Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
PLoS Pathog ; 18(9): e1010738, 2022 09.
Article in English | MEDLINE | ID: mdl-36067220

ABSTRACT

While Epstein-Barr virus causes mostly asymptomatic infection, associated malignancies, and autoimmune and lymphoproliferative diseases occur. To dissect the evolution of humoral immune responses over the course of EBV infection and to gain a better understanding of the potential contribution of antibody (Ab) function to viral control, we comprehensively profiled Ab specificities and Fc-functionalities using systems serology and VirScan. Ab functions against latent (EBNA1), early (p47/54) and two late (gp350/220 and VCA-p18) EBV proteins were overall modest and/or short-lived, differing from humoral responses induced during acute infection by other viruses such as HIV. In the first year post infection, only p18 elicited robust IgM-driven complement deposition and IgG-driven neutrophil phagocytosis while responses against EBNA-1 were largely Fc-functionally silent and only matured during chronic infection to drive phagocytosis. In contrast, Abs against Influenza virus readily mediated broad Fc-activity in all participants. These data suggest that EBV evades the induction of robust Fc-functional Abs, potentially due to the virus' life cycle, switching from lytic to latent stages during infection.


Subject(s)
Epstein-Barr Virus Infections , Antibodies, Viral , Epstein-Barr Virus Nuclear Antigens , Herpesvirus 4, Human , Humans , Immunoglobulin G , Immunoglobulin M
2.
Biomacromolecules ; 19(4): 1234-1244, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29539266

ABSTRACT

Porcine glutaraldehyde-fixed pericardium is widely used to replace human heart valves. Despite the stabilizing effects of glutaraldehyde fixation, the lack of endothelialization and the occurrence of immune reactions contribute to calcification and structural valve deterioration, which is particularly significant in young patients, in whom valve longevity is crucial. This report shows an optimization system with which to enhance endothelialization of fixed pericardium to mimic the biological function of a native heart valve. The glutaraldehyde detoxification, together with the application of a biodegradable methacrylated chondroitin sulfate hydrogel, reduces aldehydes cytotoxicity, increases the migration and proliferation of endothelial cells and the recruitment of endothelial cell progenitors, and confers thromboresistance in fixed pericardium. The combination of glutaraldehyde detoxification and a coating with chondroitin sulfate hydrogel promotes in situ mechanisms of endothelialization in fixed pericardium. We offer a new solution for improving the long life of bioprosthetic valves and exploring the means of making valves suitable to endothelialization.


Subject(s)
Chondroitin Sulfates/chemistry , Heart Valves/drug effects , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Pericardium/drug effects , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Chondroitin Sulfates/pharmacology , Clinical Deterioration , Disease Models, Animal , Endothelial Cells/drug effects , Glutaral/chemistry , Heart Valves/physiopathology , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Pericardium/physiopathology , Swine
3.
J Biomech ; 71: 296-301, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29452756

ABSTRACT

Stent thrombosis is a major complication of coronary stent and scaffold intervention. While often unanticipated and lethal, its incidence is low making mechanistic examination difficult through clinical investigation alone. Thus, throughout the technological advancement of these devices, experimental models have been indispensable in furthering our understanding of device safety and efficacy. As we refine model systems to gain deeper insight into adverse events, it is equally important that we continue to refine our measurement methods. We used digital signal processing in an established flow loop model to investigate local flow effects due to geometric stent features and ultimately its relationship to thrombus formation. A new metric of clot distribution on each microCT slice termed normalized clot ratio was defined to quantify this distribution. Three under expanded coronary bare-metal stents were run in a flow loop model to induce clotting. Samples were then scanned in a MicroCT machine and digital signal processing methods applied to analyze geometric stent conformation and spatial clot formation. Results indicated that geometric stent features play a significant role in clotting patterns, specifically at a frequency of 0.6225 Hz corresponding to a geometric distance of 1.606 mm. The magnitude-squared coherence between geometric features and clot distribution was greater than 0.4 in all samples. In stents with poor wall apposition, ranging from 0.27 mm to 0.64 mm maximum malapposition (model of real-world heterogeneity), clots were found to have formed in between stent struts rather than directly adjacent to struts. This early work shows how the combination of tools in the areas of image processing and signal analysis can advance the resolution at which we are able to define thrombotic mechanisms in in vitro models, and ultimately, gain further insight into clinical performance.


Subject(s)
Stents/adverse effects , Thrombosis/etiology , Coronary Vessels/diagnostic imaging , Coronary Vessels/surgery , Humans , Thrombosis/diagnostic imaging , X-Ray Microtomography
4.
EuroIntervention ; 13(12): e1384-e1385, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29208577
5.
Sci Transl Med ; 9(417)2017 Nov 22.
Article in English | MEDLINE | ID: mdl-29167396

ABSTRACT

Chronic kidney disease (CKD/uremia) remains vexing because it increases the risk of atherothrombosis and is also associated with bleeding complications on standard antithrombotic/antiplatelet therapies. Although the associations of indolic uremic solutes and vascular wall proteins [such as tissue factor (TF) and aryl hydrocarbon receptor (AHR)] are being defined, the specific mechanisms that drive the thrombotic and bleeding risks are not fully understood. We now present an indolic solute-specific animal model, which focuses on solute-protein interactions and shows that indolic solutes mediate the hyperthrombotic phenotype across all CKD stages in an AHR- and TF-dependent manner. We further demonstrate that AHR regulates TF through STIP1 homology and U-box-containing protein 1 (STUB1). As a ubiquitin ligase, STUB1 dynamically interacts with and degrades TF through ubiquitination in the uremic milieu. TF regulation by STUB1 is supported in humans by an inverse relationship of STUB1 and TF expression and reduced STUB1-TF interaction in uremic vessels. Genetic or pharmacological manipulation of STUB1 in vascular smooth muscle cells inhibited thrombosis in flow loops. STUB1 perturbations reverted the uremic hyperthrombotic phenotype without prolonging the bleeding time, in contrast to heparin, the standard-of-care antithrombotic in CKD patients. Our work refines the thrombosis axis (STUB1 is a mediator of indolic solute-AHR-TF axis) and expands the understanding of the interconnected relationships driving the fragile thrombotic state in CKD. It also establishes a means of minimizing the uremic hyperthrombotic phenotype without altering the hemostatic balance, a long-sought-after combination in CKD patients.


Subject(s)
Hemorrhage/metabolism , Thrombosis/metabolism , Ubiquitin-Protein Ligases/metabolism , Uremia/metabolism , Analysis of Variance , Animals , Female , Hemorrhage/enzymology , Hemorrhage/pathology , Male , Mice , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Thrombosis/enzymology , Thrombosis/pathology , Ubiquitin-Protein Ligases/genetics , Uremia/enzymology , Uremia/pathology
6.
EuroIntervention ; 12(17): 2148-2156, 2017 Apr 20.
Article in English | MEDLINE | ID: mdl-27993749

ABSTRACT

AIMS: Simple surface modifications can enhance coronary stent performance. Ultra-hydrophilic surface (UHS) treatment of contemporary bare metal stents (BMS) was assessed in vivo to verify whether such stents can provide long-term efficacy comparable to second-generation drug-eluting stents (DES) while promoting healing comparably to BMS. METHODS AND RESULTS: UHS-treated BMS, untreated BMS and corresponding DES were tested for three commercial platforms. A thirty-day and a 90-day porcine coronary model were used to characterise late tissue response. Three-day porcine coronary and seven-day rabbit iliac models were used for early healing assessment. In porcine coronary arteries, hydrophilic treatment reduced intimal hyperplasia relative to the BMS and corresponding DES platforms (1.5-fold to threefold reduction in 30-day angiographic and histological stenosis; p<0.04). Endothelialisation was similar on UHS-treated BMS and untreated BMS, both in swine and rabbit models, and lower on DES. Elevation in thrombotic indices was infrequent (never observed with UHS, rare with BMS, most often with DES), but, when present, correlated with reduced endothelialisation (p<0.01). CONCLUSIONS: Ultra-hydrophilic surface treatment of contemporary stents conferred good healing while moderating neointimal and thrombotic responses. Such surfaces may offer safe alternatives to DES, particularly when rapid healing and short dual antiplatelet therapy (DAPT) are crucial.


Subject(s)
Hydrophobic and Hydrophilic Interactions , Percutaneous Coronary Intervention/instrumentation , Stents , Animals , Neointima/prevention & control , Rabbits , Swine , Thrombosis/prevention & control
7.
Brain Behav Immun ; 61: 36-49, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27825953

ABSTRACT

Neuropeptide hormone oxytocin has roles in social bonding, energy metabolism, and wound healing contributing to good physical, mental and social health. It was previously shown that feeding of a human commensal microbe Lactobacillus reuteri (L. reuteri) is sufficient to up-regulate endogenous oxytocin levels and improve wound healing capacity in mice. Here we show that oral L. reuteri-induced skin wound repair benefits extend to human subjects. Further, dietary supplementation with a sterile lysate of this microbe alone is sufficient to boost systemic oxytocin levels and improve wound repair capacity. Oxytocin-producing cells were found to be increased in the caudal paraventricular nucleus [PVN] of the hypothalamus after feeding of a sterile lysed preparation of L. reuteri, coincident with lowered blood levels of stress hormone corticosterone and more rapid epidermal closure, in mouse models. We conclude that microbe viability is not essential for regulating host oxytocin levels. The results suggest that a peptide or metabolite produced by bacteria may modulate host oxytocin secretion for potential public or personalized health goals.


Subject(s)
Limosilactobacillus reuteri , Oxytocin/metabolism , Probiotics/administration & dosage , Skin Physiological Phenomena , Skin/microbiology , Wound Healing/physiology , Adult , Animals , Corticosterone/blood , Dietary Supplements , Female , Humans , Mice , Mice, Knockout , Oxytocin/blood , Oxytocin/genetics , Up-Regulation , Young Adult
8.
PLoS One ; 11(2): e0149178, 2016.
Article in English | MEDLINE | ID: mdl-26906566

ABSTRACT

BACKGROUND: Stacking cross-sectional intravascular images permits three-dimensional rendering of endovascular implants, yet introduces between-frame uncertainties that limit characterization of device placement and the hemodynamic microenvironment. In a porcine coronary stent model, we demonstrate enhanced OCT reconstruction with preservation of between-frame features through fusion with angiography and a priori knowledge of stent design. METHODS AND RESULTS: Strut positions were extracted from sequential OCT frames. Reconstruction with standard interpolation generated discontinuous stent structures. By computationally constraining interpolation to known stent skeletons fitted to 3D 'clouds' of OCT-Angio-derived struts, implant anatomy was resolved, accurately rendering features from implant diameter and curvature (n = 1 vessels, r2 = 0.91, 0.90, respectively) to individual strut-wall configurations (average displacement error ~15 µm). This framework facilitated hemodynamic simulation (n = 1 vessel), showing the critical importance of accurate anatomic rendering in characterizing both quantitative and basic qualitative flow patterns. Discontinuities with standard approaches systematically introduced noise and bias, poorly capturing regional flow effects. In contrast, the enhanced method preserved multi-scale (local strut to regional stent) flow interactions, demonstrating the impact of regional contexts in defining the hemodynamic consequence of local deployment errors. CONCLUSION: Fusion of planar angiography and knowledge of device design permits enhanced OCT image analysis of in situ tissue-device interactions. Given emerging interests in simulation-derived hemodynamic assessment as surrogate measures of biological risk, such fused modalities offer a new window into patient-specific implant environments.


Subject(s)
Hemodynamics , Models, Cardiovascular , Stents , Animals , Swine
9.
Ann Biomed Eng ; 44(7): 2251-60, 2016 07.
Article in English | MEDLINE | ID: mdl-26732391

ABSTRACT

Up to 80% of all endovascular stents have malapposed struts, and while some impose catastrophic events others are inconsequential. Thirteen stents were implanted in coronary arteries of seven healthy Yorkshire pigs, using specially-designed cuffed balloons inducing controlled stent malapposition and under-expansion. Optical coherence tomography (OCT) imaging confirmed that 25% of struts were malapposed (strut-wall distance

Subject(s)
Coronary Vessels , Models, Cardiovascular , Percutaneous Coronary Intervention/adverse effects , Stents/adverse effects , Tomography, Optical Coherence , Animals , Coronary Vessels/diagnostic imaging , Coronary Vessels/physiopathology , Elasticity , Swine
10.
J Am Soc Nephrol ; 27(1): 189-201, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26019318

ABSTRACT

Patients with CKD suffer high rates of thrombosis, particularly after endovascular interventions, yet few options are available to improve management and reduce thrombotic risk. We recently demonstrated that indoxyl sulfate (IS) is a potent CKD-specific prothrombotic metabolite that induces tissue factor (TF) in vascular smooth muscle cells (vSMCs), although the precise mechanism and treatment implications remain unclear. Because IS is an agonist of the aryl hydrocarbon receptor (AHR), we first examined the relationship between IS levels and AHR-inducing activity in sera of patients with ESRD. IS levels correlated significantly with both vSMC AHR activity and TF activity. Mechanistically, we demonstrated that IS activates the AHR pathway in primary human aortic vSMCs, and further, that AHR interacts directly with and stabilizes functional TF. Antagonists directly targeting AHR enhanced TF ubiquitination and degradation and suppressed thrombosis in a postinterventional model of CKD and endovascular injury. Furthermore, AHR antagonists inhibited TF in a manner dependent on circulating IS levels. In conclusion, we demonstrated that IS regulates TF stability through AHR signaling and uncovered AHR as an antithrombotic target and AHR antagonists as a novel class of antithrombotics. Together, IS and AHR have potential as uremia-specific biomarkers and targets that may be leveraged as a promising theranostic platform to better manage the elevated thrombosis rates in patients with CKD.


Subject(s)
Receptors, Aryl Hydrocarbon/physiology , Thromboplastin/physiology , Thrombosis/etiology , Thrombosis/prevention & control , Uremia/complications , Adult , Female , Humans , Indican/physiology , Male , Middle Aged , Protein Stability , Receptors, Aryl Hydrocarbon/antagonists & inhibitors
11.
Interv Cardiol Clin ; 5(3): 295-306, 2016 07.
Article in English | MEDLINE | ID: mdl-28582028

ABSTRACT

The coronary stent has propelled our understanding of the term "biocompatibility." Stents are expanded at sites of arterial blockage and mechanically reestablish blood flow. This simplicity belies the complex reactions that occur when a stent contacts living substrates. Biocompatible seek to elicit the intended response; stents should perform rather than merely exist. Because performance is assessed in the patient, stent biocompatibility is the multiscale examination of material and cell, and of material, structure, and device in the context of cell, tissue, and organism. This review tracks major biomaterial advances in coronary stent design and discusses biocompatibility clinical performance.


Subject(s)
Coronary Stenosis/surgery , Prosthesis Design/instrumentation , Stents , Coronary Restenosis/prevention & control , Drug-Eluting Stents , Humans , Stents/trends
12.
Acta Biomater ; 26: 195-204, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26277377

ABSTRACT

Drug-eluting bioresorbable vascular scaffolds (BVSs) have the potential to restore lumen patency, enable recovery of the native vascular environment, and circumvent late complications associated with permanent endovascular devices. To ensure therapeutic effects persist for sufficient times prior to scaffold resorption and resultant functional loss, many factors dictating BVS performance must be identified, characterized and optimized. While some factors relate to BVS design and manufacturing, others depend on device deployment and intrinsic vascular properties. Importantly, these factors interact and cannot be considered in isolation. The objective of this study is to quantify the extent to which degree of radial expansion modulates BVS performance, specifically in the context of modifying device erosion kinetics and evolution of structural mechanics and local drug elution. We systematically varied degree of radial expansion in model BVS constructs composed of poly dl-lactide-glycolide and generated in vitro metrics of device microstructure, degradation, erosion, mechanics and drug release. Experimental data permitted development of computational models that predicted transient concentrations of scaffold-derived soluble species and drug in the arterial wall, thus enabling speculation on the short- and long-term effects of differential expansion. We demonstrate that degree of expansion significantly affects scaffold properties critical to functionality, underscoring its relevance in BVS design and optimization. STATEMENT OF SIGNIFICANCE: Bioresorbable vascular scaffold (BVS) therapy is beginning to transform the treatment of obstructive artery disease, owing to effective treatment of short term vessel closure while avoiding long term consequences such as in situ, late stent thrombosis - a fatal event associated with permanent implants such as drug-eluting stents. As device scaffolding and drug elution are temporary for BVS, the notion of using this therapy in lieu of existing, clinically approved devices seems attractive. However, there is still a limited understanding regarding the optimal lifetime and performance characteristics of erodible endovascular implants. Several engineering criteria must be met and clinical endpoints confirmed to ensure these devices are both safe and effective. In this manuscript, we sought to establish general principles for the design and deployment of erodible, drug-eluting endovascular scaffolds, with focus on how differential expansion can modulate device performance.


Subject(s)
Absorbable Implants , Blood Vessel Prosthesis , Drug Implants/chemistry , Drug-Eluting Stents , Paclitaxel/chemistry , Tissue Scaffolds , Body Fluids/chemistry , Computer-Aided Design , Corrosion , Diffusion , Drug Implants/administration & dosage , Equipment Design , Equipment Failure Analysis , Lactic Acid/chemistry , Paclitaxel/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Vascular Grafting/instrumentation
14.
Eur Respir J ; 46(3): 651-62, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25837041

ABSTRACT

Uncontrolled pilot studies demonstrated promising results of endoscopic lung volume reduction using emphysematous lung sealant (ELS) in patients with advanced, upper lobe predominant emphysema. We aimed to evaluate the safety and efficacy of ELS in a randomised controlled setting.Patients were randomised to ELS plus medical treatment or medical treatment alone. Despite early termination for business reasons and inability to assess the primary 12-month end-point, 95 out of 300 patients were successfully randomised, providing sufficient data for 3- and 6-month analysis.57 patients (34 treatment and 23 control) had efficacy results at 3 months; 34 (21 treatment and 13 control) at 6 months. In the treatment group, 3-month lung function, dyspnoea, and quality of life improved significantly from baseline when compared to control. Improvements persisted at 6 months with >50% of treated patients experiencing clinically important improvements, including some whose lung function improved by >100%. 44% of treated patients experienced adverse events requiring hospitalisation (2.5-fold more than control, p=0.01), with two deaths in the treated cohort. Treatment responders tended to be those experiencing respiratory adverse events.Despite early termination, results show that minimally invasive ELS may be efficacious, yet significant risks (probably inflammatory) limit its current utility.


Subject(s)
Fibrin Tissue Adhesive/therapeutic use , Pneumonectomy/methods , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/surgery , Quality of Life , Aged , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pulmonary Emphysema/diagnosis , Pulmonary Emphysema/mortality , Respiratory Function Tests , Risk Assessment , Severity of Illness Index , Survival Rate , Time Factors , Tomography, X-Ray Computed/methods , Treatment Outcome
15.
J R Soc Interface ; 12(104): 20141073, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25652458

ABSTRACT

Computational modelling of physical and biochemical processes has emerged as a means of evaluating medical devices, offering new insights that explain current performance, inform future designs and even enable personalized use. Yet resource limitations force one to compromise with reduced order computational models and idealized assumptions that yield either qualitative descriptions or approximate, quantitative solutions to problems of interest. Considering endovascular drug delivery as an exemplary scenario, we used a supervised machine learning framework to process data generated from low fidelity coarse meshes and predict high fidelity solutions on refined mesh configurations. We considered two models simulating drug delivery to the arterial wall: (i) two-dimensional drug-coated balloons and (ii) three-dimensional drug-eluting stents. Simulations were performed on computational mesh configurations of increasing density. Supervised learners based on Gaussian process modelling were constructed from combinations of coarse mesh setting solutions of drug concentrations and nearest neighbourhood distance information as inputs, and higher fidelity mesh solutions as outputs. These learners were then used as computationally inexpensive surrogates to extend predictions using low fidelity information to higher levels of mesh refinement. The cross-validated, supervised learner-based predictions improved fidelity as compared with computational simulations performed at coarse level meshes--a result consistent across all outputs and computational models considered. Supervised learning on coarse mesh solutions can augment traditional physics-based modelling of complex physiologic phenomena. By obtaining efficient solutions at a fraction of the computational cost, this framework has the potential to transform how modelling approaches can be applied in the evaluation of medical technologies and their real-time administration in an increasingly personalized fashion.


Subject(s)
Arteries/pathology , Drug Delivery Systems , Drug-Eluting Stents , Arteries/drug effects , Artificial Intelligence , Computer Simulation , Coronary Artery Disease/drug therapy , Coronary Vessels/drug effects , Finite Element Analysis , Humans , Models, Statistical , Normal Distribution , Reproducibility of Results , Software
16.
Eur Heart J ; 35(46): 3267-76, 2014 Dec 07.
Article in English | MEDLINE | ID: mdl-25265973

ABSTRACT

Despite evidence-based interventions, coronary heart disease (CHD) remains a leading cause of global mortality. As therapies advance, patient non-adherence to established treatments is well recognized. Non-adherence is a powerful confounder of evidence-based practice and can affect daily patient management, resulting in inappropriate therapeutic escalation with greater costs and potential for harm. Moreover, it increases risk for adverse cardiac events, including mortality. Yet, non-adherence is complex, remains difficult to define, and provider ability to identify its presence accurately remains limited. Improved screening tools are needed to detect at-risk patients, enabling appropriate targeting of interventions. Given the rapidly expanding global population with CHD and emerging clinical and cost-benefits of adherence, addressing non-adherence to prescribed therapies is a top priority.


Subject(s)
Cardiovascular Agents/therapeutic use , Coronary Disease/drug therapy , Coronary Disease/prevention & control , Medication Adherence/statistics & numerical data , Global Health/statistics & numerical data , Humans , Patient Education as Topic , Patient Satisfaction/statistics & numerical data , Risk Factors , Treatment Outcome
17.
Cardiovasc Res ; 103(1): 37-46, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24841070

ABSTRACT

AIMS: Atherogenesis, evolution of plaque, and outcomes following endovascular intervention depend heavily on the unique vascular architecture of each individual. Patient-specific, multiscale models able to correlate changes in microscopic cellular responses with relevant macroscopic flow, and structural conditions may help understand the progression of occlusive arterial disease, providing insights into how to mitigate adverse responses in specific settings and individuals. METHODS AND RESULTS: Vascular architectures mimicking coronary and carotid bifurcations were derived from clinical imaging and used to generate conjoint computational meshes for in silico analysis and biocompatible scaffolds for in vitro models. In parallel with three-dimensional flow simulations, geometrically realistic scaffolds were seeded with human smooth muscle cells (SMC) or endothelial cells and exposed to relevant, physiological flows. In vitro surrogates of endothelial health, atherosclerotic progression, and thrombosis were locally quantified and correlated best with an quantified extent of flow recirculation occurring within the bifurcation models. Oxidized low-density lipoprotein uptake, monocyte adhesion, and tissue factor expression locally rose up to three-fold, and phosphorylated endothelial nitric oxide synthase and Krüppel-like factor 2 decreased up to two-fold in recirculation areas. Isolated testing in straight-tube idealized constructs subject to static, oscillatory, and pulsatile conditions, indicative of different recirculant conditions corroborated these flow-mediated dependencies. CONCLUSIONS: Flow drives variations in vascular reactivity and vascular beds. Endothelial health was preserved by arterial flow but jeopardized in regions of flow recirculation in a quasi-linear manner. Similarly, SMC exposed to flow were more thrombogenic in large recirculating regions. Health, thrombosis, and atherosclerosis biomarkers correlate with the extent of recirculation in vascular cells lining certain vascular geometries.


Subject(s)
Arteries/pathology , Arteries/physiopathology , Atherosclerosis/physiopathology , Models, Cardiovascular , Thrombosis/physiopathology , Atherosclerosis/pathology , Biomarkers/metabolism , Biomechanical Phenomena , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Computer Simulation , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Disease Progression , Endothelial Cells/pathology , Endothelial Cells/physiology , Humans , Kruppel-Like Transcription Factors/metabolism , Lipoproteins, LDL/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/physiology , Nitric Oxide Synthase Type III/metabolism , Regional Blood Flow/physiology , Thromboplastin/metabolism , Thrombosis/pathology , Vascular Cell Adhesion Molecule-1/metabolism
18.
J Biomech ; 47(4): 908-21, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24495755

ABSTRACT

Through decades of use and refinement, endovascular stents have become part and parcel of the management of obstructive atherosclerotic lesions. Upon stent placement, a variety of biophysical reactions ensue, governed not only by the mechanical and material properties of the device, but also the impact these properties have on the local vascular biology. Anatomic changes and vascular deformations give rise to solid mechanical and fluid forces that are the proximate, functional drivers of the induced reparative response. Powerful computational tools and advanced imaging techniques allow us to define these forces with high precision and increasingly, at a patient-specific level. We have also gained fundamental insights into how these forces influence subcellular and cellular processes, and, through application of a variety of model systems, how they subsequently drive an integrated tissue response. Clinical studies extend understanding to actual patients and pathophysiologic scenarios. These tools and insights take on added weight given the real risks that accompany the many substantial benefits of stenting. Complex lesions remain difficult to manage and continue to be associated with worse outcomes. While many patients respond well to treatment, others suffer treatment failures and recurrent events - sometimes catastrophic. Overcoming such variability requires that we move towards individualized treatment plans. Doing so necessitates that we develop not just a qualitative understanding of involved phenomena, but a quantitative ability to predict integrated outcomes. Given the multi-scale nature of the vascular response to stenting, it is critical that models, be they computational, bench-top, animal, or clinical, can be verified, validated, and made interrelated. This review provides an overview of the biophysics governing endovascular stenting, their integration in real-world endovascular settings, and how simulation and statistical approaches are helping to bridge the gap between qualitative model understanding and quantitative clinical prediction.


Subject(s)
Coronary Artery Disease/pathology , Coronary Artery Disease/therapy , Endovascular Procedures , Models, Cardiovascular , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/therapy , Animals , Comorbidity , Humans , Predictive Value of Tests , Stents , Uncertainty
19.
Circulation ; 127(3): 365-76, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23269489

ABSTRACT

BACKGROUND: Stent thrombosis (ST), a postinterventional complication with a mortality rate of 50%, has an incidence that rises precipitously in patients at risk. Chronic renal failure and end-stage renal disease have emerged as particularly strong ST risk factors, yet the mechanism remains elusive. Tissue factor (TF) is a crucial mediator of injury-related thrombosis and has been implicated for ST. We posit that uremia modulates TF in the local vessel wall to induce postinterventional thrombosis in patients with end-stage renal disease. METHODS AND RESULTS: As a model of the de-endothelialized, postinterventional state, we exposed primary human vascular smooth muscle cells (vSMCs) pretreated with uremic serum (obtained from ESRD patients on hemodialysis) to coronary-like blood flow. vSMC TF expression, activity, stability, and posttranslational modification were examined after vSMCs were treated with uremic serum or solutes. We found significantly greater clot formation after uremic serum exposure, which was substantially reduced with the prior treatment with anti-TF neutralizing antibody. Uremic sera induced 2- to 3-fold higher TF expression and activity in vSMCs independent of diabetes mellitus. Relevant concentrations of isolated uremic solutes such as indole-3-acetic acid (3.5 µg/mL), indoxyl sulfate (25 µg/mL), and uric acid (80 µg/mL) recapitulated these effects in cell culture and the flow loop model. We show further that TF undergoes ubiquitination at baseline and that uremic serum, indole-3-acetic acid, and indoxyl sulfate significantly prolong TF half-life by inhibiting its ubiquitination. CONCLUSIONS: The uremic milieu is profoundly thrombogenic and upregulates vSMC TF levels by increasing TF stability and decreasing its ubiquitination. Together, these data demonstrate for the first time that the posttranslational regulation of TF in uremia may have a causative role in the increased ST risk observed in uremic patients. These data suggest that interventions that reduce vSMC TF may help to prevent ST and that uremic solutes should be considered as novel risk factors for ST in patients with chronic renal failure.


Subject(s)
Kidney Failure, Chronic/blood , Myocytes, Smooth Muscle/metabolism , Serum , Stents/adverse effects , Thromboplastin/metabolism , Thrombosis/epidemiology , Uremia/blood , Adult , Aged , Cell Line , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , In Vitro Techniques , Indican/pharmacology , Indoleacetic Acids/pharmacology , Kidney Failure, Chronic/therapy , Male , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Renal Dialysis , Risk Factors , Ubiquitination/drug effects
20.
Circulation ; 123(13): 1400-9, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21422389

ABSTRACT

BACKGROUND: Stent thrombosis is a lethal complication of endovascular intervention. Concern has been raised about the inherent risk associated with specific stent designs and drug-eluting coatings, yet clinical and animal support is equivocal. METHODS AND RESULTS: We examined whether drug-eluting coatings are inherently thrombogenic and if the response to these materials was determined to a greater degree by stent design and deployment with custom-built stents. Drug/polymer coatings uniformly reduce rather than increase thrombogenicity relative to matched bare metal counterparts (0.65-fold; P=0.011). Thick-strutted (162 µm) stents were 1.5-fold more thrombogenic than otherwise identical thin-strutted (81 µm) devices in ex vivo flow loops (P<0.001), commensurate with 1.6-fold greater thrombus coverage 3 days after implantation in porcine coronary arteries (P=0.004). When bare metal stents were deployed in malapposed or overlapping configurations, thrombogenicity increased compared with apposed, length-matched controls (1.58-fold, P=0.001; and 2.32-fold, P<0.001). The thrombogenicity of polymer-coated stents with thin struts was lowest in all configurations and remained insensitive to incomplete deployment. Computational modeling-based predictions of stent-induced flow derangements correlated with spatial distribution of formed clots. CONCLUSIONS: Contrary to popular perception, drug/polymer coatings do not inherently increase acute stent clotting; they reduce thrombosis. However, strut dimensions and positioning relative to the vessel wall are critical factors in modulating stent thrombogenicity. Optimal stent geometries and surfaces, as demonstrated with thin stent struts, help reduce the potential for thrombosis despite complex stent configurations and variability in deployment.


Subject(s)
Drug-Eluting Stents/adverse effects , Drug-Eluting Stents/standards , Polymers/administration & dosage , Prosthesis Design/standards , Thrombosis/etiology , Thrombosis/prevention & control , Animals , Cattle , Risk Factors , Swine , Thrombosis/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...