Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Biomech ; 128: 110720, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34482227

ABSTRACT

Atherosclerosis is a lipid driven chronic inflammatory disease that is characterized by the formation of plaques at predilection sites. These predilection sites (side branches, curved segments, and bifurcations) have often been associated with disturbed shear stress profiles. However, in addition to shear stress, endothelial cells also experience artery wall strain that could contribute to atherosclerosis progression. Herein, we describe a method to accurately obtain these shear stress and strain profiles. We developed a fluid-structure interaction (FSI) framework for modelling arteries within a commercially available package (Abaqus, version 6.14) that included known prestresses (circumferential, axial and pressure associated). In addition, we co-registered 3D histology to a micro-CT-derived 3D reconstruction of an atherosclerotic carotid artery from a cholesterol-fed ApoE-/- mouse to include the spatial distribution of lipids within a subject-specific model. The FSI model also incorporated a nonlinear hyperelastic material model with regionally-varying properties that distinguished between healthy vessel wall and plaque. FSI predicted a lower shear stress than CFD (~-12%), but further decreases in plaque regions with softer properties (~-24%) were dependent on the approach used to implement the prestresses in the artery wall. When implemented with our new hybrid approach (zero prestresses in regions of lipid deposition), there was significant heterogeneity in endothelial shear stress in the atherosclerotic artery due to variations in stiffness and, in turn, wall strain. In conclusion, when obtaining endothelial shear stress and strain in diseased arteries, a careful consideration of prestresses is necessary. This paper offers a way to implement them.


Subject(s)
Atherosclerosis , Models, Cardiovascular , Animals , Carotid Arteries , Endothelial Cells , Mice , Shear Strength , Stress, Mechanical
2.
J Vasc Res ; 57(6): 367-375, 2020.
Article in English | MEDLINE | ID: mdl-32937637

ABSTRACT

Aortic aneurysms and dissections are silent and lethal conditions, whose pathogenesis remains incompletely understood. Although angiotensin II (AngII)-infused ApoE-/- mice have been widely used to study aortic aneurysm and dissection, early morphofunctional alterations preceding the onset of these conditions remain unknown. The goal of this study was to unveil early morphofunctional changes underlying the onset of aneurysm and dissection. At 3 days post-AngII infusion, suprarenal abdominal aorta presented significant volumetric dilatation and microstructural damage. Ex vivo assessment of vascular reactivity of the suprarenal dissection-prone aorta and its side branches, showed an endothelial and contractile dysfunctions that were severe in the suprarenal aorta, moderate distally, and absent in the side branches, mirroring the susceptibility to dissection of these different vascular segments. Early and specific morphofunctional changes of the suprarenal aorta may contribute to the regional onset of aortic aneurysm and dissection by exacerbating the biomechanical burden arising from its side branches.


Subject(s)
Angiotensin II , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/pathology , Aortic Dissection/pathology , Vascular Remodeling , Aortic Dissection/chemically induced , Aortic Dissection/diagnostic imaging , Aortic Dissection/physiopathology , Animals , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/physiopathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/physiopathology , Aortography , Computed Tomography Angiography , Dilatation, Pathologic , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Time Factors , Vasoconstriction , X-Ray Microtomography
3.
Biomech Model Mechanobiol ; 19(1): 81-97, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31273562

ABSTRACT

Animal models of aortic aneurysm and dissection can enhance our limited understanding of the etiology of these lethal conditions particularly because early-stage longitudinal data are scant in humans. Yet, the pathogenesis of often-studied mouse models and the potential contribution of aortic biomechanics therein remain elusive. In this work, we combined micro-CT and synchrotron-based imaging with computational biomechanics to estimate in vivo aortic strains in the abdominal aorta of angiotensin-II-infused ApoE-deficient mice, which were compared with mouse-specific aortic microstructural damage inferred from histopathology. Targeted histology showed that the 3D distribution of micro-CT contrast agent that had been injected in vivo co-localized with precursor vascular damage in the aortic wall at 3 days of hypertension, with damage predominantly near the ostia of the celiac and superior mesenteric arteries. Computations similarly revealed higher mechanical strain in branching relative to non-branching regions, thus resulting in a positive correlation between high strain and vascular damage in branching segments that included the celiac, superior mesenteric, and right renal arteries. These results suggest a mechanically driven initiation of damage at these locations, which was supported by 3D synchrotron imaging of load-induced ex vivo delaminations of angiotensin-II-infused suprarenal abdominal aortas. That is, the major intramural delamination plane in the ex vivo tested aortas was also near side branches and specifically around the celiac artery. Our findings thus support the hypothesis of an early mechanically mediated formation of microstructural defects at aortic branching sites that subsequently propagate into a macroscopic medial tear, giving rise to aortic dissection in angiotensin-II-infused mice.


Subject(s)
Angiotensin II/administration & dosage , Aorta/pathology , Stress, Mechanical , Animals , Aorta/diagnostic imaging , Aortic Rupture/diagnostic imaging , Aortic Rupture/pathology , Computer Simulation , Contrast Media/chemistry , Finite Element Analysis , Imaging, Three-Dimensional , Male , Mice, Inbred C57BL
4.
J R Soc Interface ; 16(155): 20190179, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31238834

ABSTRACT

In computational aortic biomechanics, aortic and arterial tissue are typically modelled as a homogeneous layer, making abstraction not only of the layered structure of intima, media and adventitia but also of the microstructure that exists within these layers. Here, we present a novel method to visualize the microstructure of the tunica media along the entire circumference of the vessel. To that end, we developed a pressure-inflation device that is compatible with synchrotron-based phase-contrast imaging. Using freshly excised left common carotid arteries from n = 12 mice, we visualized how the lamellae and interlamellar layers inflate as the luminal pressure is increased from 0 to 120 mm Hg in quasi-static steps. A graph-based segmentation algorithm subsequently allowed us to automatically segment each of the three lamellae, resulting in a three-dimensional geometry that represents lamellae, interlamellar layers and adventitia at nine different pressure levels. Our results demonstrate that the three elastic lamellae unfold and stretch simultaneously as luminal pressure is increased. In the long term, we believe that the results presented in this work can be a first step towards a better understanding of the mechanics of the arterial microstructure.


Subject(s)
Blood Pressure/physiology , Carotid Artery, Common , Models, Cardiovascular , Synchrotrons , Vascular Stiffness/physiology , Animals , Carotid Artery, Common/diagnostic imaging , Carotid Artery, Common/physiology , Male , Mice , Mice, Knockout, ApoE
5.
Sci Rep ; 8(1): 2223, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396472

ABSTRACT

In order to show the advantage and potential of propagation-based phase-contrast synchrotron imaging in vascular pathology research, we analyzed aortic medial ruptures in BAPN/AngII-infused mice, a mouse model for aortic dissection. Ascending and thoraco-abdominal samples from n = 3 control animals and n = 10 BAPN/AngII-infused mice (after 3, 7 and 14 days of infusion, total of 24 samples) were scanned. A steep increase in the number of ruptures was already noted after 3 days of BAPN/AngII-infusion. The largest ruptures were found at the latest time points. 133 ruptures affected only the first lamella while 135 ruptures affected multiple layers. Medial ruptures through all lamellar layers, leading to false channel formation and intramural hematoma, occurred only in the thoraco-abdominal aorta and interlamellar hematoma formation in the ascending aorta could be directly related to ruptures of the innermost lamellae. The advantages of this technique are (i) ultra-high resolution that allows to visualize the individual elastic lamellae in the aorta; (ii) quantitative and qualitative analysis of medial ruptures; (iii) 3D analysis of the complete aorta; (iv) high contrast for qualitative information extraction, reducing the need for histology coupes; (v) earlier detection of (micro-) ruptures.


Subject(s)
Aorta, Abdominal/pathology , Aorta, Thoracic/pathology , Aortic Aneurysm, Abdominal/pathology , Aortic Dissection/pathology , Aortic Rupture/pathology , Aortic Dissection/diagnostic imaging , Angiotensin II/administration & dosage , Animals , Aorta, Abdominal/diagnostic imaging , Aorta, Thoracic/diagnostic imaging , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Rupture/diagnostic imaging , Disease Models, Animal , Imaging, Three-Dimensional , Male , Mice , Mice, Inbred C57BL , Microscopy, Phase-Contrast/methods , Synchrotrons
6.
Ann Biomed Eng ; 46(1): 159-170, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29071528

ABSTRACT

In order to advance the state-of-the-art in computational aortic biomechanics, we investigated the influence of (i) a non-uniform wall thickness, (ii) minor aortic side branches and (iii) a non-uniform axial stretch distribution on the location of predicted hotspots of principal strain in a mouse model for dissecting aneurysms. After 3 days of angiotensin II infusion, a murine abdominal aorta was scanned in vivo with contrast-enhanced micro-CT. The animal was subsequently sacrificed and its aorta was scanned ex vivo with phase-contrast X-ray tomographic microscopy (PCXTM). An automatic morphing framework was developed to map the non-pressurized, non-stretched PCXTM geometry onto the pressurized, stretched micro-CT geometry. The output of the morphing model was a structural FEM simulation where the output strain distribution represents an estimation of the wall deformation, not only due to the pressurization, but also due to the local axial stretch field. The morphing model also included minor branches and a mouse-specific wall thickness. A sensitivity study was then performed to assess the influence of each of these novel features on the outcome of the simulations. The results were supported by comparing the computed hotspots of principal strain to hotspots of early vascular damage as detected on PCXTM. Non-uniform axial stretch, non-uniform wall thickness and minor subcostal arteries significantly alter the locations of calculated hotspots of maximal principal strain. Even if experimental data on these features are often not available in clinical practice, one should be aware of the important implications that simplifications in the model might have on the final simulated result.


Subject(s)
Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/physiology , Angiotensin II/pharmacology , Animals , Aorta, Abdominal/drug effects , Male , Mice , Mice, Knockout, ApoE , Tomography, X-Ray/methods
7.
Cardiovasc Res ; 113(10): 1230-1242, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28898997

ABSTRACT

AIMS: Angiotensin II-infused ApoE-/- mice are a popular mouse model for preclinical aneurysm research. Here, we provide insight in the often-reported but seldom-explained variability in shape of dissecting aneurysms in these mice. METHODS AND RESULTS: N = 45 excised aortas were scanned ex vivo with phase-contrast X-ray tomographic microscopy. Micro-ruptures were detected near the ostium of celiac and mesenteric arteries in 8/11 mice that were sacrificed after 3 days of angiotensin II-infusion. At later time points (after 10, 18, and 28 days) the variability in shape of thoraco-abdominal lesions (occurring in 31/34 mice) was classified into 7 different categories based on the presence or absence of a medial tear (31/31), an intramural hematoma (23/31) or a false channel (11/23). Medial tears were detected both in the thoracic and the abdominal aorta and were most prevalent at the left and ventral aspects of celiac and mesenteric arteries. The axial length of the hematoma strongly correlated to the total number of ruptured branch ostia (r2 = 0.78) and in 22/23 mice with a hematoma the ostium of the left suprarenal artery had ruptured. Supraceliac diameters at baseline were significantly lower for mice that did not develop an intramural hematoma, and the formation of a false channel within that intramural hematoma depended on the location, rather than the length, of the medial tear. CONCLUSION: Based on our observations we propose an elaborate hypothesis that explains how aortic side branches (i) affect the initiation and propagation of medial tears and the subsequent adventitial dissection and (ii) affect the variability in shape of dissecting aneurysms. This hypothesis was partially validated through the live visualization of a dissecting aneurysm that formed during micro-CT imaging, and led us to the conclusion that angiotensin II-infused mice are more clinically relevant for the study of aortic dissections than for the study of abdominal aortic aneurysms.


Subject(s)
Angiotensin II , Aorta, Abdominal/pathology , Aorta, Thoracic/pathology , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Thoracic/pathology , Aortic Dissection/pathology , Aortic Dissection/chemically induced , Aortic Dissection/genetics , Aortic Dissection/metabolism , Animals , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/metabolism , Aorta, Thoracic/diagnostic imaging , Aorta, Thoracic/metabolism , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Thoracic/chemically induced , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortography/methods , Computed Tomography Angiography , Disease Models, Animal , Disease Progression , Hematoma/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Time Factors , Ultrasonography, Doppler, Pulsed , Vascular Remodeling , X-Ray Microtomography
8.
Arterioscler Thromb Vasc Biol ; 36(4): 673-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26891740

ABSTRACT

OBJECTIVE: To understand the anatomy and physiology of ascending aortic aneurysms in angiotensin II-infused ApoE(-/-) mice. APPROACH AND RESULTS: We combined an extensive in vivo imaging protocol (high-frequency ultrasound and contrast-enhanced microcomputed tomography at baseline and after 3, 10, 18, and 28 days of angiotensin II infusion) with synchrotron-based ultrahigh resolution ex vivo imaging (phase contrast X-ray tomographic microscopy) in n=47 angiotensin II-infused mice and 6 controls. Aortic regurgitation increased significantly over time, as did the luminal volume of the ascending aorta. In the samples that were scanned ex vivo, we observed one or several focal dissections, with the largest located in the outer convex aspect of the ascending aorta. The volume of the dissections moderately correlated to the volume of the aneurysm as measured in vivo (r(2)=0.46). After 3 days of angiotensin II infusion, we found an interlaminar hematoma in 7/12 animals, which could be linked to an intimal tear. There was also a significant increase in single laminar ruptures, which may have facilitated a progressive enlargement of the focal dissections over time. At later time points, the hematoma was resorbed and the medial and adventitial thickness increased. Fatal transmural dissection occurred in 8/47 mice at an early stage of the disease, before adventita remodeling. CONCLUSIONS: We visualized and quantified the dissections that lead to ascending aortic aneurysms in angiotensin II-infused mice and provided unique insight into the temporal evolution of these lesions.


Subject(s)
Aorta/pathology , Aortic Aneurysm, Abdominal/pathology , Aortic Dissection/pathology , Aortic Rupture/pathology , Vascular Remodeling , Aortic Dissection/chemically induced , Aortic Dissection/diagnostic imaging , Angiotensin II , Animals , Aorta/diagnostic imaging , Aorta/metabolism , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Rupture/chemically induced , Aortic Rupture/diagnostic imaging , Aortic Valve Insufficiency/etiology , Aortography/methods , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Dilatation, Pathologic , Disease Models, Animal , Disease Progression , Elastic Tissue/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Time Factors , Ultrasonography, Doppler, Pulsed , X-Ray Microtomography
9.
ALTEX ; 33(1): 13-28, 2016.
Article in English | MEDLINE | ID: mdl-26555250

ABSTRACT

At a time of growing concern over the ethics of animal experimentation, mouse models are still an indispensable source of insight into the cardiovascular system and its most frequent pathologies. Nevertheless, reference data on the murine cardiovascular anatomy and physiology are lacking. In this work, we developed and validated an in silico, one dimensional model of the murine systemic arterial tree consisting of 85 arterial segments. Detailed aortic dimensions were obtained in vivo from contrast-enhanced micro-computed tomography in 3 male, C57BL/6J anesthetized mice and 3 male ApoE(-/-) mice, all 12-weeks old. Physiological input data were gathered from a wide range of literature data. The integrated form of the Navier-Stokes equations was solved numerically to yield pressures and flows throughout the arterial network. The resulting model predictions have been validated against invasive pressure waveforms and non-invasive velocity and diameter waveforms that were measured in vivo on an independent set of 47 mice. In conclusion, we present a validated one-dimensional model of the anesthetized murine cardiovascular system that can serve as a versatile tool in the field of preclinical cardiovascular research.


Subject(s)
Animal Testing Alternatives , Arteries/physiology , Computer Simulation , Models, Cardiovascular , Algorithms , Animals , Arteries/anatomy & histology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Ultrasonography, Doppler, Duplex , X-Ray Microtomography/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...