Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
2.
Cardiovasc Res ; 114(3): 468-475, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29186373

ABSTRACT

AIMS: Circulating monocytes infiltrate the plaque and differentiate into macrophages, contributing to an inflammatory environment which is associated with higher risk of cardiovascular events. Although the pivotal role of circulating monocytes in plaque inflammation has been firmly established, the search continues to identify specific monocyte subsets that may be especially atherogenic. Therefore, we evaluated the relation between monocyte phenotype, particularly surface receptor expression, and arterial wall inflammation in patients at increased cardiovascular risk. METHODS AND RESULTS: We performed a multivariate linear regression analysis in 79 patients at increased cardiovascular risk who had both an 18F-fluorodeoxyglucose positron emission tomography/computed tomography to assess arterial wall inflammation and extensive monocyte characterization (using flow cytometry). We found that CCR2, a monocyte chemokine receptor essential for transmigration, significantly correlates with arterial wall inflammation. This relationship was independent of traditional cardiovascular risk factors and statin use (ß = 0.429, P = 0.015). We found no relation between arterial wall inflammation and monocyte count or monocyte subsets, namely CD14+CD16-, CD14+CD16+, CD14+CD16 ++, CCR5+, CD18+, CD11b+, or CD11c+ monocytes. CONCLUSION: Monocyte CCR2 expression is associated with arterial wall inflammation in patients at increased cardiovascular risk. Our data warrant further studies to assess if inhibition of CCR2 may attenuate atherosclerotic plaque inflammation.


Subject(s)
Arteries/diagnostic imaging , Arteritis/diagnostic imaging , Atherosclerosis/diagnostic imaging , Fluorodeoxyglucose F18/administration & dosage , Monocytes/metabolism , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/administration & dosage , Receptors, CCR2/metabolism , Aged , Arteries/metabolism , Arteries/pathology , Arteritis/metabolism , Arteritis/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers/metabolism , Female , Humans , Male , Middle Aged , Phenotype , Plaque, Atherosclerotic , Predictive Value of Tests
3.
Eur Heart J ; 38(20): 1584-1593, 2017 May 21.
Article in English | MEDLINE | ID: mdl-28329114

ABSTRACT

AIMS: Migration of monocytes into the arterial wall contributes to arterial inflammation and atherosclerosis progression. Since elevated low-density lipoprotein cholesterol (LDL-C) levels have been associated with activation of plasma monocytes, intensive LDL-C lowering may reverse these pro-inflammatory changes. Using proprotein convertase subtilisin/kexin type 9 (PCSK9) monoclonal antibodies (mAbs) which selectively reduce LDL-C, we studied the impact of LDL-C lowering on monocyte phenotype and function in patients with familial hypercholesterolaemia (FH) not using statins due to statin-associated muscle symptoms. METHODS AND RESULTS: We assessed monocyte phenotype and function using flow cytometry and a trans-endothelial migration assay in FH patients (n = 22: LDL 6.8 ± 1.9 mmol/L) and healthy controls (n = 18, LDL 2.9 ± 0.8 mmol/L). Monocyte chemokine receptor (CCR) 2 expression was approximaterly three-fold higher in FH patients compared with controls. C-C chemokine receptor type 2 (CCR2) expression correlated significantly with plasma LDL-C levels (r = 0.709) and was positively associated with intracellular lipid accumulation. Monocytes from FH patients also displayed enhanced migratory capacity ex vivo. After 24 weeks of PCSK9 mAb treatment (n = 17), plasma LDL-C was reduced by 49%, which coincided with reduced intracellular lipid accumulation and reduced CCR2 expression. Functional relevance was substantiated by the reversal of enhanced migratory capacity of monocytes following PCSK9 mAb therapy. CONCLUSIONS: Monocytes of FH patients have a pro-inflammatory phenotype, which is dampened by LDL-C lowering by PCSK9 mAb therapy. LDL-C lowering was paralleled by reduced intracellular lipid accumulation, suggesting that LDL-C lowering itself is associated with anti-inflammatory effects on circulating monocytes.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Hyperlipoproteinemia Type II/drug therapy , Monocytes/immunology , Proprotein Convertase 9/immunology , Analysis of Variance , Antibodies, Monoclonal, Humanized , Case-Control Studies , Cholesterol, LDL/metabolism , Drug Administration Schedule , Female , Humans , Hyperlipoproteinemia Type II/immunology , Interleukin-10/biosynthesis , Lipid Metabolism/drug effects , Male , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Receptors, CCR2/drug effects , Receptors, CCR2/metabolism , Tumor Necrosis Factors/metabolism
4.
Eur Heart J ; 38(6): 425-432, 2017 02 07.
Article in English | MEDLINE | ID: mdl-27357356

ABSTRACT

Aims: Experimental work posits that acute ischaemic events trigger haematopoietic activity, driving monocytosis, and atherogenesis. Considering the chronic low-grade inflammatory state in atherosclerosis, we hypothesized that haematopoietic hyperactivity is a persistent feature in cardiovascular disease (CVD). Therefore, we aimed to assess the activity of haematopoietic organs and haematopoietic stem and progenitor cells (HSPCs) in humans. Methods and results: First, we performed 18F-fluorodeoxyglucose positron emission tomographic (18F-FDG PET) imaging in 26 patients with stable atherosclerotic CVD (ischaemic event >12 months ago), and 25 matched controls. In splenic tissue, 18F-FDG uptake was 2.68 ± 0.65 in CVD patients vs. 1.75 ± 0.54 in controls (1.6-fold higher; P< 0.001), and in bone marrow 3.20 ± 0.76 vs. 2.72 ± 0.46 (1.2-fold higher; P = 0.003), closely related to LDL cholesterol levels (LDLc, r = 0.72). Subsequently, we determined progenitor potential of HSPCs harvested from 18 patients with known atherosclerotic CVD and 30 matched controls; both groups were selected from a cohort of cancer patients undergoing autologous stem cell transplantation. In CVD patients, the normalized progenitor potential, expressed as the number of colony-forming units-granulocyte/monocyte (CFU-GM) colonies/CD34+ cell, was 1.6-fold higher compared with matched controls (P < 0.001). Finally, we assessed the effects of native and oxidized lipoproteins on HSPCs harvested from healthy donors in vitro. Haematopoietic stem and progenitor cells displayed a 1.5-fold increased CFU-GM capacity in co-culture with oxidized LDL in vitro (P = 0.002), which was inhibited by blocking oxidized phospholipids via E06 (P = 0.001). Conclusion: Collectively, these findings strengthen the case for a chronically affected haematopoietic system, potentially driving the low-grade inflammatory state in patients with atherosclerosis.


Subject(s)
Coronary Artery Disease/pathology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Vascular Calcification/pathology , Bone Marrow/physiology , Case-Control Studies , Cells, Cultured , Cholesterol, LDL/physiology , Female , Fluorodeoxyglucose F18 , Hematopoiesis, Extramedullary/physiology , Humans , Male , Middle Aged , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Risk Factors
5.
J Am Soc Nephrol ; 28(4): 1278-1285, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27799487

ABSTRACT

CKD associates with a 1.5- to 3.5-fold increased risk for cardiovascular disease. Both diseases are characterized by increased inflammation, and in patients with CKD, elevated C-reactive protein level predicts cardiovascular risk. In addition to systemic inflammation, local arterial inflammation, driven by monocyte-derived macrophages, predicts future cardiovascular events in the general population. We hypothesized that subjects with CKD have increased arterial and cellular inflammation, reflected by 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography computed tomography (PET/CT) of the arterial wall and a migratory phenotype of monocytes. We assessed 18F-FDG uptake in the arterial wall in 14 patients with CKD (mean±SD age: 59±5 years, mean±SD eGFR: 37±12 ml/min per 1.73 m2) but without cardiovascular diseases, diabetes, or inflammatory conditions and in 14 control subjects (mean age: 60±11 years, mean eGFR: 86±16 ml/min per 1.73 m2). Compared with controls, patients with CKD showed increased arterial inflammation, quantified as target-to-background ratio (TBR) in the aorta (TBRmax: CKD, 3.14±0.70 versus control, 2.12±0.27; P=0.001) and the carotid arteries (TBRmax: CKD, 2.45±0.65 versus control, 1.66±0.27; P<0.001). Characterization of circulating monocytes using flow cytometry revealed increased chemokine receptor expression and enhanced transendothelial migration capacity in patients with CKD compared with controls. In conclusion, this increased arterial wall inflammation, observed in patients with CKD but without overt atherosclerotic disease and with few traditional risk factors, may contribute to the increased cardiovascular risk associated with CKD. The concomitant elevation of monocyte activity may provide novel therapeutic targets for attenuating this inflammation and thereby preventing CKD-associated cardiovascular disease.


Subject(s)
Arteritis/diagnostic imaging , Arteritis/etiology , Cardiovascular Diseases/diagnostic imaging , Cardiovascular Diseases/etiology , Fluorodeoxyglucose F18 , Inflammation/diagnostic imaging , Inflammation/etiology , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Renal Insufficiency, Chronic/complications , Cells , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Risk Factors
7.
BMC Cardiovasc Disord ; 16(1): 237, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27887576

ABSTRACT

BACKGROUND: Patients with peripheral artery disease (PAD) are at increased risk of secondary events, which is exaggerated in the presence of type 2 diabetes mellitus. Diabetes is associated with a systemic pro-inflammatory state. We therefore investigated the cumulative impact of PAD and type 2 diabetes on carotid arterial wall inflammation. As recent data suggest a detrimental role of exogenous insulin on cardiovascular disease, we also included a group of insulin users. RESULTS: 18F-fluorodeoxyglucose positron emission tomography with computed tomography (18F-FDG PET/CT) imaging showed increased carotid arterial wall inflammation, assessed as target-to-background ratio (TBR), in PAD patients without diabetes (PAD-only: n = 11, 1.97 ± 0.57) compared with matched controls (n = 12, 1.49 ± 0.57; p = 0.009), with a significant further TBR increase in PAD patients with type 2 diabetes (PAD-DM, n = 23, 2.90 ± 1, p = 0.033 vs PAD-only). TBR of insulin users (n = 12, 3.31 ± 1.14) was higher compared with patients on oral medication only (n = 11, 2.44 ± 0.76, p = 0.035), despite comparable PAD severity (Fontaine stages), BMI and CRP. Multivariate regression analysis showed that Hba1c and plasma insulin levels, but not dose of exogenous insulin, correlated with TBR. CONCLUSIONS: Concurrent diabetes significantly augments carotid arterial wall inflammation in PAD patients. A further increase in those requiring insulin was observed, which was associated with diabetes severity, rather than with the use of exogenous insulin itself.


Subject(s)
Carotid Arteries/diagnostic imaging , Diabetes Mellitus, Type 2/complications , Inflammation/etiology , Peripheral Arterial Disease/complications , Ankle Brachial Index , Cross-Sectional Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Female , Follow-Up Studies , Glycated Hemoglobin/metabolism , Humans , Inflammation/diagnosis , Insulin/blood , Male , Middle Aged , Peripheral Arterial Disease/diagnosis , Positron Emission Tomography Computed Tomography , Predictive Value of Tests , Retrospective Studies
8.
JACC Cardiovasc Imaging ; 9(10): 1198-1207, 2016 10.
Article in English | MEDLINE | ID: mdl-27639759

ABSTRACT

OBJECTIVES: This study assessed 5 frequently applied arterial 18fluorodeoxyglucose (18F-FDG) uptake metrics in healthy control subjects, those with risk factors and patients with cardiovascular disease (CVD), to derive uptake thresholds in each subject group. Additionally, we tested the reproducibility of these measures and produced recommended sample sizes for interventional drug studies. BACKGROUND: 18F-FDG positron emission tomography (PET) can identify plaque inflammation as a surrogate endpoint for vascular interventional drug trials. However, an overview of 18F-FDG uptake metrics, threshold values, and reproducibility in healthy compared with diseased subjects is not available. METHODS: 18F-FDG PET/CT of the carotid arteries and ascending aorta was performed in 83 subjects (61 ± 8 years) comprising 3 groups: 25 healthy controls, 23 patients at increased CVD risk, and 35 patients with known CVD. We quantified 18F-FDG uptake across the whole artery, the most-diseased segment, and within all active segments over several pre-defined cutoffs. We report these data with and without background corrections. Finally, we determined measurement reproducibility and recommended sample sizes for future drug studies based on these results. RESULTS: All 18F-FDG uptake metrics were significantly different between healthy and diseased subjects for both the carotids and aorta. Thresholds of physiological 18F-FDG uptake were derived from healthy controls using the 90th percentile of their target to background ratio (TBR) value (TBRmax); whole artery TBRmax is 1.84 for the carotids and 2.68 in the aorta. These were exceeded by >52% of risk factor patients and >67% of CVD patients. Reproducibility was excellent in all study groups (intraclass correlation coefficient >0.95). Using carotid TBRmax as a primary endpoint resulted in sample size estimates approximately 20% lower than aorta. CONCLUSIONS: We report thresholds for physiological 18F-FDG uptake in the arterial wall in healthy subjects, which are exceeded by the majority of CVD patients. This remains true, independent of readout vessel, signal quantification method, or the use of background correction. We also confirm the high reproducibility of 18F-FDG PET measures of inflammation. Nevertheless, because of overlap between subject categories and the relatively small population studied, these data have limited generalizability until substantiated in larger, prospective event-driven studies. (Vascular Inflammation in Patients at Risk for Atherosclerotic Disease; NTR5006).


Subject(s)
Aorta/diagnostic imaging , Aortitis/diagnostic imaging , Arteritis/diagnostic imaging , Carotid Arteries/diagnostic imaging , Carotid Artery Diseases/diagnostic imaging , Fluorodeoxyglucose F18/administration & dosage , Plaque, Atherosclerotic , Positron-Emission Tomography , Radiopharmaceuticals/administration & dosage , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Observer Variation , Predictive Value of Tests , Reproducibility of Results
9.
Circulation ; 134(8): 611-24, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27496857

ABSTRACT

BACKGROUND: Elevated lipoprotein(a) [Lp(a)] is a prevalent, independent cardiovascular risk factor, but the underlying mechanisms responsible for its pathogenicity are poorly defined. Because Lp(a) is the prominent carrier of proinflammatory oxidized phospholipids (OxPLs), part of its atherothrombosis might be mediated through this pathway. METHODS: In vivo imaging techniques including magnetic resonance imaging, (18)F-fluorodeoxyglucose uptake positron emission tomography/computed tomography and single-photon emission computed tomography/computed tomography were used to measure subsequently atherosclerotic burden, arterial wall inflammation, and monocyte trafficking to the arterial wall. Ex vivo analysis of monocytes was performed with fluorescence-activated cell sorter analysis, inflammatory stimulation assays, and transendothelial migration assays. In vitro studies of the pathophysiology of Lp(a) on monocytes were performed with an in vitro model for trained immunity. RESULTS: We show that subjects with elevated Lp(a) (108 mg/dL [50-195 mg/dL]; n=30) have increased arterial inflammation and enhanced peripheral blood mononuclear cells trafficking to the arterial wall compared with subjects with normal Lp(a) (7 mg/dL [2-28 mg/dL]; n=30). In addition, monocytes isolated from subjects with elevated Lp(a) remain in a long-lasting primed state, as evidenced by an increased capacity to transmigrate and produce proinflammatory cytokines on stimulation (n=15). In vitro studies show that Lp(a) contains OxPL and augments the proinflammatory response in monocytes derived from healthy control subjects (n=6). This effect was markedly attenuated by inactivating OxPL on Lp(a) or removing OxPL on apolipoprotein(a). CONCLUSIONS: These findings demonstrate that Lp(a) induces monocyte trafficking to the arterial wall and mediates proinflammatory responses through its OxPL content. These findings provide a novel mechanism by which Lp(a) mediates cardiovascular disease. CLINICAL TRIAL REGISTRATION: URL: http://www.trialregister.nl. Unique identifier: NTR5006 (VIPER Study).


Subject(s)
Aorta/diagnostic imaging , Aorta/metabolism , Inflammation Mediators/metabolism , Leukocytes, Mononuclear/metabolism , Lipoprotein(a)/metabolism , Phospholipids/metabolism , Adult , Aged , Cell Movement/physiology , Cells, Cultured , Female , Humans , Inflammation/diagnostic imaging , Inflammation/metabolism , Male , Middle Aged , Positron Emission Tomography Computed Tomography
10.
Atherosclerosis ; 251: 381-388, 2016 08.
Article in English | MEDLINE | ID: mdl-27263077

ABSTRACT

BACKGROUND AND AIMS: Infusion of high-density lipoprotein (HDL) mimetics aimed at reducing atherosclerotic burden has led to equivocal results, which may relate in part to the inability of HDL mimetics to adequately reach atherosclerotic lesions in humans. This study evaluated delivery of recombinant human apolipoprotein A-I (apoA-I) containing HDL mimetic CER-001 in carotid plaques in patients. METHODS: CER-001 was radiolabeled with the long-lived positron emitter zirconium-89 ((89)Zr) to enable positron emission tomography with computed tomography (PET/CT) imaging. Eight patients with atherosclerotic carotid artery disease (>50% stenosis) received a single infusion of unlabeled CER-001 (3 mg/kg), co-administered with 10 mg of (89)Zr-labeled CER-001 (18 MBq). Serial PET/CT imaging and contrast enhanced-magnetic resonance imaging (CE-MRI) were performed to evaluate targeted delivery of CER-001. RESULTS: One hour after infusion, mean plasma apoA-I levels increased by 9.9 mg/dL (p = 0.026), with a concomitant relative increase in the plasma cholesterol efflux capacity of 13.8% (p < 0.001). Using serial PET/CT imaging, we showed that arterial uptake of CER-001 expressed as target-to-background ratio (TBRmax) increased significantly 24 h after infusion, and remained increased up to 48 h (TBRmax t = 10 min: 0.98; t = 24 h: 1.14 (p = 0.001); t = 48 h: 1.12 (p = 0.007)). TBRmax was higher in plaque compared with non-plaque segments (1.18 vs. 1.05; p < 0.001). Plaque TBRmax correlated with local plaque contrast enhancement (r = 0.56; p = 0.019) as assessed by CE-MRI. CONCLUSIONS: Infusion of HDL mimetic CER-001 increases plasma apoA-I concentration and plasma cholesterol efflux capacity. Our data support the concept that CER-001 targets plaque regions in patients, which correlates with plaque contrast enhancement. These clinical findings may also guide future nanomedicine development using HDL particles for drug delivery in atherosclerosis. CLINICAL TRIAL REGISTRATION: Netherlands Trial Registry - NTR5178. http://www.trialregister.nl/trialreg/admin/rctview.asp?TC=5178.


Subject(s)
Apolipoprotein A-I/chemistry , Phospholipids/chemistry , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/drug therapy , Recombinant Proteins/chemistry , Aged , Contrast Media/chemistry , Drug Carriers , Female , Humans , Lipoproteins/chemistry , Magnetic Resonance Imaging , Male , Middle Aged , Nanomedicine , Plaque, Atherosclerotic/metabolism , Positron-Emission Tomography , Tomography, X-Ray Computed , Zirconium/chemistry
11.
Arthritis Res Ther ; 18(1): 115, 2016 05 21.
Article in English | MEDLINE | ID: mdl-27209093

ABSTRACT

BACKGROUND: Increasing numbers of patients (up to 40 %) with rheumatoid arthritis (RA) achieve remission, yet it remains to be elucidated whether this also normalizes their cardiovascular risk. Short-term treatment with TNF inhibitors lowers arterial wall inflammation, but not to levels of healthy controls. We investigated whether RA patients in long-term remission are characterized by normalized inflammatory activity of the arterial wall and if this is dependent on type of medication used (TNF-inhibitor versus nonbiological disease-modifying antirheumatic drugs (DMARDs)). METHODS: Arterial wall inflammation, bone marrow and splenic activity (index of progenitor cell activity) was assessed with (18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography/computed tomography (PET/CT) in RA patients in remission (disease activity score (DAS28) <2.6 for >6 months) and healthy controls. We performed ex vivo characterization of monocytes using flow cytometry and a transendothelial migration assay. RESULTS: Overall, arterial wall inflammation was comparable in RA patients (n = 23) in long-term remission and controls (n = 17). However, RA subjects using current anti-TNF therapy (n = 13, disease activity score 1.98[1.8-2.2]) have an almost 1.2-fold higher (18)F-FDG uptake in the arterial wall compared to those using DMARDs (but with previous anti-TNF therapy) (n = 10, disease activity score 2.24[1.3-2.5]), which seemed to be predominantly explained by longer duration of their rheumatic disease in a multivariate linear regression analysis. This coincided with increased expression of pro-adhesive (CCR2) and migratory (CD11c, CD18) surface markers on monocytes and a concomitant increased migratory capacity. Finally, we found increased activity in bone marrow and spleen in RA patients using anti-TNF therapy compared to those with DMARDs and controls. CONCLUSIONS: A subset of patients with RA in clinical remission have activated monocytes and increased inflammation in the arterial wall, despite the use of potent TNF blocking therapies. In these subjects, RA disease duration was the most important contributor to the level of arterial wall inflammation. This increased inflammatory state implies higher cardiovascular risk in these patients, who thus may require more stringent CV risk management.


Subject(s)
Aorta/pathology , Arthritis, Rheumatoid/pathology , Inflammation/pathology , Aged , Antirheumatic Agents/therapeutic use , Aorta/diagnostic imaging , Arthritis, Rheumatoid/drug therapy , Cohort Studies , Cross-Sectional Studies , Female , Flow Cytometry , Humans , Male , Middle Aged , Positron Emission Tomography Computed Tomography , Remission Induction
12.
Vascul Pharmacol ; 82: 51-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27189780

ABSTRACT

BACKGROUND: Macrophages play a central role in atherosclerosis development and progression, hence, targeting macrophage activity is considered an attractive therapeutic. Recently, we documented nanomedicinal delivery of the anti-inflammatory compound prednisolone to atherosclerotic plaque macrophages in patients, which did however not translate into therapeutic efficacy. This unanticipated finding calls for in-depth screening of drugs intended for targeting plaque macrophages. METHODS AND RESULTS: We evaluated the effect of several candidate drugs on macrophage activity, rating overall performance with respect to changes in cytokine release, oxidative stress, lipid handling, endoplasmic reticulum (ER) stress, and proliferation of macrophages. Using this in vitro approach, we observed that the anti-inflammatory effect of prednisolone was counterbalanced by multiple adverse effects on other key pathways. Conversely, pterostilbene, T0901317 and simvastatin had an overall anti-atherogenic effect on multiple pathways, suggesting their potential for liposomal delivery. CONCLUSION: This dedicated assay setup provides a framework for high-throughput assessment. Further in vivo studies are warranted to determine the predictive value of this macrophage-based screening approach and its potential value in nanomedicinal drug development for cardiovascular patients.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atherosclerosis/drug therapy , Inflammation/drug therapy , Macrophage Activation/drug effects , Macrophages/drug effects , Plaque, Atherosclerotic , Signal Transduction/drug effects , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Proliferation/drug effects , Cytokines/metabolism , Endoplasmic Reticulum Stress/drug effects , High-Throughput Screening Assays , Humans , Hydrocarbons, Fluorinated/pharmacology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Lipid Metabolism/drug effects , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress/drug effects , Prednisolone/pharmacology , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Simvastatin/pharmacology , Stilbenes/pharmacology , Sulfonamides/pharmacology , Transfection
13.
Ann Rheum Dis ; 75(10): 1848-51, 2016 10.
Article in English | MEDLINE | ID: mdl-27084958

ABSTRACT

BACKGROUND: Ankylosing spondylitis (AS) is a chronic inflammatory disease with involvement of axial and sacroiliac joints. In addition, patients with AS have increased risk of cardiovascular disease (CVD), which might be attributed to enhanced inflammatory activity of the arterial wall. In the present study, we compared the level of carotid arterial wall inflammation in patients with AS with healthy controls using (18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography with CT. As arterial wall inflammation is reduced by statin therapy, we subsequently assessed the effect of 3-month statin therapy on arterial wall inflammation in AS. METHODS AND RESULTS: We included 24 patients with AS (age 44±10, 72% males) without a history of CVD and 20 controls matched for age and gender. Patients with AS had lower high-density lipoprotein cholesterol and increased C reactive protein (CRP) compared with controls. The 10-year CVD risk was 2% in both groups. Notwithstanding, patients with AS had a 20% increase in arterial wall (18)F-FDG uptake compared with controls. Three-month atorvastatin 40 mg daily significantly lowered low-density lipoprotein cholesterol (baseline 3.55±1.15 mmol/L, -53%) and CRP (baseline 5.0 (1.5-9.3) mg/L, -58%) with a concomitant decrease of carotid arterial wall inflammation (maximum target-to-background ratio from 1.90±0.30 to 1.67±0.27; p=0.009). CONCLUSIONS: Patients with AS and without other CVD risk factors have increased arterial wall inflammation, which decreases upon statin therapy. These subjects are not identified as being at risk in current cardiovascular prevention guidelines. Our data support the need to revise CV disease management in AS, with perhaps a role for early statin therapy.


Subject(s)
Atorvastatin/administration & dosage , Carotid Arteries/drug effects , Carotid Artery Diseases/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Spondylitis, Ankylosing/complications , Adult , C-Reactive Protein/analysis , Cardiovascular Diseases/etiology , Carotid Arteries/pathology , Carotid Artery Diseases/etiology , Carotid Artery Diseases/pathology , Case-Control Studies , Cholesterol, LDL/blood , Female , Humans , Lipoproteins, HDL/blood , Male , Middle Aged , Risk Factors , Spondylitis, Ankylosing/blood , Spondylitis, Ankylosing/pathology , Treatment Outcome
14.
Nanomedicine ; 12(6): 1463-70, 2016 08.
Article in English | MEDLINE | ID: mdl-27015770

ABSTRACT

Atherosclerosis is a lipid-driven inflammatory disease, for which nanomedicinal interventions are under evaluation. Previously, we showed that liposomal nanoparticles loaded with prednisolone (LN-PLP) accumulated in plaque macrophages, however, induced proatherogenic effects in patients. Here, we confirmed in low-density lipoprotein receptor knockout (LDLr(-/-)) mice that LN-PLP accumulates in plaque macrophages. Next, we found that LN-PLP infusions at 10mg/kg for 2weeks enhanced monocyte recruitment to plaques. In follow up, after 6weeks of LN-PLP exposure we observed (i) increased macrophage content, (ii) more advanced plaque stages, and (iii) larger necrotic core sizes. Finally, in vitro studies showed that macrophages become lipotoxic after LN-PLP exposure, exemplified by enhanced lipid loading, ER stress and apoptosis. These findings indicate that liposomal prednisolone may paradoxically accelerate atherosclerosis by promoting macrophage lipotoxicity. Hence, future (nanomedicinal) drug development studies are challenged by the multifactorial nature of atherosclerotic inflammation.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Prednisolone/administration & dosage , Animals , Humans , Liposomes , Macrophages/pathology , Mice , Plaque, Atherosclerotic
15.
J Lipid Res ; 57(9): 1612-8, 2016 09.
Article in English | MEDLINE | ID: mdl-26637277

ABSTRACT

Lipoprotein (a) [Lp(a)] is a human plasma lipoprotein with unique structural and functional characteristics. Lp(a) is an assembly of two components: a central core with apoB and an additional glycoprotein, called apo(a). Ever since the strong association between elevated levels of Lp(a) and an increased risk for CVD was recognized, interest in the therapeutic modulation of Lp(a) levels has increased. Here, the past and present therapies aiming to lower Lp(a) levels will be reviewed, demonstrating that these agents have had varying degrees of success. The next challenge will be to prove that Lp(a) lowering also leads to cardiovascular benefit in patients with elevated Lp(a) levels. Therefore, highly specific and potent Lp(a)-lowering strategies are awaited urgently.


Subject(s)
Apolipoprotein B-100/blood , Apolipoproteins A/blood , Cardiovascular Diseases/drug therapy , Lipoprotein(a)/blood , Cardiovascular Diseases/blood , Humans , Hypolipidemic Agents/therapeutic use , Lipoprotein(a)/drug effects , Risk Factors
16.
Curr Opin Lipidol ; 26(6): 580-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26382552

ABSTRACT

PURPOSE OF REVIEW: Atherosclerosis is a chronic disease of the arterial wall largely driven by inflammation; hence, therapeutics targeting inflammatory pathways are considered an attractive strategy in atherosclerotic cardiovascular disease (ASCVD). The purpose of this review is to describe the randomized, placebo-controlled clinical trials currently investigating the impact of anti-inflammatory strategies in ASCVD patients, to discuss novel insights and targets into the role of innate immunity in atherosclerosis and to address the promise of local drug delivery as opposed to systemic therapies in atherosclerotic disease. RECENT FINDINGS: The first clinical trials using systemic anti-inflammatory drugs in ASCVD patients might be able to strengthen the case for immunomodulation once showing an improved ASCVD outcome. Several specific targets in innate immunity bear therapeutic potential, of which some have already entered the clinical arena. To prevent immunosuppression by systemic effects, drug delivery systems are increasingly being applied to locally attenuate plaque inflammation. SUMMARY: Anti-inflammatory therapies seem promising for future treatment of ASCVD. In view of the risk of immunosuppression in case of long term and systemic use of anti-inflammatory drugs, there is a clinical need for highly selective and targeted therapies in patients with atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Molecular Targeted Therapy/methods , Animals , Atherosclerosis/immunology , Clinical Trials as Topic , Drug Delivery Systems , Humans , Immunity, Innate/drug effects , Inflammation/drug therapy
18.
Arterioscler Thromb Vasc Biol ; 35(7): 1663-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26109739

ABSTRACT

OBJECTIVE: We previously demonstrated that subjects with functional ATP-binding cassette (ABC) A1 mutations have increased atherosclerosis, which has been attributed to the role of ABCA1 in reverse cholesterol transport. More recently, a proinflammatory effect of Abca1 deficiency was shown in mice, potentially contributing to atherogenesis. In this study, we investigated whether ABCA1 deficiency was associated with proinflammatory changes in humans. APPROACH AND RESULTS: Thirty-one heterozygous, 5 homozygous ABCA1 mutation carriers, and 21 matched controls were studied. (18)Fluorodeoxyglucose positron emission tomography with computed tomographic scanning was performed in a subset of carriers and controls to assess arterial wall inflammation (target:background ratio). Heterozygous ABCA1 mutation carriers had a 20% higher target:background ratio than in controls (target:background ratio; P=0.008). In carriers using statins (n=7), target:background ratio was 21% reduced than in nonstatin users (n=7; P=0.03). We then measured plasma cytokine levels. Tumor necrosis factor α, monocyte chemoattractant protein-1, and interleukin-6 levels were increased in heterozygous and homozygous ABCA1 mutation carriers. We isolated monocytes from carriers and controls and measured inflammatory gene expression. Only TNFα mRNA was increased in monocytes from heterozygous ABCA1 mutation carriers. Additional studies in THP-1 macrophages showed that both ABCA1 deficiency and lipoprotein-deficient plasma from ABCA1 mutation carriers increased inflammatory gene expression. CONCLUSIONS: Our data suggest a proinflammatory state in ABCA1 mutation carriers as reflected by an increased positron emission tomography-MRI signal in nonstatin using subjects, and increased circulating cytokines. The increased inflammation in ABCA1 mutation carriers seems to be attenuated by statins.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Inflammation/genetics , Mutation , Plaque, Atherosclerotic/genetics , Apolipoproteins B/blood , Biological Transport , Cholesterol/blood , Cytokines/blood , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/metabolism , Inflammation/prevention & control , Lipoproteins/blood , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/prevention & control
20.
Nanomedicine ; 11(5): 1133-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25791805

ABSTRACT

The present study describes the development of a good manufacturing practice (GMP)-grade liposomal nanotherapy containing prednisolone phosphate for the treatment of inflammatory diseases. After formulation design, GMP production was commenced which yielded consistent, stable liposomes sized 100nm±10nm, with a prednisolone phosphate (PLP) incorporation efficiency of 3%-5%. Pharmacokinetics and toxicokinetics of GMP-grade liposomal nanoparticles were evaluated in healthy rats, which were compared to daily and weekly administration of free prednisolone phosphate, revealing a long circulatory half-life with minimal side effects. Subsequently, non-invasive multimodal clinical imaging after liposomal nanotherapy's intravenous administration revealed anti-inflammatory effects on the vessel wall of atherosclerotic rabbits. The present program led to institutional review board approval for two clinical trials with patients with atherosclerosis. FROM THE CLINICAL EDITOR: In drug discovery, bringing production to industrial scale is an essential process. In this article the authors describe the development of an anti-inflammatory nanoparticle according to good manufacturing practice. As a result, this paves the way for translating laboratory studies to clinical trials in humans.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Atherosclerosis/drug therapy , Chemistry, Pharmaceutical/methods , Glucocorticoids/administration & dosage , Prednisolone/analogs & derivatives , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/toxicity , Aorta/drug effects , Aorta/pathology , Atherosclerosis/pathology , Glucocorticoids/pharmacokinetics , Glucocorticoids/therapeutic use , Glucocorticoids/toxicity , Half-Life , Humans , Liposomes , Male , Prednisolone/administration & dosage , Prednisolone/pharmacokinetics , Prednisolone/therapeutic use , Prednisolone/toxicity , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...