Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Eur Cell Mater ; 22: 202-13, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-22048898

ABSTRACT

A eukaryotic cell attaches and spreads on substrates, whether it is the extracellular matrix naturally produced by the cell itself, or artificial materials, such as tissue-engineered scaffolds. Attachment and spreading require the cell to apply forces in the nN range to the substrate via adhesion sites, and these forces are balanced by the elastic response of the substrate. This mechanical interaction is one determinant of cell morphology and, ultimately, cell phenotype. In this paper we use a finite element model of a cell, with a tensegrity structure to model the cytoskeleton of actin filaments and microtubules, to explore the way cells sense the stiffness of the substrate and thereby adapt to it. To support the computational results, an analytical 1D model is developed for comparison. We find that (i) the tensegrity hypothesis of the cytoskeleton is sufficient to explain the matrix-elasticity sensing, (ii) cell sensitivity is not constant but has a bell-shaped distribution over the physiological matrix-elasticity range, and (iii) the position of the sensitivity peak over the matrix-elasticity range depends on the cytoskeletal structure and in particular on the F-actin organisation. Our model suggests that F-actin reorganisation observed in mesenchymal stem cells (MSCs) in response to change of matrix elasticity is a structural-remodelling process that shifts the sensitivity peak towards the new value of matrix elasticity. This finding discloses a potential regulatory role of scaffold stiffness for cell differentiation.


Subject(s)
Cell Adhesion/physiology , Elasticity , Tissue Scaffolds , Actin Cytoskeleton/physiology , Actins , Cell Movement , Extracellular Matrix/physiology , Finite Element Analysis , Mesenchymal Stem Cells/physiology , Microtubules/physiology , Models, Biological
2.
J Mech Behav Biomed Mater ; 4(8): 1880-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22098887

ABSTRACT

Computational modelling is becoming ever more important for obtaining regulatory approval for new medical devices. An accepted approach is to infer performance in a population from an analysis conducted for an idealised or 'average' patient; we present here a method for predicting the performance of an orthopaedic implant when released into a population--effectively simulating a clinical trial. Specifically we hypothesise that an analysis based on a method for predicting the performance in a population will lead to different conclusions than an analysis based on an idealised or 'average' patient. To test this hypothesis we use a finite element model of an intramedullary implant in a bone whose size and remodelling activity is different for each individual in the population. We compare the performance of a low Young's modulus implant (E=20 GPa) to one with a higher Young's modulus (200 GPa). Cyclic loading is applied and failure is assumed when the migration of the implant relative to the bone exceeds a threshold magnitude. The analysis for an idealised of 'average' patient predicts that the lower modulus device survives longer whereas the analysis simulating a clinical trial predicts no statistically-significant tendency (p=0.77) for the low modulus device to perform better. It is concluded that population-based simulations of implant performance-simulating a clinical trial-present a very valuable opportunity for more realistic computational pre-clinical testing of medical devices.


Subject(s)
Bone and Bones , Clinical Trials as Topic , Computer Simulation , Prostheses and Implants , Weight-Bearing , Biomechanical Phenomena , Bone Remodeling , Bone and Bones/cytology , Bone and Bones/physiopathology , Finite Element Analysis , Prosthesis Failure
3.
Med Eng Phys ; 32(10): 1180-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20933453

ABSTRACT

Three-dimensional reconstruction from volumetric medical images (e.g. CT, MRI) is a well-established technology used in patient-specific modelling. However, there are many cases where only 2D (planar) images may be available, e.g. if radiation dose must be limited or if retrospective data is being used from periods when 3D data was not available. This study aims to address such cases by proposing an automated method to create 3D surface models from planar radiographs. The method consists of (i) contour extraction from the radiograph using an Active Contour (Snake) algorithm, (ii) selection of a closest matching 3D model from a library of generic models, and (iii) warping the selected generic model to improve correlation with the extracted contour. This method proved to be fully automated, rapid and robust on a given set of radiographs. Measured mean surface distance error values were low when comparing models reconstructed from matching pairs of CT scans and planar X-rays (2.57-3.74mm) and within ranges of similar studies. Benefits of the method are that it requires a single radiographic image to perform the surface reconstruction task and it is fully automated. Mechanical simulations of loaded bone with different levels of reconstruction accuracy showed that an error in predicted strain fields grows proportionally to the error level in geometric precision. In conclusion, models generated by the proposed technique are deemed acceptable to perform realistic patient-specific simulations when 3D data sources are unavailable.


Subject(s)
Femur/diagnostic imaging , Imaging, Three-Dimensional/methods , Models, Biological , Pattern Recognition, Automated/methods , Radiographic Image Interpretation, Computer-Assisted/methods , Femur Head/diagnostic imaging , Femur Neck/diagnostic imaging , Finite Element Analysis , Humans , Reproducibility of Results , Sensitivity and Specificity , X-Rays
4.
Philos Trans A Math Phys Eng Sci ; 368(1921): 2919-35, 2010 Jun 28.
Article in English | MEDLINE | ID: mdl-20478914

ABSTRACT

The design of medical devices could be very much improved if robust tools were available for computational simulation of tissue response to the presence of the implant. Such tools require algorithms to simulate the response of tissues to mechanical and chemical stimuli. Available methodologies include those based on the principle of mechanical homeostasis, those which use continuum models to simulate biological constituents, and the cell-centred approach, which models cells as autonomous agents. In the latter approach, cell behaviour is governed by rules based on the state of the local environment around the cell; and informed by experiment. Tissue growth and differentiation requires simulating many of these cells together. In this paper, the methodology and applications of cell-centred techniques--with particular application to mechanobiology--are reviewed, and a cell-centred model of tissue formation in the lumen of an artery in response to the deployment of a stent is presented. The method is capable of capturing some of the most important aspects of restenosis, including nonlinear lesion growth with time. The approach taken in this paper provides a framework for simulating restenosis; the next step will be to couple it with more patient-specific geometries and quantitative parameter data.


Subject(s)
Computer Simulation , Models, Biological , Stents , Tunica Intima/cytology , Biomechanical Phenomena , Extracellular Space/metabolism , Finite Element Analysis , Humans , Myocytes, Smooth Muscle/cytology
5.
Ann Biomed Eng ; 38(5): 1767-79, 2010 May.
Article in English | MEDLINE | ID: mdl-20217480

ABSTRACT

A role for mechanical stimulation in the control of cell fate has been proposed and mechanical conditioning of mesenchymal stem cells (MSCs) is of interest in directing MSC behavior for tissue engineering applications. This study investigates strain-induced differentiation and proliferation of MSCs, and investigates the cellular mechanisms of mechanotransduction. MSCs were seeded onto a collagen-coated silicone substrate and exposed to cyclic tensile mechanical strain of 2.5% at 0.17 Hz for 1-14 days. To examine mechanotransduction, cells were strained in the presence of the stretch-activated cation channel (SACC) blocker, gadolinium chloride (GdCl(3)); the extracellular regulated kinase (ERK) inhibitor, U0126; the p38 inhibitor, SB203580; and the phosphatidylinosito1 3-kinase (PI3-kinase) inhibitor, LY294002. Following exposure to strain, the osteogenic markers Cbfalpha1, collagen type I, osteocalcin, and BMP2 were temporally expressed. Exposure to strain in the presence of GdCl(3) (10 microM) reduced the induction of collagen I expression, thus identifying a role for SACC, at least in part, as mechanosensors in strain-induced MSC differentiation. The strain-induced synthesis of BMP2 was found to be reduced by inhibitors of the kinases, ERK, p38, and PI3 kinase. Additionally, mechanical strain reduced the rate of MSC proliferation. The identification of the mechanical control of MSC proliferation and the molecular link between mechanical stimulation and osteogenic differentiation has consequences for regenerative medicine through the development of a functional tissue engineering approach.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Tissue Engineering/methods , Animals , Chromones/pharmacology , Collagen/metabolism , Collagen Type I/metabolism , Extracellular Matrix/metabolism , Male , Mesenchymal Stem Cells/metabolism , Morpholines/pharmacology , Osteocalcin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Wistar
6.
J Biomech Eng ; 130(5): 051015, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19045522

ABSTRACT

The geometry of an implant surface to best promote osseointegration has been the subject of several experimental studies, with porous beads and woven mesh surfaces being among the options available. Furthermore, it is unlikely that one surface geometry is optimal for all loading conditions. In this paper, a computational method is used to simulate tissue differentiation and osseointegration on a smooth surface, a surface covered with sintered beads (this simulated the experiment (Simmons, C., and Pilliar, R., 2000, Biomechanical Study of Early Tissue Formation Around Bone-Interface Implants: The Effects of Implant Surface Geometry," Bone Engineering, J. E. Davies, ed., Emsquared, Chap. A, pp. 369-379) and established that the method gives realistic results) and a surface covered by porous tantalum. The computational method assumes differentiation of mesenchymal stem cells in response to fluid flow and shear strain and models cell migration and proliferation as continuum processes. The results of the simulation show a higher rate of bone ingrowth into the surfaces with porous coatings as compared with the smooth surface. It is also shown that a thicker interface does not increase the chance of fixation failure.


Subject(s)
Bone and Bones/cytology , Bone and Bones/physiology , Cell Differentiation/physiology , Models, Biological , Osteoblasts/physiology , Osteogenesis/physiology , Prostheses and Implants , Animals , Computer Simulation , Humans , Surface Properties
7.
J Biomech Eng ; 130(6): 061001, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19045530

ABSTRACT

Tissue engineering often involves seeding cells into porous scaffolds and subjecting the scaffold to mechanical stimulation. Current experimental techniques have provided a plethora of data regarding cell responses within scaffolds, but the quantitative understanding of the load transfer process within a cell-seeded scaffold is still relatively unknown. The objective of this work was to develop a finite element representation of the transient and heterogeneous nature of a cell-seeded collagen-GAG-scaffold. By undertaking experimental investigation, characteristics such as scaffold architecture and shrinkage, cellular attachment patterns, and cellular dimensions were used to create a finite element model of a cell-seeded porous scaffold. The results demonstrate that a very wide range of microscopic strains act at the cellular level when a sample value of macroscopic (apparent) strain is applied to the collagen-GAG-scaffold. An external uniaxial strain of 10% generated a cellular strain as high as 49%, although the majority experienced less than approximately 5% strain. The finding that the strain on some cells could be higher than the macroscopic strain was unexpected and proves contrary to previous in vitro investigations. These findings indicate a complex system of biophysical stimuli created within the scaffolds and the difficulty of inducing the desired cellular responses from artificial environments. Future in vitro studies could also corroborate the results from this computational prediction to further explore mechanoregulatory mechanisms in tissue engineering.


Subject(s)
Extracellular Matrix/chemistry , Mechanotransduction, Cellular/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Models, Biological , Tissue Engineering/methods , Animals , Cell Culture Techniques/methods , Cells, Cultured , Computer Simulation , Elastic Modulus/physiology , Finite Element Analysis , Male , Physical Stimulation/methods , Porosity , Rats , Rats, Wistar , Shear Strength/physiology , Stem Cells , Stress, Mechanical
8.
J Biomech Eng ; 130(6): 061004, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19045533

ABSTRACT

Mechanical conditioning of mesenchymal stem cells (MSCs) has been adopted widely as a biophysical signal to aid tissue engineering applications. The replication of in vivo mechanical signaling has been used in in vitro environments to regulate cell differentiation, and extracellular matrix synthesis, so that both the chemical and mechanical properties of the tissue-engineered construct are compatible with the implant site. While research in these areas contributes to tissue engineering, the effects of mechanical strain on MSC apoptosis remain poorly defined. To evaluate the effects of uniaxial cyclic tensile strain on MSC apoptosis and to investigate mechanotransduction associated with strain-mediated cell death, MSCs seeded on a 2D silicone membrane were stimulated by a range of strain magnitudes for 3 days. Mechanotransduction was investigated using the stretch-activated cation channel blocker gadolinium chloride, the L-type voltage-activated calcium channel blocker nicardipine, the c-jun NH(2)-terminal kinase (JNK) blocker D-JNK inhibitor 1, and the calpain inhibitor MDL 28170. Apoptosis was assessed through DNA fragmentation using the terminal deoxynucleotidyl transferase mediated-UTP-end nick labeling method. Results demonstrated that tensile strains of 7.5% or greater induce apoptosis in MSCs. L-type voltage-activated calcium channels coupled mechanical stress to activation of calpain and JNK, which lead to apoptosis through DNA fragmentation. The definition of the in vitro boundary conditions for tensile strain and MSCs along with a proposed mechanism for apoptosis induced by mechanical events positively contributes to the development of MSC biology, bioreactor design for tissue engineering, and development of computational methods for mechanobiology.


Subject(s)
Apoptosis/physiology , Calcium Channels/metabolism , Calpain/metabolism , MAP Kinase Kinase 4/metabolism , Mechanotransduction, Cellular/physiology , Mesenchymal Stem Cells/physiology , Animals , Cells, Cultured , Elastic Modulus/physiology , Mesenchymal Stem Cells/cytology , Physical Stimulation/methods , Rats , Rats, Wistar , Stress, Mechanical
9.
Regen Med ; 3(5): 743-59, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18729798

ABSTRACT

Osteochondral tissues are those that form the synovial joints, namely cartilage and bone, including sub-chondral bone. The biomechanical purpose of synovial joints is to provide lubricated contact between the moving surfaces with as little frictional forces as possible. This is achieved by separating the cartilage layers by a thin film of fluid and supporting the cartilage layers by a bony trabecular network that becomes dense and more calcified immediately underneath the cartilage layer. Each tissue's biomechanical behavior is well understood after several decades of research and this behavior is briefly reviewed here, as are the concepts relating to the mechanical induction of cartilage degradation (osteoarthritis) with a discussion of clinical strategies for repair. Focusing on tissue-engineering strategies, the following concepts are reviewed: scaffolds, bioreactors and computational simulations, with an analysis of how these elements may be combined in future.


Subject(s)
Biomechanical Phenomena , Cartilage, Articular/cytology , Chondrocytes/cytology , Stem Cells/cytology , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Bioreactors , Cartilage, Articular/pathology , Cell Lineage , Collagen/chemistry , Computer Simulation , Humans , Knee Joint , Osteoarthritis/pathology , Regenerative Medicine/methods
10.
Ir J Med Sci ; 177(4): 289-96, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18641919

ABSTRACT

This paper presents the 14th Samuel Haughton lecture delivered on the 26th of January 2008. The lecture began by describing Haughton's research on animal mechanics. Haughton opposed Charles Darwin's theory of natural selection using the argument that the skeleton obeys the 'principle of least action' and therefore must have been designed with that principle in mind. In the course of his research he dissected many animals, including albatrosses, cassowaries, llamas, tigers, jackals and jaguars. He took anatomical measurements and did calculations to prove that muscle attachment sites were optimally located. The relationship between optimality and evolution continues to be studied. Computer simulations show optimality is difficult to achieve. This is because, even if optimality could be defined, the gene recombinations required to evolve an optimal phenotype may not exist. The drive towards optimality occurs under gravitational forces. Simulations to predict mechano-regulation of tissue differentiation and remodelling have been developed and tested. They have been used to design biomechanically optimized scaffolds for regenerative medicine and to identify the mechanoregularory mechanisms in osteoporosis. It is proposed that an important development in bioengineering will be the discovery of algorithms that can be used for the prediction of mechano-responsiveness in biological tissues.


Subject(s)
Osteoporosis/therapy , Stem Cells , Tissue Scaffolds , Bone Remodeling , Computational Biology , Humans , Models, Theoretical , Musculoskeletal System , Stress, Mechanical , Tissue Engineering
11.
Arch Orthop Trauma Surg ; 128(10): 1193-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-17940780

ABSTRACT

INTRODUCTION: The number of revision hip arthroplasties is increasing but several aspects of this procedure could be improved. One method of reducing intra-operative complications is the cement-in-cement technique. This procedure entails cementing a smaller femoral prosthesis into the existing stable cement mantle. The aim of this systematic review is to provide a concise overview of the existing historical, operative, biomechanical and clinical literature on the cement-in-cement construct. RESULTS: Four biomechanical publications exist in authoritative journals and these were reviewed. Simple specimens were produced and these were tested by static means. Although these published tests support the cement-in-cement technique, they cannot be regarded as conclusive. Areas which could be subject to further research are identified. Five clinical publications on patients undergoing cement-in-cement revisions were also reviewed. Patient numbers were generally low (7-53) apart from one study containing 354 patients. Long-term patient follow-up was not available except in Hubble's study (41 patients followed for 8 years). Outcomes of these patients were very satisfactory for the period of follow-up. Three expert reviews of cemented femoral revisions outline the cement in cement procedure. If other Orthopaedic Centres can emulate the results of the clinical research presented, complication rates, operative times and financial costs may be decreased. CONCLUSION: The analysis presented in this paper consolidates the latest biomechanical and clinical information on cement-in-cement revision hip arthroplasty. Although we find evidence to support the use of the method clinically, we do note that the scientific basis needs further investigation.


Subject(s)
Arthroplasty, Replacement, Hip/methods , Cementation/methods , Biomechanical Phenomena , Bone Cements , Humans , Reoperation
12.
Biomech Model Mechanobiol ; 7(6): 443-61, 2008 Dec.
Article in English | MEDLINE | ID: mdl-17972123

ABSTRACT

Modelling the course of healing of a long bone subjected to loading has been the subject of several investigations. These have succeeded in predicting the differentiation of tissues in the callus in response to a static mechanical load and the diffusion of biological factors. In this paper an approach is presented which includes both mechanoregulation of tissue differentiation and the diffusion and proliferation of cell populations (mesenchymal stem cells, fibroblasts, chondrocytes, and osteoblasts). This is achieved in a three-dimensional poroelastic finite element model which, being poroelastic, can model the effect of the frequency of dynamic loading. Given the number of parameters involved in the simulation, a parameter variation study is reported, and final parameters are selected based on comparison with an in vivo experiment. The model predicts that asymmetric loading creates an asymmetric distribution of tissues in the callus, but only for high bending moments. Furthermore the frequency of loading is predicted to have an effect. In conclusion, a numerical algorithm is presented incorporating both mechanoregulation and evolution of cell populations, and it proves capable of predicting realistic difference in bone healing in a 3D fracture callus.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Fracture Healing/physiology , Fractures, Bone/physiopathology , Mechanotransduction, Cellular/physiology , Animals , Bone Regeneration/physiology , Bony Callus/physiopathology , Calibration , Computer Simulation , Finite Element Analysis , Reproducibility of Results , Sheep
13.
Med Biol Eng Comput ; 46(3): 283-98, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17899238

ABSTRACT

Mandibular symphyseal distraction osteogenesis is a common clinical procedure to modify the geometrical shape of the mandible for correcting problems of dental overcrowding and arch shrinkage. In spite of consolidated clinical use, questions remain concerning the optimal latency period and the influence of mastication loading on osteogenesis within the callus prior to the first distraction of the mandible. This work utilized a mechano-regulation model to assess bone regeneration within the callus of an osteotomized mandible. A 3D model of the mandible was reconstructed from CT scan data and meshed using poroelastic finite elements (FE). The stimulus regulating tissue differentiation within the callus was hypothesized to be a function of the strain and fluid flow computed by the FE model. This model was then used to analyse tissue differentiation during a 15-day latency period, defined as the time between the day of the osteotomy and the day when the first distraction is given to the device. The following predictions are made: (1) the mastication forces generated during the latency period support osteogenesis in certain regions of the callus, and that during the latency period the percentage of progenitor cells differentiating into osteoblasts increases; (2) reducing the mastication load by 70% during the latency period increases the number of progenitor cells differentiating into osteoblasts; (3) the stiffness of new tissue increases at a slower rate on the side of bone callus next to the occlusion of the mandibular ramus which could cause asymmetries in the bone tissue formation with respect to the middle sagittal plane. Although the model predicts that the mastication loading generates such asymmetries, their effects on the spatial distribution of callus mechanical properties are insignificant for typical latency periods used clinically. It is also predicted that a latency period of longer than a week will increase the risk of premature bone union across the callus.


Subject(s)
Bone Regeneration , Mandible/surgery , Osteogenesis, Distraction , Algorithms , Finite Element Analysis , Humans , Mandible/diagnostic imaging , Mandible/physiopathology , Mastication , Mechanotransduction, Cellular , Osteotomy , Tomography, X-Ray Computed
14.
Technol Health Care ; 15(1): 19-31, 2007.
Article in English | MEDLINE | ID: mdl-17264410

ABSTRACT

Adult mesenchymal stem cells (MSCs) have the capability to differentiate along several lineages including those of bone, cartilage, tendon and muscle, thus offering huge potential for the field of tissue engineering. The purpose of this study was to characterise the differentiation capacity of rat MSCs cultured on standard plastic coverslips in 2 dimensions and on a novel collagen glycosaminoglycan scaffold in the presence of a standard combination of osteoinductive factors. Cells were cultured for 3, 7, 14 and 21 days and several markers of osteogenesis were analysed. While the initial response of the cells in 3-D seemed to be faster than cells cultured in 2-D, as evidenced by collagen type I expression, later markers showed that osteogenic differentiation of MSCs took longer in the 3-D environment of the collagen GAG scaffold compared to standard 2-D culture conditions. Furthermore, it was shown that complete scaffold mineralisation could be evoked within a 6 week timeframe. This study further demonstrates the potential use of MSC-seeded collagen GAG scaffolds for bone tissue engineering applications.


Subject(s)
Cell Culture Techniques , Cell-Matrix Junctions/chemistry , Collagen/chemistry , Mesenchymal Stem Cells , Osteogenesis/physiology , Tissue Engineering/methods , Animals , Cell Differentiation , Glycosaminoglycans , Osteocalcin/biosynthesis , RNA, Messenger , Rats , Rats, Wistar
15.
Cell Tissue Res ; 327(2): 293-300, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17024413

ABSTRACT

The interface between any newly engineered tissue and pre-existing tissue is of great importance to tissue engineering; however, this process has so far been largely ignored, with few reports regarding the mechanical strength of newly integrated connective tissues surfaces. A new model system has been developed to generate a well-defined interface between two collagen lattices: one pre-contracted by resident fibroblasts and the other a cell-free wrapping gel. This construct can be cultured for prolonged periods (>2 weeks) and can also be fitted onto a mechanical testing system to measure the interface adhesive strength at the end of the culture time. Interface adhesive strength shows a six-fold increase after 1 week in culture, compared with the time-zero baseline. Observations of cell migration across the interface suggest that cell translocation in the three-dimensional matrix might play an important role in the integration process. In this new controlled geometry, normal and shear stresses at the interface can be analysed by finite element modelling and the areas at which debonding starts can be defined. The current experimental design permits solid multiple (homogeneous or heterogeneous) interface formation in vitro with a well-defined geometry and the possibility of measuring mechanical linkage. This design should enable many other factors affecting cell-driven interface strengthening to be investigated.


Subject(s)
Collagen/chemistry , Tissue Engineering/methods , Animals , Biomechanical Phenomena , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Movement , Cells, Cultured , Connective Tissue/chemistry , Fibroblasts/cytology , Fibroblasts/physiology , Finite Element Analysis , Gels/chemistry , Materials Testing , Rats , Stress, Mechanical , Tissue Engineering/instrumentation
16.
J Biomech ; 40(10): 2244-53, 2007.
Article in English | MEDLINE | ID: mdl-17173925

ABSTRACT

Computational models have shown that biophysical stimuli can be correlated with observed patterns of tissue differentiation, and simulations have been performed that predict the time course of tissue differentiation in, for example, long bone fracture healing. Some simulations have used a diffusion model to simulate the migration and proliferation of cells with the differentiating tissue. However, despite the convenience of the diffusion model, diffusion is not the mechanism of cell dispersal: cells disperse by crawling or proliferation, or are transported in a moving fluid. In this paper, a random-walk model (i.e., a stochastic model), with and without a preferred direction, is studied as an approach to simulate cell proliferation/migration in differentiating tissues and it is compared with the diffusion model. A simulation of tissue differentiation of gap tissue in a two-dimensional model of a bone/implant interface was performed to demonstrate the differences between diffusion vs. random walk with a preferred direction. Results of diffusion and random-walk models are similar with respect to the change in the stiffness of the gap tissue but rather different results are obtained regarding tissue patterning in the differentiating tissues; the diffusion approach predicted continuous patterns of tissue differentiation whereas the random-walk model showed a more discontinuous pattern-histological results are not available that can unequivocally establish which is most similar to experimental observation. Comparing isotropic to anisotropic random walk (preferred direction of proliferation and cell migration), a more rapid reduction of the relative displacement between implant and bone is predicted. In conclusion, we have shown how random-walk models of cell dispersal and proliferation can be implemented, and shown where differences between them exist. Further study of the random-walk model is warranted, given the importance of cell seeding and cell dispersal/proliferation in many mechanobiological problems.


Subject(s)
Bone and Bones/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Mechanotransduction, Cellular/physiology , Mitosis/physiology , Models, Biological , Animals , Bone and Bones/cytology , Computer Simulation , Humans , Prostheses and Implants
17.
HFSP J ; 1(3): 181-91, 2007 Sep.
Article in English | MEDLINE | ID: mdl-19404445

ABSTRACT

During numerous biological processes, cell adhesion, cell migration and cell spreading are vital. These basic biological functions are regulated by the interaction of cells with their extracellular environment. To examine the morphology and mechanical changes occurring in mesenchymal stem cells cultured on a mechanically rigid substrate, atomic force microscopy and fluorescence microscopy were employed. Investigations of the cells revealed both linear and geodesic F-actin configurations. No particular cell characteristics or intra-cellular location were implicated in the appearance of the geodesic structures. However, the length of time the cells were cultured on the substrate correlated with the percentage appearance of the geodesic structures. Calculating energy dissipation from cell images acquired by dynamic mode atomic force microscopy, it was observed that the vertices of the geodesic structures had significantly higher energy dissipation compared to the linear F-actin and the glass. This supports work by Lazarides [J. Cell Biol. 68, 202-219 (1976)], who postulated that the vertices of these geodesic structures should have a greater flexibility. Our results also support predictions based on the microfilament tensegrity model. By understanding the basic principles of cell ultrastructure and cell mechanics in relation to different extracellular environments, a better understanding of physiological and pathological process will be elicited.

18.
Ir J Med Sci ; 175(3): 69-75, 2006.
Article in English | MEDLINE | ID: mdl-17073253

ABSTRACT

Biomechanics is a subject that draws on knowledge from many disciplines. One of its great practitioners in the last century was the Irish anatomist M.A. MacConaill. In this paper, we review some of MacConaill's fundamental contributions to biomechanics, namely: the hydrodynamic theory of synovial joint lubrication, the kinematics of joint motion and conjunct rotations; and the theory of spurt and shunt muscles. The aim is to revisit these topics in the light of current research, and to draw some conclusions about the import of his research in the context of recent developments in the field. The paper concludes with a discussion of science in Ireland, the development of the field of biomechanics since MacConaill's time, and some other matters.


Subject(s)
Biomechanical Phenomena/history , Kinesiology, Applied/history , History, 20th Century , Ireland , Joints/physiology , Muscles/physiology
19.
Bone ; 39(2): 392-400, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16644297

ABSTRACT

Hormone therapy (HT) drugs and bisphosphonates prevent osteoporosis by inhibiting osteoclastic bone resorption. However, the effects of osteoporosis and anti-resorptive drugs on the mechanical behavior of the bone tissue constituting individual trabeculae have not yet been quantified. In this study, we test the hypothesis that the mechanical properties of bone trabecular tissue will differ for normal, ovariectomized and drug-treated rat bones over the course of ageing. Microtensile testing is carried on individual trabeculae from tibial bone of ovariectomized (OVX) rats, OVX rats treated with tibolone and placebo-treated controls. The method developed minimizes errors due to misalignment and stress concentrations at the grips. The local mineralization of single trabeculae is compared using micro-CT images calibrated for bone mineral content assessment. Our results indicate that ovariectomy in rats increases the stiffness, yield strength, yield strain and ultimate stress of the mineralized tissue constituting trabecular bone relative to normal; we found significant differences (P < 0.05) at 14, 34 and 54 weeks of treatment. These increases are complemented by a significant increase in the mineral content at the tissue level, although overall bone mineral density and mass are reduced. With drug treatment, the properties remain at, or slightly below, the placebo-treated controls levels for 54 weeks. The higher bone strength in the OVX group may cause the trabecular architecture to adapt as seen during osteopenia/osteoporosis, or alternately it may compensate for loss of trabecular architecture. These findings suggest that, in addition to the effects of osteoporosis and subsequent treatment on bone architecture, there are also more subtle processes ongoing to alter bone strength at the tissue level.


Subject(s)
Aging/physiology , Estrogen Receptor Modulators/pharmacology , Norpregnenes/pharmacology , Ovariectomy , Tibia/drug effects , Tibia/physiology , Animals , Biomechanical Phenomena , Bone Density/drug effects , Female , Finite Element Analysis , Rats , Rats, Wistar , Stress, Mechanical , Tensile Strength , Tomography, X-Ray Computed , Weight-Bearing
20.
J Biomech ; 39(4): 734-41, 2006.
Article in English | MEDLINE | ID: mdl-16439243

ABSTRACT

Analyses of the distributions of stress and strain within individual bone trabeculae have not yet been reported. In this study, four trabeculae were imaged and finite elements models were generated in an attempt to quantify the variability of stress/strain in real trabeculae. In three of these trabeculae, cavities were identified with depths comparable to values reported for resorption lacunae ( approximately 50 microm)-although we cannot be certain, it is most probable that they are indeed resorption lacunae. A tensile load was applied to each trabeculum to simulate physiological loading and to ensure that bending was minimized. The force carried by each trabecula was calculated from this value using the average cross sectional area of each trabecula. The analyses predict that very high stresses (>100 MPa) existed within bone trabecular tissue. Stress and strain distributions were highly heterogeneous in all cases, more so in trabeculae with the presumptive resorption lacunae where at least 30% of the tissue had a strain greater than 4000 micoepsilon in all cases. Stresses were elevated at the pit of the lacunae, and peak stress concentrations were located in the longitudinal direction ahead of the lacunae. Given these high strains, we suggest that microdamage is inevitable around resorption lacunae in trabecular bone, and may cause the bone multicellular unit to proceed to resorb a packet of bone in the trabeculum rather than just resorb whatever localized area was initially targeted.


Subject(s)
Bone Resorption/physiopathology , Models, Biological , Tibia/physiopathology , Weight-Bearing , Animals , Compressive Strength , Computer Simulation , Elasticity , In Vitro Techniques , Radiography , Rats , Rats, Wistar , Stress, Mechanical , Tibia/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...