Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Biomech Eng ; 141(12)2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31633165

ABSTRACT

Ascending thoracic aortic aneurysms (ATAAs) are anatomically complex in terms of architecture and geometry, and both complexities contribute to unpredictability of ATAA dissection and rupture in vivo. The goal of this work was to examine the mechanism of ATAA failure using a combination of detailed mechanical tests on human tissue and a multiscale computational model. We used (1) multiple, geometrically diverse, mechanical tests to characterize tissue properties; (2) a multiscale computational model to translate those results into a broadly usable form; and (3) a model-based computer simulation of the response of an ATAA to the stresses generated by the blood pressure. Mechanical tests were performed in uniaxial extension, biaxial extension, shear lap, and peel geometries. ATAA tissue was strongest in circumferential extension and weakest in shear, presumably because of the collagen and elastin in the arterial lamellae. A multiscale, fiber-based model using different fiber properties for collagen, elastin, and interlamellar connections was specified to match all of the experimental data with one parameter set. Finally, this model was used to simulate ATAA inflation using a realistic geometry. The predicted tissue failure occurred in regions of high stress, as expected; initial failure events involved almost entirely interlamellar connections, consistent with arterial dissection-the elastic lamellae remain intact, but the connections between them fail. The failure of the interlamellar connections, paired with the weakness of the tissue under shear loading, is suggestive that shear stress within the tissue may contribute to ATAA dissection.

2.
J Biomech Eng ; 141(9)2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31141605

ABSTRACT

The scar that forms after a myocardial infarction is often characterized by a highly disordered architecture but generally exhibits some degree of collagen fiber orientation, with a resulting mechanical anisotropy. When viewed in finer detail, however, the heterogeneity of the sample is clear, with different subregions exhibiting different fiber orientations. In this work, we used a multiscale finite element model to explore the consequences of the heterogeneity in terms of mechanical behavior. To do so, we used previously obtained fiber alignment maps of rat myocardial scar slices (n = 15) to generate scar-specific finite element meshes that were populated with fiber models based on the local alignment state. These models were then compared to isotropic models with the same sample shape and fiber density, and to homogeneous models with the same sample shape, fiber density, and average fiber alignment as the scar-specific models. All simulations involved equibiaxial extension of the sample with free motion in the third dimension. We found that heterogeneity led to a lower degree of mechanical anisotropy and a higher level of local stress concentration than the corresponding homogeneous model, and also that fibers failed in the heterogeneous model at much lower macroscopic strains than in the isotropic and homogeneous models. Taken together, these results suggest that scar heterogeneity may impair myocardial mechanical function both in terms of anisotropy and strength, and that individual variations in scar heterogeneity could be an important consideration for understanding scar remodeling and designing therapeutic interventions for patients after myocardial infarction.

3.
Am J Respir Crit Care Med ; 198(4): 486-496, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29579397

ABSTRACT

RATIONALE: The lung extracellular matrix (ECM) in idiopathic pulmonary fibrosis (IPF) mediates progression of fibrosis by decreasing fibroblast expression of miR-29 (microRNA-29), a master negative regulator of ECM production. The molecular mechanism is undefined. IPF-ECM is stiffer than normal. Stiffness drives fibroblast ECM production in a YAP (yes-associated protein)-dependent manner, and YAP is a known regulator of miR-29. Therefore, we tested the hypothesis that negative regulation of miR-29 by IPF-ECM was mediated by mechanotransduction of stiffness. OBJECTIVES: To determine how IPF-ECM negatively regulates miR-29. METHODS: We decellularized lung ECM using detergents and prepared polyacrylamide hydrogels of defined stiffness by varying acrylamide concentrations. Mechanistic studies were guided by immunohistochemistry of IPF lung and used cell culture, RNA-binding protein assays, and xenograft models. MEASUREMENTS AND MAIN RESULTS: Contrary to our hypothesis, we excluded fibroblast mechanotransduction of ECM stiffness as the primary mechanism deregulating miR-29. Instead, systematic examination of miR-29 biogenesis revealed a microRNA processing defect that impeded processing of miR-29 into its mature bioactive forms. Immunohistochemical analysis of the microRNA processing machinery in IPF lung specimens revealed decreased Dicer1 expression in the procollagen-rich myofibroblastic core of fibroblastic foci compared with the focus perimeter and adjacent alveolar walls. Mechanistically, IPF-ECM increased association of the Dicer1 transcript with RNA binding protein AUF1 (AU-binding factor 1), and Dicer1 knockdown conferred primary human lung fibroblasts with cell-autonomous fibrogenicity in zebrafish and mouse lung xenograft models. CONCLUSIONS: Our data identify suppression of fibroblast Dicer1 expression in the myofibroblast-rich IPF fibroblastic focus core as a central step in the mechanism by which the ECM sustains fibrosis progression in IPF.


Subject(s)
DEAD-box RNA Helicases/genetics , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , MicroRNAs/metabolism , Ribonuclease III/genetics , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibrosis/genetics , Fibrosis/pathology , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Lung/pathology , Mice , Zebrafish
4.
J Biomech Eng ; 139(7)2017 07 01.
Article in English | MEDLINE | ID: mdl-28334369

ABSTRACT

The von Mises (VM) stress is a common stress measure for finite element models of tissue mechanics. The VM failure criterion, however, is inherently isotropic, and therefore may yield incorrect results for anisotropic tissues, and the relevance of the VM stress to anisotropic materials is not clear. We explored the application of a well-studied anisotropic failure criterion, the Tsai­Hill (TH) theory, to the mechanically anisotropic porcine aorta. Uniaxial dogbones were cut at different angles and stretched to failure. The tissue was anisotropic, with the circumferential failure stress nearly twice the axial (2.67 ± 0.67 MPa compared to 1.46 ± 0.59 MPa). The VM failure criterion did not capture the anisotropic tissue response, but the TH criterion fit the data well (R2 = 0.986). Shear lap samples were also tested to study the efficacy of each criterion in predicting tissue failure. Two-dimensional failure propagation simulations showed that the VM failure criterion did not capture the failure type, location, or propagation direction nearly as well as the TH criterion. Over the range of loading conditions and tissue geometries studied, we found that problematic results that arise when applying the VM failure criterion to an anisotropic tissue. In contrast, the TH failure criterion, though simplistic and clearly unable to capture all aspects of tissue failure, performed much better. Ultimately, isotropic failure criteria are not appropriate for anisotropic tissues, and the use of the VM stress as a metric of mechanical state should be reconsidered when dealing with anisotropic tissues.


Subject(s)
Stress, Mechanical , Animals , Anisotropy , Aorta, Abdominal/cytology , Biomechanical Phenomena , Finite Element Analysis , Materials Testing , Swine
5.
J Biomech Eng ; 139(3)2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27893044

ABSTRACT

The ascending thoracic aorta is poorly understood mechanically, especially its risk of dissection. To make better predictions of dissection risk, more information about the multidimensional failure behavior of the tissue is needed, and this information must be incorporated into an appropriate theoretical/computational model. Toward the creation of such a model, uniaxial, equibiaxial, peel, and shear lap tests were performed on healthy porcine ascending aorta samples. Uniaxial and equibiaxial tests showed anisotropy with greater stiffness and strength in the circumferential direction. Shear lap tests showed catastrophic failure at shear stresses (150-200 kPa) much lower than uniaxial tests (750-2500 kPa), consistent with the low peel tension (∼60 mN/mm). A novel multiscale computational model, including both prefailure and failure mechanics of the aorta, was developed. The microstructural part of the model included contributions from a collagen-reinforced elastin sheet and interlamellar connections representing fibrillin and smooth muscle. Components were represented as nonlinear fibers that failed at a critical stretch. Multiscale simulations of the different experiments were performed, and the model, appropriately specified, agreed well with all experimental data, representing a uniquely complete structure-based description of aorta mechanics. In addition, our experiments and model demonstrate the very low strength of the aorta in radial shear, suggesting an important possible mechanism for aortic dissection.


Subject(s)
Aorta, Thoracic , Mechanical Phenomena , Models, Biological , Animals , Biomechanical Phenomena , Male , Materials Testing , Stress, Mechanical , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...