Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Comput Biol Med ; 136: 104717, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34426166

RESUMEN

When striving for reconstructing and predicting bone remodeling processes by means of mathematical models, cell population models have become a popular option. From a conceptual point of view, these models are able to take into account an arbitrary amount of regulatory mechanisms driving the development of bone cells and their activities. However, in most cases, the models include a large number of parameters; and most of those parameters cannot be measured, which certainly compromises the credibility of cell population models. Here, new insights are presented as to the potential improvement of this unsatisfactory situation. In particular, a previously published bone remodeling model was considered, and based on combination and merging of the original parameters, the total number of parameters could be reduced from 28 to 18, without impairing the model's versatility and significance. Furthermore, a comprehensive number of one- and two-variable sensitivity studies were performed, pointing out which parameters (alone and in combination with other parameters) influence the model predictions significantly - for that purpose, the mean squared relative error (MSRE) between simulations based on the original parameters and based on varied parameters was considered as failure measure. It has turned out that the model is significantly more sensitive to parameters which can be considered as phenomenological (such as differentiation, proliferation, and apoptosis rates) than to parameters which are directly related to specific processes (such as dissociation rate constants, and maximum concentrations of the involved factors). Using common correlation measures (such as Pearson, Spearman, and partial ranked correlation coefficients), correlation studies revealed that the correlations between most parameters and the MSRE are weak, while a few parameters exhibited moderate correlations. In conclusion, the results shown in this paper provide valuable insights concerning the design of new experiments allowing for measurement of the parameters which are most influential in the context of bone remodeling simulation.


Asunto(s)
Remodelación Ósea , Modelos Teóricos , Simulación por Computador , Correlación de Datos
2.
Chaos Solitons Fractals ; 136: 109891, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32508398

RESUMEN

The COVID-19 pandemic has world-widely motivated numerous attempts to properly adjust classical epidemiological models, namely those of the SEIR-type, to the spreading characteristics of the novel Corona virus. In this context, the fundamental structure of the differential equations making up the SEIR models has remained largely unaltered-presuming that COVID-19 may be just "another epidemic". We here take an alternative approach, by investigating the relevance of one key ingredient of the SEIR models, namely the death kinetics law. The latter is compared to an alternative approach, which we call infection-to-death delay rule. For that purpose, we check how well these two mathematical formulations are able to represent the publicly available country-specific data on recorded fatalities, across a selection of 57 different nations. Thereby, we consider that the model-governing parameters-namely, the death transmission coefficient for the death kinetics model, as well as the apparent fatality-to-case fraction and the characteristic fatal illness period for the infection-to-death delay rule-are time-invariant. For 55 out of the 57 countries, the infection-to-death delay rule turns out to represent the actual situation significantly more precisely than the classical death kinetics rule. We regard this as an important step towards making SEIR-approaches more fit for the COVID-19 spreading prediction challenge.

3.
Biomech Model Mechanobiol ; 19(5): 1765-1780, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32100180

RESUMEN

One of only a few approved and available anabolic treatments for severe osteoporosis is daily injections of PTH (1-34). This drug has a specific dual action which can act either anabolically or catabolically depending on the type of administration, i.e. intermittent or continuous, respectively. In this paper, we present a mechanistic pharmacokinetic-pharmacodynamic model of the action of PTH in postmenopausal osteoporosis. This model accounts for anabolic and catabolic activities in bone remodelling under intermittent and continuous administration of PTH. The model predicts evolution of common bone biomarkers and bone volume fraction (BV/TV) over time. We compared the relative changes in BV/TV resulting from a daily injection of 20 [Formula: see text]g of PTH with experimental data from the literature. Simulation results indicate a site-specific bone gain of 8.66[Formula: see text] (9.4 ± 1.13[Formula: see text]) at the lumbar spine and 3.14[Formula: see text] (2.82 ± 0.72[Formula: see text]) at the femoral neck. Bone gain depends nonlinearly on the administered dose, being, respectively, 0.68[Formula: see text], 3.4[Formula: see text] and 6.16[Formula: see text] for a 10, 20 and 40 [Formula: see text]g PTH dose at the FN over 2 years. Simulations were performed also taking into account a bone mechanical disuse to reproduce elderly frail subjects. The results show that mechanical disuse ablates the effects of PTH and leads to a 1.08% reduction of bone gain at the FN over a 2-year treatment period for the 20 [Formula: see text]g of PTH. The developed model can simulate a range of pathological conditions and treatments in bones including different PTH doses, different mechanical loading environments and combinations. Consequently, the model can be used for testing and generating hypotheses related to synergistic action between PTH treatment and physical activity.


Asunto(s)
Modelos Biológicos , Osteoporosis Posmenopáusica/tratamiento farmacológico , Osteoporosis Posmenopáusica/fisiopatología , Hormona Paratiroidea/farmacocinética , Hormona Paratiroidea/uso terapéutico , Fenómenos Biomecánicos , Simulación por Computador , Femenino , Humanos , Tamaño de los Órganos/efectos de los fármacos , Osteoporosis Posmenopáusica/sangre , Hormona Paratiroidea/administración & dosificación , Hormona Paratiroidea/sangre , Soporte de Peso
4.
J Theor Biol ; 473: 67-79, 2019 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-31009612

RESUMEN

This paper presents a pharmacokinetic/pharmacodynamic (PK/PD) model of the action of PTH(1-34) on bone modelling and remodelling, developed for quantitatively investigating the dose- and administration pattern-dependency of the bone tissue response to this drug. Firstly, a PK model of PTH(1-34) was developed, accounting for administration via subcutaneous injections. Subsequently, the PK model was coupled to a (mechanistic) bone cell population model of bone modelling and remodelling, taking into account the effects of PTH(1-34) on the differentiation of lining cells into active osteoblasts, on the apoptosis of active osteoblasts, and on proliferation of osteoblast precursors, as well as on the key regulatory pathways of bone cell activities. Numerical simulations show that the coupled PK/PD model is able to distinguish between continuous and intermittent administration patterns of PTH(1-34), in terms of yielding both catabolic bone responses (if drug administration is carried out continuously) and anabolic bone responses (if drug administration is carried out intermittently). The model also features a non-linear relation between bone gain and drug dose (as known from experiments); doubling the dose from 80 µg/kg/day to 160 µg/kg/day induced a 1.3-fold increase of the bone volume-to-total volume ratio. Furthermore, the model presented in this paper confirmed that bone modelling represents an essential mechanism of the anabolic response of bone to PTH(1-34) administration in rat models, and that the large amount of bone formation observed in such models cannot be explained via remodelling alone.


Asunto(s)
Modelos Biológicos , Osteoporosis/tratamiento farmacológico , Hormona Paratiroidea/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Calibración , Modelos Animales de Enfermedad , Femenino , Humanos , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Osteoporosis/sangre , Ovariectomía , Hormona Paratiroidea/sangre , Hormona Paratiroidea/farmacocinética , Hormona Paratiroidea/farmacología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
5.
Mater Sci Eng C Mater Biol Appl ; 95: 389-396, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30573263

RESUMEN

Nowadays, the assessment of the mechanical competence of tissue engineering scaffolds based on computer simulations is a well-accepted technology. Typically, such simulations are performed by means of the Finite Element (FE) method, with the underlying structural model being created based on micro-computed tomography (microCT). Here, this analysis modality is applied to a new, ternary composite, consisting of PHBV, i.e. poly(3-hydroxybutyrate-co-3-hydroxyvalerate), PLGA, i.e. poly(lactic-co-glycolide), as well as of TCP, i.e. tricalcium phosphate hydrate. The studied scaffold structure is made up by fibers of this new composite material, manufactured by means of the rapid prototyping method. The data collected from microCT is utilized for adequately defining the mechanical properties of the FE model. In particular, the three-dimensional field of grey values is interpreted in terms of the underlying field of attenuation coefficients, taking into account the photon energy employed in microCT imaging, eventually allowing for calculation of the three-dimensionally distributed, voxel-specific composition of the studied material. For the sake of keeping the FE simulations as efficient as possible, groups of voxels are combined into one finite element; the grey value of the latter is obtained by volume averaging. Employing a two-step micromechanical homogenization scheme, the experimentally accessible stiffness of the three constituents (PHBV, PLGA, and TCP) is then, finite element by finite element, upscaled to the composition-dependent stiffness of the composite material. The plausibility and adequacy of the FE model is demonstrated by simulating the effects of uniaxial compression on the scaffold structure, in terms of resulting stress and strain fields, highlighting the importance of the fiber junctions (as they are the mechanically most stressed regions), and that neglecting the material heterogeneity would lead to a potentially significant underestimation of stresses and strains. Finally, a comparison is made of the employed analysis modality of microCT data with a previously pursued, simplified analysis strategy, highlighting the conceptual superiority of the former, and pointing out the application limits of the latter.


Asunto(s)
Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ingeniería de Tejidos/métodos , Análisis de Elementos Finitos , Andamios del Tejido/química , Microtomografía por Rayos X
6.
J Theor Biol ; 453: 125-135, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-29778649

RESUMEN

A combined experimental/theoretical approach is presented, for improving the predictability of Saccharomyces cerevisiae fermentations. In particular, a mathematical model was developed explicitly taking into account the main mechanisms of the fermentation process, allowing for continuous computation of key process variables, including the biomass concentration and the respiratory quotient (RQ). For model calibration and experimental validation, batch and fed-batch fermentations were carried out. Comparison of the model-predicted biomass concentrations and RQ developments with the corresponding experimentally recorded values shows a remarkably good agreement for both batch and fed-batch processes, confirming the adequacy of the model. Furthermore, sensitivity studies were performed, in order to identify model parameters whose variations have significant effects on the model predictions: our model responds with significant sensitivity to the variations of only six parameters. These studies provide a valuable basis for model reduction, as also demonstrated in this paper. Finally, optimization-based parametric studies demonstrate how our model can be utilized for improving the efficiency of Saccharomyces cerevisiae fermentations.


Asunto(s)
Etanol/metabolismo , Fermentación , Modelos Biológicos , Saccharomyces cerevisiae/metabolismo , Biomasa , Calibración , Cinética , Modelos Teóricos , Consumo de Oxígeno/fisiología , Proyectos de Investigación , Biología de Sistemas
7.
Bone ; 107: 208-221, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29170108

RESUMEN

While bone tissue is a hierarchically organized material, mathematical formulations of bone remodeling are often defined on the level of a millimeter-sized representative volume element (RVE), "smeared" over all types of bone microstructures seen at lower observation scales. Thus, there is no explicit consideration of the fact that the biological cells and biochemical factors driving bone remodeling are actually located in differently sized pore spaces: active osteoblasts and osteoclasts can be found in the vascular pores, whereas the lacunar pores host osteocytes - bone cells originating from former osteoblasts which were then "buried" in newly deposited extracellular bone matrix. We here propose a mathematical description which considers size and shape of the pore spaces where the biological and biochemical events take place. In particular, a previously published systems biology formulation, accounting for biochemical regulatory mechanisms such as the rank-rankl-opg pathway, is cast into a multiscale framework coupled to a poromicromechanical model. The latter gives access to the vascular and lacunar pore pressures arising from macroscopic loading. Extensive experimental data on the biological consequences of this loading strongly suggest that the aforementioned pore pressures, together with the loading frequency, are essential drivers of bone remodeling. The novel approach presented here allows for satisfactory simulation of the evolution of bone tissue under various loading conditions, and for different species; including scenarios such as mechanical dis- and overuse of murine and human bone, or in osteocyte-free bone.


Asunto(s)
Remodelación Ósea/fisiología , Mecanotransducción Celular/fisiología , Modelos Biológicos , Modelos Teóricos , Animales , Huesos/metabolismo , Humanos , Osteocitos/metabolismo
8.
Front Physiol ; 7: 383, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27708584

RESUMEN

We here explore for the very first time how an advanced multiscale mathematical modeling approach may support the design of a provenly successful tissue engineering concept for mandibular bone. The latter employs double-porous, potentially cracked, single millimeter-sized granules packed into an overall conglomerate-type scaffold material, which is then gradually penetrated and partially replaced by newly grown bone tissue. During this process, the newly developing scaffold-bone compound needs to attain the stiffness of mandibular bone under normal physiological conditions. In this context, the question arises how the compound stiffness is driven by the key design parameters of the tissue engineering system: macroporosity, crack density, as well as scaffold resorption/bone formation rates. We here tackle this question by combining the latest state-of-the-art mathematical modeling techniques in the field of multiscale micromechanics, into an unprecedented suite of highly efficient, semi-analytically defined computation steps resolving several levels of hierarchical organization, from the millimeter- down to the nanometer-scale. This includes several types of homogenization schemes, namely such for porous polycrystals with elongated solid elements, for cracked matrix-inclusion composites, as well as for assemblies of coated spherical compounds. Together with the experimentally known stiffnesses of hydroxyapatite crystals and mandibular bone tissue, the new mathematical model suggests that early stiffness recovery (i.e., within several weeks) requires total avoidance of microcracks in the hydroxyapatite scaffolds, while mid-term stiffness recovery (i.e., within several months) is additionally promoted by provision of small granule sizes, in combination with high bone formation and low scaffold resorption rates.

9.
Biomech Model Mechanobiol ; 15(1): 9-28, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26220453

RESUMEN

Mechanical loads which are macroscopically acting onto bony organs, are known to influence the activities of biological cells located in the pore spaces of bone, in particular so the signaling and production processes mediated by osteocytes. The exact mechanisms by which osteocytes are actually able to "feel" the mechanical loading and changes thereof, has been the subject of numerous studies, and, while several hypotheses have been brought forth over time, this topic has remained a matter of debate. Relaxation times reported in a recent experimental study of Gardinier et al. (Bone 46(4):1075-1081, 2010) strongly suggest that the lacunar pores are likely to experience, during typical physiological load cycles, not only fluid transport, but also undrained conditions. The latter entail the buildup of lacunar pore pressures, which we here quantify by means of a thorough multiscale modeling approach. In particular, the proposed model is based on classical poroelasticity theory, and able to account for multiple pore spaces. First, the model reveals distinct nonlinear dependencies of the resulting lacunar (and vascular) pore pressures on the underlying bone composition, highlighting the importance of a rigorous multiscale approach for appropriate computation of the aforementioned pore pressures. Then, the derived equations are evaluated for macroscopic (uniaxial as well as hydrostatic) mechanical loading of physiological magnitude. The resulting model-predicted pore pressures agree very well with the pressures that have been revealed, by means of in vitro studies, to be of adequate magnitude for modulating the responses of biological cells, including osteocytes. This underlines that osteocytes may respond to many types of loading stimuli at the same time, in particular so to fluid flow and hydrostatic pressure.


Asunto(s)
Huesos/fisiología , Osteocitos/fisiología , Presión , Envejecimiento/fisiología , Animales , Fenómenos Biomecánicos , Bovinos , Articulación de la Cadera/fisiología , Humanos , Presión Hidrostática , Ratones Endogámicos C57BL , Porosidad , Ratas , Estrés Mecánico , Caminata , Soporte de Peso
10.
Int J Fract ; 202(2): 217-235, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-32269408

RESUMEN

Bone tissue engineering materials must blend in the targeted physiological environment, in terms of both the materials' biocompatibility and mechanical properties. As for the latter, a well-adjusted stiffness ensures that the biomaterial's deformation behavior fits well to the deformation behavior of the surrounding biological tissue, whereas an appropriate strength provides sufficient load-carrying capacity of the biomaterial. Here, a mathematical modeling approach for estimating the macroscopic load that initiates failure of a hierarchically organized, granular, hydroxyapatite-based biomaterial is presented. For this purpose, a micromechanics model is developed for downscaling macroscopically prescribed stress (or strain) states to the level of the needle-shaped hydroxyapatite crystals. Presuming that the biomaterial fails due to the quasi-brittle failure of the most unfavorably stressed hydroxyapatite needle, the downscaled stress tensors are fed into a suitable, Mohr-Coulomb-type failure criterion, based on which the macroscopic failure load is deduced. The change of the biomaterial's composition in response to placing it in physiological solution, caused by growth of new bone tissue on the granules's surfaces, on the one hand, and by resorption of the hydroxyapatite crystals, on the other hand, is taken into account by means of suitable evolution laws. Numerical studies show how the macroscopic load-carrying capacity of the biomaterial is influenced by its design parameters. The presented modeling approach could prove beneficial for the design process of the studied biomaterials (as well as similarly composed biomaterials), particularly in terms of optimizing its mechanical performance.

11.
Comput Med Imaging Graph ; 45: 36-46, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26253382

RESUMEN

In dentistry, clinical radiographs (also called X-ray images) reflect the intensity loss of an X-ray when being transmitted through the mandibular objects, and this loss is quantified in terms of grey values. While such images are standardly used for pathology detection by the experienced dentist, we here present a new method for getting more quantitative information out of such 2D radiographs, "extending" them into the third dimension. This "extension" requires consistent combination of X-ray physics (namely, X-ray intensity loss quantification along paths orthogonal to the panoramic clinical image and X-ray attenuation averaging for composite materials) with anatomically known upper and lower limits of vascular porosities in cortical and trabecular bone compartments. Correspondingly computed ranges of overall organ thicknesses are extremely narrow, suggesting adequate estimation of thickness characteristics from 2D radiographic panoramas used clinically, while predicted cortical and trabecular thickness ranges vary by ±8.47% and ±16.13%, respectively. The proposed method also identifies variations between thicknesses at similar anatomical locations left and right of the face's symmetry axis, and molar regions turn out to be thicker than those close to incisors. This paves the way to more detailed diagnostic activities, e.g. in combination with Finite Element simulations.


Asunto(s)
Absorciometría de Fotón/métodos , Densidad Ósea/fisiología , Imagenología Tridimensional/métodos , Mandíbula/diagnóstico por imagen , Mandíbula/fisiología , Radiografía Panorámica/métodos , Humanos , Interpretación de Imagen Radiográfica Asistida por Computador/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
Artículo en Inglés | MEDLINE | ID: mdl-22563708

RESUMEN

Extracellular bone material can be characterised as a nanocomposite where, in a liquid environment, nanometre-sized hydroxyapatite crystals precipitate within as well as between long fibre-like collagen fibrils (with diameters in the 100 nm range), as evidenced from neutron diffraction and transmission electron microscopy. Accordingly, these crystals are referred to as 'interfibrillar mineral' and 'extrafibrillar mineral', respectively. From a topological viewpoint, it is probable that the mineralisations start on the surfaces of the collagen fibrils ('mineral-encrusted fibrils'), from where the crystals grow both into the fibril and into the extrafibrillar space. Since the mineral concentration depends on the pore spaces within the fibrils and between the fibrils (there is more space between them), the majority of the crystals (but clearly not all of them) typically lie in the extrafibrillar space. There, larger crystal agglomerations or clusters, spanning tens to hundreds of nanometers, develop in the course of mineralisation, and the micromechanics community has identified the pivotal role, which this extrafibrillar mineral plays for tissue elasticity. In such extrafibrillar crystal agglomerates, single crystals are stuck together, their surfaces being covered with very thin water layers. Recently, the latter have caught our interest regarding strength properties (Fritsch et al. 2009 J Theor Biol. 260(2): 230-252) - we have identified these water layers as weak interfaces in the extrafibrillar mineral of bone. Rate-independent gliding effects of crystals along the aforementioned interfaces, once an elastic threshold is surpassed, can be related to overall elastoplastic material behaviour of the hierarchical material 'bone'. Extending this idea, the present paper is devoted to viscous gliding along these interfaces, expressing itself, at the macroscale, in the well-known experimentally evidenced phenomenon of bone viscoelasticity. In this context, a multiscale homogenisation scheme is extended to viscoelasticity, mineral-cluster-specific creep parameters are identified from three-point bending tests on hydrated bone samples, and the model is validated by statistically and physically independent experiments on partially dried samples. We expect this model to be relevant when it comes to prediction of time-dependent phenomena, e.g. in the context of bone remodelling.


Asunto(s)
Huesos/fisiología , Modelos Biológicos , Agua/fisiología , Huesos/química , Durapatita/química , Elasticidad , Viscosidad
13.
Biomaterials ; 30(12): 2411-9, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19135717

RESUMEN

Synchrotron radiation micro-computed tomography (SRmuCT) revealed the microstructure of a CEL2 glass-ceramic scaffold with macropores of several hundred microns characteristic length, in terms of the voxel-by-voxel 3D distribution of the attenuation coefficients throughout the scanned space. The probability density function of all attenuation coefficients related to the macroporous space inside the scaffold gives access to the tomograph-specific machine error included in the SRmuCT measurements (also referred to as instrumental resolution function). After Lorentz function-based clearing of the measured CT data from the systematic resolution error, the voxel-specific attenuation information of the voxels representing the solid skeleton is translated into the composition of the material inside one voxel, in terms of the nanoporosity embedded in a dense CEL2 glass-ceramic matrix. Based on voxel-invariant elastic properties of dense CEL2 glass-ceramic, continuum micromechanics allows for translation of the voxel-specific nanoporosity into voxel-specific elastic properties. They serve as input for Finite Element analyses of the scaffold structure. Young's modulus of a specific CT-scanned macroporous scaffold sample, predicted from a Finite Element simulation of a uniaxial compression test, agrees well with the experimental value obtained from an ultrasonic test on the same sample. This highlights the satisfactory predictive capabilities of the presented approach.


Asunto(s)
Huesos/diagnóstico por imagen , Tomografía Computarizada por Rayos X/métodos , Fenómenos Biomecánicos , Elasticidad , Modelos Biológicos , Nanoestructuras , Porosidad , Ingeniería de Tejidos , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...