Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Arterioscler Thromb Vasc Biol ; 41(1): 430-445, 2021 01.
Article in English | MEDLINE | ID: mdl-33147993

ABSTRACT

OBJECTIVE: To determine whether global reduction of CD68 (cluster of differentiation) macrophages impacts the development of experimental pulmonary arterial hypertension (PAH) and whether this reduction affects the balance of pro- and anti-inflammatory macrophages within the lung. Additionally, to determine whether there is evidence of an altered macrophage polarization in patients with PAH. Approach and Results: Macrophage reduction was induced in mice via doxycycline-induced CD68-driven cytotoxic diphtheria toxin A chain expression (macrophage low [MacLow] mice). Chimeric mice were generated using bone marrow transplant. Mice were phenotyped for PAH by echocardiography and closed chest cardiac catheterization. Murine macrophage phenotyping was performed on lungs, bone marrow-derived macrophages, and alveolar macrophages using immunohistochemical and flow cytometry. Monocyte-derived macrophages were isolated from PAH patients and healthy volunteers and polarization capacity assessed morphologically and by flow cytometry. After 6 weeks of macrophage depletion, male but not female MacLow mice developed PAH. Chimeric mice demonstrated a requirement for both MacLow bone marrow and MacLow recipient mice to cause PAH. Immunohistochemical analysis of lung sections demonstrated imbalance in M1/M2 ratio in male MacLow mice only, suggesting that this imbalance may drive the PAH phenotype. M1/M2 imbalance was also seen in male MacLow bone marrow-derived macrophages and PAH patient monocyte-derived macrophages following stimulation with doxycycline and IL (interleukin)-4, respectively. Furthermore, MacLow-derived alveolar macrophages showed characteristic differences in terms of their polarization and expression of diphtheria toxin A chain following stimulation with doxycycline. CONCLUSIONS: These data further highlight a sex imbalance in PAH and further implicate immune cells into this paradigm. Targeting imbalance of macrophage population may offer a future therapeutic option.


Subject(s)
Macrophage Activation , Macrophages, Alveolar/pathology , Muscle, Smooth, Vascular/pathology , Pulmonary Arterial Hypertension/pathology , Vascular Remodeling , Adult , Aged , Animals , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Case-Control Studies , Cell Proliferation , Diphtheria Toxin/genetics , Disease Models, Animal , Female , Humans , Hypoxia/complications , Macrophages, Alveolar/metabolism , Male , Mice, Transgenic , Middle Aged , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Paracrine Communication , Peptide Fragments/genetics , Phenotype , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Sex Factors
3.
J Dairy Res ; 87(4): 397-399, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33168112

ABSTRACT

This research communication presents a study evaluating the effects of dried sainfoin (Onobrychis viciifolia) supplemented to dairy goats on their milking performance and feed protein efficiency under commercial conditions. During July and August 2015, a herd of 20 Alpine goats was divided into two treatments (n = 10), balanced by milk yield and days in milk. They were supplied with either 700 g/d sainfoin pellets (condensed tannins: 4.0 g/kg DM) or 700 g/d lucerne (Medicago sativa) pellets (condensed tannins: 0.3 g/kg DM). The goats remained in one herd and were separated by treatments only during milking. In the milking parlour each goat received 350 g of the respective pellets, twice daily. During the day, the herd had 5 h access to a high-quality pasture (crude protein >200 g/kg DM), whilst during the rest of the day and the night animals were housed and offered grass hay ad libitum. The experiment lasted for seven weeks. Individual milk yields and composition were controlled in weeks 1, 3, 5, and 7 after the start of the experiment. No differences between the treatments were found, either for milk, protein or urea yields, nor for protein, urea and fat concentrations. Urea to protein ratio in milk was lower with the sainfoin treatment. In conclusion, sainfoin compared to lucerne, supplied for 7 weeks to dairy goats at approximately 25% of the diet, had only weak beneficial and no adverse effects on milking performance, milk composition and feed protein efficiency under commercial conditions of pasture-based dairy production.


Subject(s)
Animal Feed , Dietary Proteins , Fabaceae/chemistry , Goats/physiology , Tannins , Animal Nutritional Physiological Phenomena , Animals , Diet/veterinary , Feeding Behavior , Female , Nutritive Value
4.
Nat Commun ; 10(1): 5183, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729368

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare but fatal disease. Current treatments increase life expectancy but have limited impact on the progressive pulmonary vascular remodelling that drives PAH. Osteoprotegerin (OPG) is increased within serum and lesions of patients with idiopathic PAH and is a mitogen and migratory stimulus for pulmonary artery smooth muscle cells (PASMCs). Here, we report that the pro-proliferative and migratory phenotype in PASMCs stimulated with OPG is mediated via the Fas receptor and that treatment with a human antibody targeting OPG can attenuate pulmonary vascular remodelling associated with PAH in multiple rodent models of early and late treatment. We also demonstrate that the therapeutic efficacy of the anti-OPG antibody approach in the presence of standard of care vasodilator therapy is mediated by a reduction in pulmonary vascular remodelling. Targeting OPG with a therapeutic antibody is a potential treatment strategy in PAH.


Subject(s)
Antibodies/administration & dosage , Familial Primary Pulmonary Hypertension/drug therapy , Osteoprotegerin/metabolism , Animals , Cell Movement/drug effects , Disease Models, Animal , Familial Primary Pulmonary Hypertension/genetics , Familial Primary Pulmonary Hypertension/metabolism , Familial Primary Pulmonary Hypertension/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Osteoprotegerin/genetics , Protein Binding , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats , Rats, Wistar , Vascular Remodeling/drug effects
5.
Am J Respir Crit Care Med ; 199(2): 199-210, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30211629

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is characterized by vascular cell proliferation and endothelial cell apoptosis. TLR3 (Toll-like receptor 3) is a receptor for double-stranded RNA and has been recently implicated in vascular protection. OBJECTIVES: To study the expression and role of TLR3 in PAH and to determine whether a TLR3 agonist reduces pulmonary hypertension in preclinical models. METHODS: Lung tissue and endothelial cells from patients with PAH were investigated by polymerase chain reaction, immunofluorescence, and apoptosis assays. TLR3-/- and TLR3+/+ mice were exposed to chronic hypoxia and SU5416. Chronic hypoxia or chronic hypoxia/SU5416 rats were treated with the TLR3 agonist polyinosinic/polycytidylic acid (Poly[I:C]). MEASUREMENTS AND MAIN RESULTS: TLR3 expression was reduced in PAH patient lung tissue and endothelial cells, and TLR3-/- mice exhibited more severe pulmonary hypertension following exposure to chronic hypoxia/SU5416. TLR3 knockdown promoted double-stranded RNA signaling via other intracellular RNA receptors in endothelial cells. This was associated with greater susceptibility to apoptosis, a known driver of pulmonary vascular remodeling. Poly(I:C) increased TLR3 expression via IL-10 in rat endothelial cells. In vivo, high-dose Poly(I:C) reduced pulmonary hypertension in both rat models in proof-of-principle experiments. In addition, Poly(I:C) also reduced right ventricular failure in established pulmonary hypertension. CONCLUSIONS: Our work identifies a novel role for TLR3 in PAH based on the findings that reduced expression of TLR3 contributes to endothelial apoptosis and pulmonary vascular remodeling.


Subject(s)
Hypertension, Pulmonary/genetics , Toll-Like Receptor 3/genetics , Animals , Disease Models, Animal , Humans , Hypertension, Pulmonary/metabolism , Lung/metabolism , Mice , Rats , Signal Transduction , Toll-Like Receptor 3/metabolism
6.
BMC Pulm Med ; 18(1): 41, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29499691

ABSTRACT

BACKGROUND: Aldosterone is a mineralocorticoid hormone critically involved in arterial blood pressure regulation. Although pharmacological aldosterone antagonism reduces mortality and morbidity among patients with severe left-sided heart failure, the contribution of aldosterone to the pathobiology of pulmonary arterial hypertension (PAH) and right ventricular (RV) heart failure is not fully understood. METHODS: The effects of Eplerenone (0.1% Inspra® mixed in chow) on pulmonary vascular and RV remodeling were evaluated in mice with pulmonary hypertension (PH) caused by Sugen5416 injection with concomitant chronic hypoxia (SuHx) and in a second animal model with established RV dysfunction independent from lung remodeling through surgical pulmonary artery banding. RESULTS: Preventive Eplerenone administration attenuated the development of PH and pathological remodeling of pulmonary arterioles. Therapeutic aldosterone antagonism - starting when RV dysfunction was established - normalized mineralocorticoid receptor gene expression in the right ventricle without direct effects on either RV structure (Cardiomyocyte hypertrophy, Fibrosis) or function (assessed by non-invasive echocardiography along with intra-cardiac pressure volume measurements), but significantly lowered systemic blood pressure. CONCLUSIONS: Our data indicate that aldosterone antagonism with Eplerenone attenuates pulmonary vascular rather than RV remodeling in PAH.


Subject(s)
Eplerenone/pharmacology , Heart Ventricles/pathology , Hypertension, Pulmonary/drug therapy , Ventricular Dysfunction, Right/drug therapy , Ventricular Remodeling/drug effects , Animals , Arterial Pressure/drug effects , Disease Models, Animal , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mineralocorticoid Receptor Antagonists/pharmacology , Pulmonary Artery/physiopathology , Ventricular Dysfunction, Right/physiopathology
7.
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.

8.
J Clin Invest ; 126(7): 2495-508, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27214554

ABSTRACT

Loss of the growth-suppressive effects of bone morphogenetic protein (BMP) signaling has been demonstrated to promote pulmonary arterial endothelial cell dysfunction and induce pulmonary arterial smooth muscle cell (PASMC) proliferation, leading to the development of pulmonary arterial hypertension (PAH). MicroRNAs (miRs) mediate higher order regulation of cellular function through coordinated modulation of mRNA targets; however, miR expression is altered by disease development and drug therapy. Here, we examined treatment-naive patients and experimental models of PAH and identified a reduction in the levels of miR-140-5p. Inhibition of miR-140-5p promoted PASMC proliferation and migration in vitro. In rat models of PAH, nebulized delivery of miR-140-5p mimic prevented the development of PAH and attenuated the progression of established PAH. Network and pathway analysis identified SMAD-specific E3 ubiquitin protein ligase 1 (SMURF1) as a key miR-140-5p target and regulator of BMP signaling. Evaluation of human tissue revealed that SMURF1 is increased in patients with PAH. miR-140-5p mimic or SMURF1 knockdown in PASMCs altered BMP signaling, further supporting these factors as regulators of BMP signaling. Finally, Smurf1 deletion protected mice from PAH, demonstrating a critical role in disease development. Together, these studies identify both miR-140-5p and SMURF1 as key regulators of disease pathology and as potential therapeutic targets for the treatment of PAH.


Subject(s)
Hypertension, Pulmonary/metabolism , MicroRNAs/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Aged , Animals , Humans , Hypoxia , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Phenotype , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction
9.
Thromb Haemost ; 116(1): 181-90, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27075869

ABSTRACT

Blood flow generates wall shear stress (WSS) which alters endothelial cell (EC) function. Low WSS promotes vascular inflammation and atherosclerosis whereas high uniform WSS is protective. Ivabradine decreases heart rate leading to altered haemodynamics. Besides its cardio-protective effects, ivabradine protects arteries from inflammation and atherosclerosis via unknown mechanisms. We hypothesised that ivabradine protects arteries by increasing WSS to reduce vascular inflammation. Hypercholesterolaemic mice were treated with ivabradine for seven weeks in drinking water or remained untreated as a control. En face immunostaining demonstrated that treatment with ivabradine reduced the expression of pro-inflammatory VCAM-1 (p<0.01) and enhanced the expression of anti-inflammatory eNOS (p<0.01) at the inner curvature of the aorta. We concluded that ivabradine alters EC physiology indirectly via modulation of flow because treatment with ivabradine had no effect in ligated carotid arteries in vivo, and did not influence the basal or TNFα-induced expression of inflammatory (VCAM-1, MCP-1) or protective (eNOS, HMOX1, KLF2, KLF4) genes in cultured EC. We therefore considered whether ivabradine can alter WSS which is a regulator of EC inflammatory activation. Computational fluid dynamics demonstrated that ivabradine treatment reduced heart rate by 20 % and enhanced WSS in the aorta. In conclusion, ivabradine treatment altered haemodynamics in the murine aorta by increasing the magnitude of shear stress. This was accompanied by induction of eNOS and suppression of VCAM-1, whereas ivabradine did not alter EC that could not respond to flow. Thus ivabradine protects arteries by altering local mechanical conditions to trigger an anti-inflammatory response.


Subject(s)
Arteries/drug effects , Arteritis/prevention & control , Benzazepines/pharmacology , Heart Rate/drug effects , Animals , Arteries/physiology , Arteritis/physiopathology , Biomechanical Phenomena , Cardiovascular Agents/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Heart Rate/physiology , Human Umbilical Vein Endothelial Cells , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/drug therapy , Hypercholesterolemia/physiopathology , Ivabradine , Kruppel-Like Factor 4 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Stress, Mechanical , Vascular Cell Adhesion Molecule-1/metabolism
10.
Circ Cardiovasc Interv ; 8(11): e002569, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26553697

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension is a devastating disease with high morbidity and mortality and limited treatment options. Recent studies have shown that pulmonary artery denervation improves pulmonary hemodynamics in an experimental model and in an early clinical trial. We aimed to evaluate the nerve distribution around the pulmonary artery, to determine the effect of radiofrequency pulmonary artery denervation on acute pulmonary hypertension induced by vasoconstriction, and to demonstrate denervation of the pulmonary artery at a histological level. METHODS AND RESULTS: Histological evaluation identified a circumferential distribution of nerves around the proximal pulmonary arteries. Nerves were smaller in diameter, greater in number, and located in closer proximity to the luminal aspect of the pulmonary arterial wall beyond the pulmonary artery bifurcation. To determine the effect of pulmonary arterial denervation acute pulmonary hypertension was induced in 8 pigs by intravenous infusion of thromboxane A2 analogue. Animals were assigned to either pulmonary artery denervation, using a prototype radiofrequency catheter and generator, or a sham procedure. Pulmonary artery denervation resulted in reduced mean pulmonary artery pressure and pulmonary vascular resistance and increased cardiac output. Ablation lesions on the luminal surface of the pulmonary artery were accompanied by histological and biochemical alteration in adventitial nerves and correlated with improved hemodynamic parameters. CONCLUSIONS: Pulmonary artery denervation offers the possibility of a new treatment option for patients with pulmonary arterial hypertension. Further work is required to determine the long-term efficacy and safety.


Subject(s)
Autonomic Pathways/surgery , Catheter Ablation , Denervation , Hypertension, Pulmonary/therapy , Pulmonary Artery/surgery , Acute Disease , Animals , Autonomic Pathways/metabolism , Blood Pressure , Cardiac Output , Disease Models, Animal , Hemodynamics , Humans , Pulmonary Artery/innervation , Pulmonary Artery/pathology , Swine , Vascular Resistance , Vasoconstriction
11.
EuroIntervention ; 10(6): 709-16, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25330503

ABSTRACT

AIMS: The success of percutaneous coronary intervention (PCI) has been limited by restenosis and stent thrombosis. Delayed or incomplete endothelial regeneration is believed to be a key factor responsible for these events. Developing a stent with an accelerated healing profile may be of benefit. We aimed to evaluate the feasibility and safety of seeding a bare metal stent (BMS) with human trophoblastic endovascular progenitor cells (hTEC) derived from human embryonic stem cells. A porcine coronary artery model was used to compare the rate and extent of endothelial regeneration and the degree of neointimal proliferation. Characterisation of hTEC confirmed a mixed progenitor and endothelial cell phenotype. The biodistribution and fate of hTEC were studied using radiolabelled 111Indium oxine and fluorescent in situ hybridisation. Scanning electron microscopy showed earlier endothelial coverage in hTEC-seeded stents as compared to similar BMS. hTEC-seeded BMS achieved complete stent coverage in three days. Quantitative coronary angiography, intravascular ultrasound assessment and histomorphometry showed no difference in neointimal hyperplasia between hTEC-seeded and control BMS. hTEC seeding of coronary stents is a novel and safe approach to accelerate endothelial regeneration without increasing neointimal proliferation.


Subject(s)
Endothelium, Vascular/physiology , Guided Tissue Regeneration , Stem Cells/physiology , Stents , Animals , Cell Proliferation , Cells, Cultured , Coronary Angiography , Embryonic Stem Cells/physiology , Microscopy, Electron, Scanning , Models, Animal , Neointima/pathology , Swine , Trophoblasts/cytology , Ultrasonography, Interventional
12.
J Exp Med ; 209(11): 1919-35, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23071256

ABSTRACT

Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by the progressive narrowing and occlusion of small pulmonary arteries. Current therapies fail to fully reverse this vascular remodeling. Identifying key pathways in disease pathogenesis is therefore required for the development of new-targeted therapeutics. We have previously reported tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) immunoreactivity within pulmonary vascular lesions from patients with idiopathic PAH and animal models. Because TRAIL can induce both endothelial cell apoptosis and smooth muscle cell proliferation in the systemic circulation, we hypothesized that TRAIL is an important mediator in the pathogenesis of PAH. We demonstrate for the first time that TRAIL is a potent stimulus for pulmonary vascular remodeling in human cells and rodent models. Furthermore, antibody blockade or genetic deletion of TRAIL prevents the development of PAH in three independent rodent models. Finally, anti-TRAIL antibody treatment of rodents with established PAH reverses pulmonary vascular remodeling by reducing proliferation and inducing apoptosis, improves hemodynamic indices, and significantly increases survival. These preclinical investigations are the first to demonstrate the importance of TRAIL in PAH pathogenesis and highlight its potential as a novel therapeutic target to direct future translational therapies.


Subject(s)
Antibodies/pharmacology , Hypertension, Pulmonary/prevention & control , Myocytes, Smooth Muscle/drug effects , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Adult , Animals , Antibodies/immunology , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Expression/drug effects , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology , Young Adult
13.
Thorax ; 67(9): 796-803, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22496351

ABSTRACT

BACKGROUND: The death receptor ligand tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) shows considerable clinical promise as a therapeutic agent. TRAIL induces leukocyte apoptosis, reducing acute inflammatory responses in the lung. It is not known whether TRAIL modifies chronic lung injury or whether TRAIL has a role in human idiopathic pulmonary fibrosis (IPF). We therefore explored the capacity of TRAIL to modify chronic inflammatory lung injury and studied TRAIL expression in patients with IPF. METHODS: TRAIL(-/-) and wild-type mice were instilled with bleomycin and inflammation assessed at various time points by bronchoalveolar lavage and histology. Collagen deposition was measured by tissue hydroxyproline content. TRAIL expression in human IPF lung samples was assessed by immunohistochemistry and peripheral blood TRAIL measured by ELISA. RESULTS: TRAIL(-/-) mice had an exaggerated delayed inflammatory response to bleomycin, with increased neutrophil numbers (mean 3.19±0.8 wild type vs 11.5±5.4×10(4) TRAIL(-/-), p<0.0001), reduced neutrophil apoptosis (5.42±1.6% wild type vs 2.47±0.5% TRAIL(-/-), p=0.0003) and increased collagen (3.45±0.2 wild type vs 5.8±1.3 mg TRAIL(-/-), p=0.005). Immunohistochemical analysis showed induction of TRAIL in bleomycin-treated wild-type mice. Patients with IPF demonstrated lower levels of TRAIL expression than in control lung biopsies and their serum levels of TRAIL were significantly lower compared with matched controls (38.1±9.6 controls vs 32.3±7.2 pg/ml patients with IPF, p=0.002). CONCLUSION: These data suggest TRAIL may exert beneficial, anti-inflammatory actions in chronic pulmonary inflammation in murine models and that these mechanisms may be compromised in human IPF.


Subject(s)
Lung Injury/metabolism , Pulmonary Fibrosis/metabolism , TNF-Related Apoptosis-Inducing Ligand/deficiency , Animals , Biomarkers/metabolism , Bleomycin , Bronchoalveolar Lavage , Case-Control Studies , Collagen/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hydroxyproline/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Respiratory Function Tests
14.
Biology (Basel) ; 1(1): 43-57, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-24832046

ABSTRACT

Development of the atherosclerotic plaque involves a complex interplay between a number of cell types and an extensive inter-cellular communication via cell bound as well as soluble mediators. The family of tribbles proteins has recently been identified as novel controllers of pro-inflammatory signal transduction. The objective of this study was to address the expression pattern of all three tribbles proteins in atherosclerotic plaques from a mouse model of atherosclerosis. Each tribbles were expressed in vascular smooth muscle cells, endothelial cells as well as in resident macrophages of mouse atherosclerotic plaques. The role of IL-1 mediated inflammatory events in controlling tribbles expression was also addressed by inducing experimental atherosclerosis in ApoE-/-IL1R1-/- (double knockout) mice. Immunohistochemical analysis of these mice showed a selective decrease in the percentage of trb-1 expressing macrophages, compared to the ApoE-/- cohort (14.7% ± 1.55 vs. 26.3% ± 1.19). The biological significance of this finding was verified in vitro where overexpression of trb-1 in macrophages led to a significant attenuation (~70%) of IL-6 production as well as a suppressed IL-12 expression induced by a proinflammatory stimulus. In this in vitro setting, expression of truncated trb-1 mutants suggests that the kinase domain of this protein is sufficient to exert this inhibitory action.

15.
Am J Pathol ; 179(4): 1693-705, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21835155

ABSTRACT

Inflammatory mechanisms are proposed to play a significant role in the pathogenesis of pulmonary arterial hypertension (PAH). Previous studies have described PAH in fat-fed apolipoprotein E knockout (ApoE(-/-)) mice. We have reported that signaling in interleukin-1-receptor-knockout (IL-1R1(-/-)) mice leads to a reduction in diet-induced systemic atherosclerosis. We subsequently hypothesized that double-null (ApoE(-/-)/IL-1R1(-/-)) mice would show a reduced PAH phenotype compared with that of ApoE(-/-) mice. Male IL-1R1(-/-), ApoE(-/-), and ApoE(-/-)/IL-1R1(-/-) mice were fed regular chow or a high-fat diet (Paigen diet) for 8 weeks before phenotyping for PAH. No abnormal phenotype was observed in the IL-1R1(-/-) mice. Fat-fed ApoE(-/-) mice developed significantly increased right ventricular systolic pressure and substantial pulmonary vascular remodeling. Surprisingly, ApoE(-/-)/IL-1R1(-/-) mice showed an even more severe PAH phenotype. Further molecular investigation revealed the expression of a putative, alternatively primed IL-1R1 transcript expressed within the lungs but not aorta of ApoE(-/-)/IL-1R1(-/-) mice. Treatment of ApoE(-/-) and ApoE(-/-)/IL-1R1(-/-) mice with IL-1-receptor antagonist prevented progression of the PAH phenotype in both strains. Blocking IL-1 signaling may have beneficial effects in treating PAH, and alternative IL-1-receptor signaling in the lung may be important in driving PAH pathogenesis.


Subject(s)
Apolipoproteins E/deficiency , Diet, High-Fat/adverse effects , Hypertension, Pulmonary/pathology , Interleukin-1/metabolism , Animals , Apolipoproteins E/metabolism , Biomarkers/metabolism , Disease Progression , Feeding Behavior/drug effects , Hemodynamics/drug effects , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/complications , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Inflammation Mediators/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/drug effects , Osteoprotegerin/metabolism , Phenotype , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Up-Regulation/drug effects , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects
16.
Ann Biomed Eng ; 39(10): 2615-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21785884

ABSTRACT

A model that combines the results of in vivo experiment, 3D image data, and computer simulation has been developed. Twelve identical stents were implanted into six healthy pigs and explanted at a range of different post-recovery periods from 6 h to 28 days. The stented vessel segments were embedded in methacrylate resin for the preparation of transverse histological sections and imaged using ultra-high resolution micro-CT. The resulting CT data was used to reconstruct the 3D geometry of the stents and one case was used to inform a 3D computational fluid dynamic model. Derived hemodynamic parameters such as wall shear stress (WSS), axial WSS, and oscillatory shear index were correlated with the distribution of neointimal hyperplasia, assessed from histomorphometric analyses. The direct comparison of hemodynamic parameters and biological response supports the hypothesis that low and oscillatory WSS lead to a greater neointimal response within the stented region. Moreover, the realistic geometry obtained from micro-CT images, characterized by proximal overexpansion and asymmetric deployment of the stent, leads to a markedly non-uniform distribution of WSS values and correlates with asymmetric neo-intimal growth. This correlation cannot be appreciated from studies of idealized geometries.


Subject(s)
Computer Simulation , Graft Occlusion, Vascular/physiopathology , Hemodynamics/physiology , Hyperplasia/physiopathology , Stents/adverse effects , Animals , Imaging, Three-Dimensional , Models, Animal , Models, Cardiovascular , Stress, Physiological/physiology , Surgery, Computer-Assisted , Swine , X-Ray Microtomography
17.
Cardiovasc Res ; 85(1): 38-44, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19633315

ABSTRACT

AIMS: Animal models of stenting are mostly limited to larger animals or involve substantial abdominal surgery in rodents. We aimed to develop a simple, direct model of murine stenting. METHODS AND RESULTS: We designed a miniature, self-expanding, nitinol wire coil stent that was pre-loaded into a metal stent sheath. This was advanced into the abdominal aorta of the mouse, via femoral access, and the stent deployed. In-stent restenosis was investigated at 1, 3, 7, and 28 days post-stenting. The model was validated by investigation of neointima formation in mice deficient in signalling via the interleukin-1 receptor (IL-1R1), compared with other injury models. Ninety-two per cent of mice undergoing the procedure were successfully stented. All stented vessels were patent. Inflammatory cells were seen in the adventitia and around the stent strut up to 3 days post-stenting. At 3 days, an early neointima was present, building to a mature neointima at 28 days. In mice lacking IL-1R1, the neointima was 64% smaller than that in wild-type controls at the 28-day timepoint, in agreement with other models. CONCLUSION: This is the first description of a successful model of murine in situ stenting, using a stent specifically tailored for use in small thin-walled arteries. The procedure can be undertaken by a single operator without the need for an advanced level of microsurgical skill and is reliable and reproducible. The utility of this model is demonstrated by a reduction in in-stent restenosis in IL-1R1-deficient mice.


Subject(s)
Models, Animal , Stents , Animals , Aorta, Abdominal/pathology , Mice , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/physiology , Stents/adverse effects , Tunica Intima/pathology
18.
Arterioscler Thromb Vasc Biol ; 27(4): 833-40, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17204666

ABSTRACT

OBJECTIVE: We aimed to develop and validate a model of angioplasty and stenting in mice that would allow investigation of the response to stent injury using genetically modified mouse strains. METHODS AND RESULTS: Aortic segments from either C57BL/6 wild-type or atherosclerotic ApoE-KO mice underwent balloon angioplasty alone or balloon angioplasty and stenting with a 1.25x2.5 mm stainless steel stent. Vessels were carotid-interposition grafted into genetically identical littermate recipients and harvested at 1, 7, 14, or 28 days. In wild-type mice, stenting generated an inflammatory vascular injury response between days 1 to 7, leading to the development of neointimal hyperplasia by day 14, which further increased in area by day 28 leading to the development of in-stent stenosis. Uninjured vessels and vessels injured by balloon angioplasty alone developed minimal neointimal hyperplasia. In stented ApoE-KO mice, neointimal area at 28 days was 30% greater compared with wild-type mice. CONCLUSIONS: By reproducing important features of human stenting in atherosclerotic mice, we provide the potential to investigate molecular pathways and evaluate novel therapeutic targets for stent injury and restenosis.


Subject(s)
Angioplasty, Balloon/adverse effects , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Atherosclerosis/metabolism , Stents/adverse effects , Animals , Aorta, Thoracic/injuries , Aorta, Thoracic/pathology , Aorta, Thoracic/transplantation , Carotid Arteries/surgery , Constriction, Pathologic/etiology , Female , Hyperplasia , Immunochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors , Tunica Intima/pathology , Vasculitis/etiology , Vasculitis/pathology
19.
Catheter Cardiovasc Interv ; 69(2): 209-15, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17195202

ABSTRACT

BACKGROUND: While several endovascular techniques have been developed for treating arterial bifurcation lesions, there is, as yet, no single, widely accepted technique for treating left main stem (LMS) bifurcation lesions with stents. The simultaneous kissing stent (SCS) technique seems particularly suited for such lesions. The authors describe a consecutive cohort of patients with LMS bifurcation stenosis treated with this technique and present mechanistic insights from a porcine model. METHODS: Thirty consecutive patients with LMS bifurcation stenosis +/- multivessel disease were treated with SCS technique using paclitaxel-eluting stents. The technique involves simultaneous implantation of two stents (LMS-LAD and LMS-Cx) so that the proximal end of both stents lie at the same level in the body of LMS. Symptom status was recorded and follow-up angiography was performed. In addition, four pigs underwent SCS implantation of the LMS and were studied by direct examination, histology, and scanning electron microscopy. RESULTS: The patients' mean age was 63, 26 were elective and 19 were male. Paclitaxel-eluting stents (Taxus, Boston Scientific) were used. There was one in-hospital death in a highly unstable patient. At 6-month follow-up angiography, two patients required target lesion revascularization. At 13 +/- 3 month follow-up, the remaining patients remain well, with no other major adverse cardiac events. The animals all survived with patent stents. SEM demonstrated full re-endothelialization and histology showed a thin but complete neointima covering the stent struts. CONCLUSION: The SCS technique for treating LMS bifurcation disease with paclitaxel-eluting stents is safe and feasible. Medium term clinical and angiographic results are good. An endothelium and a thin neointima cover the metal struts.


Subject(s)
Blood Vessel Prosthesis Implantation/instrumentation , Coronary Stenosis/therapy , Stents , Aged , Animals , Coronary Angiography , Drug Delivery Systems , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Prospective Studies , Prosthesis Design , Swine , Treatment Outcome
20.
Cardiovasc Res ; 68(3): 493-501, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16099441

ABSTRACT

OBJECTIVE: To determine the influence of IL-1 on the arterial response to experimental injury in porcine models of percutaneous coronary intervention (PCI). METHODS: An intravenous (i.v.) bolus of 0.5 mg/kg followed by a subcutaneous (s.c.) infusion of 2 mg/kg/24 h of human IL-1 receptor antagonist (IL-1ra) inhibited neutrophil recruitment in response to intradermal IL-1. Using this dose regimen, five groups of pigs were studied: Group 1, oversized balloon angioplasty of 2 coronary vessels (14-day infusion, 28th day sacrifice and analysis); Groups 2, 3, 4, and 5, oversized stenting of 2 coronary vessels (Group 2: 14-day infusion, 28th day analysis; Group 3: 14-day infusion, 14th day analysis; Group 4: 28-day infusion, 28th day analysis; Group 5: 28-day infusion, 90th day analysis). Neointimal area was quantified by standard means. RESULTS: In Group 1, IL-1ra resulted in a 23% decrease in neointimal area (p=0.04); in Group 2, a 34% increase (p=0.001); in Group 3, a 38% decrease (p<0.0001); in Group 4, a 34% decrease (p=0.0004); and in Group 5, a 41% decrease (p=0.00001). CONCLUSIONS: IL-1ra was associated with a sustained, significant reduction in neointima after vessel wall injury as long as it is given for the duration of the stimulation of the IL-1 system, in this case at least 28 days. This suggests that therapies based on the antagonism of IL-1 may modulate the coronary artery response to injury.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Coronary Restenosis/prevention & control , Coronary Vessels/injuries , Coronary Vessels/metabolism , Sialoglycoproteins/therapeutic use , Animals , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , Coronary Stenosis/metabolism , Coronary Stenosis/therapy , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/metabolism , Male , Models, Animal , Random Allocation , Sialoglycoproteins/metabolism , Swine , Time Factors , Tunica Intima/drug effects , Tunica Intima/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...