ∼20mmHg (n=16, P<0.05), but had no effect in controls. In 1K/DOCA/salt-treated mice, anakinra modestly reduced (∼30%) renal expression of some (CCL5, CCL2; n=7-8; P<0.05) but not all (ICAM-1, IL-6) inflammatory markers, and had no effect on immune cell infiltration (n=7-8, P>0.05). Anakinra reduced renal collagen content (n=6, P<0.01) but paradoxically appeared to exacerbate the renal and glomerular hypertrophy (n=8-9, P<0.001) that accompanied 1K/DOCA/salt-induced hypertension. CONCLUSION: Despite its anti-hypertensive and renal anti-fibrotic actions, anakinra had minimal effects on inflammation and leukocyte infiltration in mice with 1K/DOCA/salt-induced hypertension. Future studies will assess whether the anti-hypertensive actions of anakinra are mediated by protective actions in other BP-regulating or salt-handling organs such as the arteries, skin and brain.


Subject(s)
Blood Pressure/drug effects , Fibrosis/drug therapy , Hypertension, Renal/drug therapy , Interleukin 1 Receptor Antagonist Protein/pharmacology , Kidney Diseases/drug therapy , Kidney/drug effects , Animals , Antihypertensive Agents/pharmacology , Biomarkers/metabolism , Desoxycorticosterone Acetate/pharmacology , Fibrosis/metabolism , Hypertension, Renal/chemically induced , Hypertension, Renal/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Kidney Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Sodium Chloride, Dietary/pharmacology
18.
Clin Sci (Lond) ; 130(18): 1629-40, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27231254

ABSTRACT

Nitroxyl anion (HNO) donors are currently being assessed for their therapeutic utility in several cardiovascular disorders including heart failure. Here, we examine their effect on factors that precede atherosclerosis including endothelial cell and monocyte activation, leucocyte adhesion to the endothelium and macrophage polarization. Similar to the NO donor glyceryl trinitrate (GTN), the HNO donors Angeli's salt (AS) and isopropylamine NONOate (IPA/NO) decreased leucocyte adhesion to activated human umbilical vein endothelial cells (HUVECs) and mouse isolated aorta. This reduction in adhesion was accompanied by a reduction in intercellular adhesion molecule-1 (ICAM-1) and the cytokines monocyte chemoattractant protein 1 (MCP-1) and interleukin 6 (IL-6) which was inhibitor of nuclear factor κB (NFκB) α (IκBα)- and subsequently NFκB-dependent. Intriguingly, the effects of AS on leucocyte adhesion, like those on vasodilation, were found to not be susceptible to pharmacological tolerance, unlike those observed with GTN. As well, HNO reduces monocyte activation and promotes polarization of M2 macrophages. Taken together, our data demonstrate that HNO donors can reduce factors that are associated with and which precede atherosclerosis and may thus be useful therapeutically. Furthermore, since the effects of the HNO donors were not subject to tolerance, this confers an additional advantage over NO donors.


Subject(s)
Atherosclerosis/drug therapy , Cell Polarity/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Macrophages/drug effects , Monocytes/cytology , Monocytes/drug effects , Nitrogen Oxides/administration & dosage , Animals , Aorta/drug effects , Aorta/immunology , Aorta/physiopathology , Atherosclerosis/immunology , Atherosclerosis/physiopathology , Chemokine CCL2/immunology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/immunology , Humans , Intercellular Adhesion Molecule-1/immunology , Interleukin-6/immunology , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology
19.
Br J Pharmacol ; 173(4): 741-51, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26332942

ABSTRACT

BACKGROUND AND PURPOSE: Monocyte-derived macrophages are critical in the development of atherosclerosis and can adopt a wide range of functional phenotypes depending on their surrounding milieu. High-density lipoproteins (HDLs) have many cardio-protective properties including potent anti-inflammatory effects. We investigated the effects of HDL on human macrophage phenotype and the mechanisms by which these occur. EXPERIMENTAL APPROACH: Human blood monocytes were differentiated into macrophages in the presence or absence of HDL and were then induced to either an inflammatory macrophage (M1) or anti-inflammatory macrophage (M2) phenotype using LPS and IFN-γ or IL-4, respectively. KEY RESULTS: HDL inhibited the induction of macrophages to an M1-phenotype, as evidenced by a decrease in the expression of M1-specific cell surface markers CD192 and CD64, as well as M1-associated inflammatory genes TNF-α, IL-6 and MCP-1 (CCL2). HDL also inhibited M1 function by reducing the production of ROS. In contrast, HDL had no effect on macrophage induction to the M2-phenotype. Similarly, methyl-ß-cyclodextrin, a non-specific cholesterol acceptor also suppressed the induction of M1 suggesting that cholesterol efflux is important in this process. Furthermore, HDL decreased membrane caveolin-1 in M1 macrophages. We confirmed that caveolin-1 is required for HDL to inhibit M1 induction as bone marrow-derived macrophages from caveolin-1 knockout mice continued to polarize into M1-phenotype despite the presence of HDL. Moreover, HDL decreased ERK1/2 and STAT3 phosphorylation in M1 macrophages. CONCLUSIONS AND IMPLICATIONS: We concluded that HDL reduces the induction of macrophages to the inflammatory M1-phenotype via redistribution of caveolin-1, preventing the activation of ERK1/2 and STAT3.


Subject(s)
Caveolin 1/metabolism , Lipoproteins, HDL/pharmacology , Macrophages/cytology , Macrophages/drug effects , Animals , Caveolin 1/deficiency , Humans , Lipoproteins, HDL/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Phenotype , Reactive Oxygen Species/metabolism
20.
Br J Pharmacol ; 173(4): 729-40, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25560767

ABSTRACT

BACKGROUND AND PURPOSE: Angiotensin AT2 receptors are upregulated in disease states such as atherosclerosis and blockade of the AT2 receptors exacerbates plaque formation. Direct stimulation of these receptors is anti-atherogenic but the mechanisms and pathways involved remain unknown. We examined the effect of direct AT2 receptor stimulation with Compound 21 (C21) on the leukocyte adhesion cascade in vitro, right through to plaque formation in vivo. EXPERIMENTAL APPROACH: Effects of C21 on TNFα-induced inflammation were assessed in human umbilical vein endothelial cells (HUVECs), activation of monocytes, polarisation of monocyte-derived macrophages and in intact mouse aortae. KEY RESULTS: C21 attenuated TNFα-induced: monocyte adhesion to cultured HUVECs, adhesion molecule expression and abolished TNFα-induced ROS production. TNFα-induced NFκB translocation from the cytoplasm to the nucleus, essential for cytokine production, was prevented by C21. C21 did not influence monocyte activation or macrophage polarisation but did reduce TNFα and IL-6 mRNA expression in M1 macrophages. The anti-inflammatory effects of C21 were abolished by an AT2 receptor antagonist confirming that the effects of C21 were AT2 receptor-mediated. Also, leukocyte adhesion and cytokine gene expression, induced by high-fat diet (HFD), was attenuated in ApoE(-/-) mice treated with C21. Plaque size and stability were improved with C21 treatment with increased smooth muscle cell composition and decreased lipid size, compared with HFD-saline treated mice. CONCLUSION AND IMPLICATIONS: C21 prevented TNFα-induced and HFD-induced vascular inflammation in vitro and in vivo. Our data provide strong evidence that the anti-atherosclerotic actions of C21 were due to vascular anti-inflammatory effects, mediated by AT2 receptors.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Inflammation/drug therapy , Leukocytes/drug effects , Receptor, Angiotensin, Type 2/agonists , Sulfonamides/pharmacology , Thiophenes/pharmacology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Leukocytes/cytology , Leukocytes/metabolism , Male , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sulfonamides/chemistry , Thiophenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL