Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Bull Math Biol ; 84(8): 85, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35802265

ABSTRACT

We analyse mathematical models in order to understand how microstructural features of vascular networks may affect blood flow dynamics, and to identify particular characteristics that promote the onset of self-sustained oscillations. By focusing on a simple three-node motif, we predict that network "redundancy", in the form of a redundant vessel connecting two main flow-branches, together with differences in haemodynamic resistance in the branches, can promote the emergence of oscillatory dynamics. We use existing mathematical descriptions for blood rheology and haematocrit splitting at vessel branch-points to construct our flow model; we combine numerical simulations and stability analysis to study the dynamics of the three-node network and its relation to the system's multiple steady-state solutions. While, for the case of equal inlet-pressure conditions, a "trivial" equilibrium solution with no flow in the redundant vessel always exists, we find that it is not stable when other, stable, steady-state attractors exist. In turn, these "nontrivial" steady-state solutions may undergo a Hopf bifurcation into an oscillatory state. We use the branch diameter ratio, together with the inlet haematocrit rate, to construct a two-parameter stability diagram that delineates regimes in which such oscillatory dynamics exist. We show that flow oscillations in this network geometry are only possible when the branch diameters are sufficiently different to allow for a sufficiently large flow in the redundant vessel, which acts as the driving force of the oscillations. These microstructural properties, which were found to promote oscillatory dynamics, could be used to explore sources of flow instability in biological microvascular networks.


Subject(s)
Mathematical Concepts , Models, Biological , Hemodynamics , Microvessels/physiology , Models, Theoretical
2.
Biomech Model Mechanobiol ; 20(2): 585-608, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33219879

ABSTRACT

We present a mechanical model of tissue homeostasis that is specialised to the intestinal crypt. Growth and deformation of the crypt, idealised as a line of cells on a substrate, are modelled using morphoelastic rod theory. Alternating between Lagrangian and Eulerian mechanical descriptions enables us to precisely characterise the dynamic nature of tissue homeostasis, whereby the proliferative structure and morphology are static in the Eulerian frame, but there is active migration of Lagrangian material points out of the crypt. Assuming mechanochemical growth, we identify the necessary conditions for homeostasis, reducing the full, time-dependent system to a static boundary value problem characterising a spatially heterogeneous "treadmilling" state. We extract essential features of crypt homeostasis, such as the morphology, the proliferative structure, the migration velocity, and the sloughing rate. We also derive closed-form solutions for growth and sloughing dynamics in homeostasis, and show that mechanochemical growth is sufficient to generate the observed proliferative structure of the crypt. Key to this is the concept of threshold-dependent mechanical feedback, that regulates an established Wnt signal for biochemical growth. Numerical solutions demonstrate the importance of crypt morphology on homeostatic growth, migration, and sloughing, and highlight the value of this framework as a foundation for studying the role of mechanics in homeostasis.


Subject(s)
Homeostasis , Intestines/growth & development , Intestines/physiology , Animals , Biomechanical Phenomena , Humans , Models, Biological
3.
Proc Math Phys Eng Sci ; 476(2243): 20200846, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33363445

ABSTRACT

[This corrects the article DOI: 10.1098/rspa.2020.0355.].

4.
Proc Math Phys Eng Sci ; 476(2242): 20200355, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33223936

ABSTRACT

Tides are a major component of the interaction between the marine and terrestrial environments, and thus play an important part in shaping the environmental context for the evolution of shallow marine and coastal organisms. Here, we use a dedicated tidal model and palaeogeographic reconstructions from the Late Silurian to early Late Devonian (420 Ma, 400 Ma and 380 Ma, Ma = millions of years ago) to explore the potential significance of tides for the evolution of osteichthyans (bony fish) and tetrapods (land vertebrates). The earliest members of the osteichthyan crown-group date to the Late Silurian, approximately 425 Ma, while the earliest evidence for tetrapods is provided by trackways from the Middle Devonian, dated to approximately 393 Ma, and the oldest tetrapod body fossils are Late Devonian, approximately 373 Ma. Large tidal ranges could have fostered both the evolution of air-breathing organs in osteichthyans to facilitate breathing in oxygen-depleted tidal pools, and the development of weight-bearing tetrapod limbs to aid navigation within the intertidal zones. We find that tidal ranges over 4 m were present around areas of evolutionary significance for the origin of osteichthyans and the fish-tetrapod transition, highlighting the possible importance of tidal dynamics as a driver for these evolutionary processes.

5.
Eur J Appl Math ; 31(1): 143-171, 2020 Feb.
Article in English | MEDLINE | ID: mdl-33149377

ABSTRACT

Tissue engineering aims to grow artificial tissues in vitro to replace those in the body that have been damaged through age, trauma or disease. A recent approach to engineer artificial cartilage involves seeding cells within a scaffold consisting of an interconnected 3D-printed lattice of polymer fibres combined with a cast or printed hydrogel, and subjecting the construct (cell-seeded scaffold) to an applied load in a bioreactor. A key question is to understand how the applied load is distributed throughout the construct. To address this, we employ homogenisation theory to derive equations governing the effective macroscale material properties of a periodic, elastic-poroelastic composite. We treat the fibres as a linear elastic material and the hydrogel as a poroelastic material, and exploit the disparate length scales (small inter-fibre spacing compared with construct dimensions) to derive macroscale equations governing the response of the composite to an applied load. This homogenised description reflects the orthotropic nature of the composite. To validate the model, solutions from finite element simulations of the macroscale, homogenised equations are compared to experimental data describing the unconfined compression of the fibre-reinforced hydrogels. The model is used to derive the bulk mechanical properties of a cylindrical construct of the composite material for a range of fibre spacings and to determine the local mechanical environment experienced by cells embedded within the construct.

6.
J Theor Biol ; 436: 39-50, 2018 01 07.
Article in English | MEDLINE | ID: mdl-28970093

ABSTRACT

Immunotherapies exploit the immune system to target and kill cancer cells, while sparing healthy tissue. Antibody therapies, an important class of immunotherapies, involve the binding to specific antigens on the surface of the tumour cells of antibodies that activate natural killer (NK) cells to kill the tumour cells. Preclinical assessment of molecules that may cause antibody-dependent cellular cytotoxicity (ADCC) involves co-culturing cancer cells, NK cells and antibody in vitro for several hours and measuring subsequent levels of tumour cell lysis. Here we develop a mathematical model of such an in vitro ADCC assay, formulated as a system of time-dependent ordinary differential equations and in which NK cells kill cancer cells at a rate which depends on the amount of antibody bound to each cancer cell. Numerical simulations generated using experimentally-based parameter estimates reveal that the system evolves on two timescales: a fast timescale on which antibodies bind to receptors on the surface of the tumour cells, and NK cells form complexes with the cancer cells, and a longer time-scale on which the NK cells kill the cancer cells. We construct approximate model solutions on each timescale, and show that they are in good agreement with numerical simulations of the full system. Our results show how the processes involved in ADCC change as the initial concentration of antibody and NK-cancer cell ratio are varied. We use these results to explain what information about the tumour cell kill rate can be extracted from the cytotoxicity assays.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Models, Immunological , Cell Line, Tumor , Humans , Numerical Analysis, Computer-Assisted
7.
Math Med Biol ; 35(3): 319-346, 2018 09 11.
Article in English | MEDLINE | ID: mdl-28520976

ABSTRACT

We develop a continuum model for the aggregation of cells cultured in a nutrient-rich medium in a culture well. We consider a 2D geometry, representing a vertical slice through the culture well, and assume that the cell layer depth is small compared with the typical lengthscale of the culture well. We adopt a continuum mechanics approach, treating the cells and culture medium as a two-phase mixture. Specifically, the cells and culture medium are treated as fluids. Additionally, the cell phase can generate forces in response to environmental cues, which include the concentration of a chemoattractant that is produced by the cells within the culture medium. The model leads to a system of coupled nonlinear partial differential equations for the volume fraction and velocity of the cell phase, the culture medium pressure and the chemoattractant concentration, which must be solved subject to appropriate boundary and initial conditions. To gain insight into the system, we consider two model reductions, appropriate when the cell layer depth is thin compared to the typical length scale of the culture well: a (simple) 1D and a (more involved) thin-film extensional flow reduction. By investigating the resulting systems of equations analytically and numerically, we identify conditions under which small amplitude perturbations to a homogeneous steady state (corresponding to a spatially uniform cell distribution) can lead to a spatially varying steady state (pattern formation). Our analysis reveals that the simpler 1D reduction has the same qualitative features as the thin-film extensional flow reduction in the linear and weakly nonlinear regimes, motivating the use of the simpler 1D modelling approach when a qualitative understanding of the system is required. However, the thin-film extensional flow reduction may be more appropriate when detailed quantitative agreement between modelling predictions and experimental data is desired. Furthermore, full numerical simulations of the two model reductions in regions of parameter space when the system is not close to marginal stability reveal significant differences in the evolution of the volume fraction and velocity of the cell phase, and chemoattractant concentration.


Subject(s)
Cell Aggregation/physiology , Models, Biological , Cell Culture Techniques , Cell Proliferation , Chemotaxis/physiology , Computer Simulation , Culture Media , Linear Models , Mathematical Concepts , Nonlinear Dynamics
8.
Math Biosci ; 292: 46-56, 2017 10.
Article in English | MEDLINE | ID: mdl-28735871

ABSTRACT

We develop a mathematical model to describe the regeneration of a hydrogel inserted into an ex vivo osteochondral explant. Specifically we use partial differential equations to describe the evolution of two populations of cells that migrate from the tissue surrounding the defect, proliferate, and compete for space and resources within the hydrogel. The two cell populations are chondrocytes and cells that infiltrate from the subchondral bone. Model simulations are used to investigate how different seeding strategies and growth factor placement within the hydrogel affect the spatial distribution of both cell types. Since chondrocyte migration is extremely slow, we conclude that the hydrogel should be seeded with chondrocytes prior to culture in order to obtain zonal chondrocyte distributions typical of those associated with healthy cartilage.


Subject(s)
Cell Movement , Chondrocytes/cytology , Hydrogel, Polyethylene Glycol Dimethacrylate , Models, Biological , Regeneration , Bone and Bones/cytology , Cartilage, Articular/cytology , Cell Proliferation , Intercellular Signaling Peptides and Proteins/metabolism , Tissue Engineering
9.
CPT Pharmacometrics Syst Pharmacol ; 5(11): 636-645, 2016 11.
Article in English | MEDLINE | ID: mdl-27863175

ABSTRACT

Experimental evidence suggests that antiangiogenic therapy gives rise to a transient window of vessel normalization, within which the efficacy of radiotherapy and chemotherapy may be enhanced. Preclinical experiments that measure components of vessel normalization are invasive and expensive. We have developed a mathematical model of vascular tumor growth from preclinical time-course data in a breast cancer xenograft model. We used a mixed-effects approach for model parameterization, leveraging tumor size data to identify a period of enhanced tumor growth that could potentially correspond to the transient window of vessel normalization. We estimated the characteristics of the window for mice treated with an anti-VEGF antibody (bevacizumab) or with a bispecific anti-VEGF/anti-angiopoietin-2 antibody (vanucizumab). We show how the mathematical model could theoretically be used to predict how to coordinate antiangiogenic therapy with radiotherapy or chemotherapy to maximize therapeutic effect, reducing the need for preclinical experiments that directly measure vessel normalization parameters.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Models, Biological , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab/pharmacology , Breast Neoplasms/blood supply , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Longitudinal Studies , Mice , Mice, SCID , Models, Statistical , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Random Allocation , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
10.
J Theor Biol ; 409: 115-132, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27590325

ABSTRACT

The bladder is a complex organ that is highly adaptive to its mechanical environment. The umbrella cells in the bladder uroepithelium are of particular interest: these cells actively change their surface area through exo- and endocytosis of cytoplasmic vesicles, and likely form a critical component in the mechanosensing process that communicates the sense of 'fullness' to the nervous system. In this paper we develop a first mechanical model for vesicle trafficking in umbrella cells in response to membrane tension during bladder filling. Recent experiments conducted on a disc of uroepithelial tissue motivate our model development. These experiments subject bladder tissue to fixed pressure differences and exhibit counterintuitive area changes. Through analysis of the mathematical model and comparison with experimental data in this setup, we gain an intuitive understanding of the biophysical processes involved and calibrate the vesicle trafficking rate parameters in our model. We then adapt the model to simulate in vivo bladder filling and investigate the potential effect of abnormalities in the vesicle trafficking machinery on bladder pathologies.


Subject(s)
Cell Membrane/metabolism , Models, Biological , Urinary Bladder/metabolism , Urothelium/metabolism , Biological Transport, Active/physiology , Humans , Surface Tension , Urinary Bladder/cytology , Urothelium/cytology
11.
J Theor Biol ; 398: 162-80, 2016 06 07.
Article in English | MEDLINE | ID: mdl-26987523

ABSTRACT

The development of anti-angiogenic drugs for cancer therapy has yielded some promising candidates, but novel approaches for interventions to angiogenesis have led to disappointing results. In addition, there is a shortage of biomarkers that are predictive of response to anti-angiogenic treatments. Consequently, the complex biochemical and physiological basis for tumour angiogenesis remains incompletely understood. We have adopted a mathematical approach to address these issues, formulating a spatially averaged multiscale model that couples the dynamics of VEGF, Ang1, Ang2 and PDGF, with those of mature and immature endothelial cells and pericyte cells. The model reproduces qualitative experimental results regarding pericyte coverage of vessels after treatment by anti-Ang2, anti-VEGF and combination anti-VEGF/anti-Ang2 antibodies. We used the steady state behaviours of the model to characterise angiogenic and non-angiogenic vascular phenotypes, and used mechanistic perturbations representing hypothetical anti-angiogenic treatments to generate testable hypotheses regarding transitions to non-angiogenic phenotypes that depend on the pre-treatment vascular phenotype. Additionally, we predicted a synergistic effect between anti-VEGF and anti-Ang2 treatments when applied to an immature pre-treatment vascular phenotype, but not when applied to a normalised angiogenic pre-treatment phenotype. Based on these findings, we conclude that changes in vascular phenotype are predicted to be useful as an experimental biomarker of response to treatment. Further, our analysis illustrates the potential value of non-spatial mathematical models for generating tractable predictions regarding the action of anti-angiogenic therapies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Blood Vessels/pathology , Models, Biological , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Angiopoietin-2/metabolism , Blood Vessels/drug effects , Computer Simulation , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Neovascularization, Pathologic/pathology , Numerical Analysis, Computer-Assisted , Phenotype , Platelet-Derived Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
12.
J Math Biol ; 72(7): 1775-809, 2016 06.
Article in English | MEDLINE | ID: mdl-26328534

ABSTRACT

Mechanical interactions between cells and the fibrous extracellular matrix (ECM) in which they reside play a key role in tissue development. Mechanical cues from the environment (such as stress, strain and fibre orientation) regulate a range of cell behaviours, including proliferation, differentiation and motility. In turn, the ECM structure is affected by cells exerting forces on the matrix which result in deformation and fibre realignment. In this paper we develop a mathematical model to investigate this mechanical feedback between cells and the ECM. We consider a three-phase mixture of collagen, culture medium and cells, and formulate a system of partial differential equations which represents conservation of mass and momentum for each phase. This modelling framework takes into account the anisotropic mechanical properties of the collagen gel arising from its fibrous microstructure. We also propose a cell-collagen interaction force which depends upon fibre orientation and collagen density. We use a combination of numerical and analytical techniques to study the influence of cell-ECM interactions on pattern formation in tissues. Our results illustrate the wide range of structures which may be formed, and how those that emerge depend upon the importance of cell-ECM interactions.


Subject(s)
Cells/metabolism , Extracellular Matrix/metabolism , Models, Biological , Anisotropy , Cells/cytology , Collagen/metabolism
13.
Biomech Model Mechanobiol ; 15(3): 663-81, 2016 06.
Article in English | MEDLINE | ID: mdl-26264498

ABSTRACT

We develop a model of wound healing in the framework of finite elasticity, focussing our attention on the processes of growth and contraction in the dermal layer of the skin. The dermal tissue is treated as a hyperelastic cylinder that surrounds the wound and is subject to symmetric deformations. By considering the initial recoil that is observed upon the application of a circular wound, we estimate the degree of residual tension in the skin and build an evolution law for mechanosensitive growth of the dermal tissue. Contraction of the wound is governed by a phenomenological law in which radial pressure is prescribed at the wound edge. The model reproduces three main phases of the healing process. Initially, the wound recoils due to residual stress in the surrounding tissue; the wound then heals as a result of contraction and growth; and finally, healing slows as contraction and growth decrease. Over a longer time period, the surrounding tissue remodels, returning to the residually stressed state. We identify the steady state growth profile associated with this remodelled state. The model is then used to predict the outcome of rewounding experiments designed to quantify the amount of stress in the tissue, and also to simulate the application of pressure treatments.


Subject(s)
Dermis/pathology , Elasticity , Models, Biological , Wound Healing , Anisotropy , Elastic Modulus , Kinetics , Numerical Analysis, Computer-Assisted , Stress, Mechanical
14.
J Theor Biol ; 379: 24-37, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-25913880

ABSTRACT

Kidney development is initiated by the outgrowth of an epithelial ureteric bud into a population of mesenchymal cells. Reciprocal morphogenetic responses between these two populations generate a highly branched epithelial ureteric tree with the mesenchyme differentiating into nephrons, the functional units of the kidney. While we understand some of the mechanisms involved, current knowledge fails to explain the variability of organ sizes and nephron endowment in mice and humans. Here we present a spatially-averaged mathematical model of kidney morphogenesis in which the growth of the two key populations is described by a system of time-dependant ordinary differential equations. We assume that branching is symmetric and is invoked when the number of epithelial cells per tip reaches a threshold value. This process continues until the number of mesenchymal cells falls below a critical value that triggers cessation of branching. The mathematical model and its predictions are validated against experimentally quantified C57Bl6 mouse embryonic kidneys. Numerical simulations are performed to determine how the final number of branches changes as key system parameters are varied (such as the growth rate of tip cells, mesenchyme cells, or component cell population exit rate). Our results predict that the developing kidney responds differently to loss of cap and tip cells. They also indicate that the final number of kidney branches is less sensitive to changes in the growth rate of the ureteric tip cells than to changes in the growth rate of the mesenchymal cells. By inference, increasing the growth rate of mesenchymal cells should maximise branch number. Our model also provides a framework for predicting the branching outcome when ureteric tip or mesenchyme cells change behaviour in response to different genetic or environmental developmental stresses.


Subject(s)
Kidney/embryology , Models, Biological , Organogenesis/physiology , Animals , Mice
15.
Math Med Biol ; 32(3): 345-66, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25323738

ABSTRACT

In this paper, we consider the derivation of macroscopic equations appropriate to describe the growth of biological tissue, employing a multiple-scale homogenization method to accommodate explicitly the influence of the underlying microscale structure of the material, and its evolution, on the macroscale dynamics. Such methods have been widely used to study porous and poroelastic materials; however, a distinguishing feature of biological tissue is its ability to remodel continuously in response to local environmental cues. Here, we present the derivation of a model broadly applicable to tissue engineering applications, characterized by cell proliferation and extracellular matrix deposition in porous scaffolds used within tissue culture systems, which we use to study coupling between fluid flow, nutrient transport, and microscale tissue growth. Attention is restricted to surface accretion within a rigid porous medium saturated with a Newtonian fluid; coupling between the various dynamics is achieved by specifying the rate of microscale growth to be dependent upon the uptake of a generic diffusible nutrient. The resulting macroscale model comprises a Darcy-type equation governing fluid flow, with flow characteristics dictated by the assumed periodic microstructure and surface growth rate of the porous medium, coupled to an advection-reaction equation specifying the nutrient concentration. Illustrative numerical simulations are presented to indicate the influence of microscale growth on macroscale dynamics, and to highlight the importance of including experimentally relevant microstructural information to correctly determine flow dynamics and nutrient delivery in tissue engineering applications.


Subject(s)
Biological Transport , Hydrodynamics , Models, Biological , Porosity , Tissue Engineering , Tissue Scaffolds
16.
J Math Biol ; 70(3): 485-532, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24615007

ABSTRACT

Angiogenesis is the formation of new blood vessels from pre-existing ones in response to chemical signals secreted by, for example, a wound or a tumour. In this paper, we propose a mesoscopic lattice-based model of angiogenesis, in which processes that include proliferation and cell movement are considered as stochastic events. By studying the dependence of the model on the lattice spacing and the number of cells involved, we are able to derive the deterministic continuum limit of our equations and compare it to similar existing models of angiogenesis. We further identify conditions under which the use of continuum models is justified, and others for which stochastic or discrete effects dominate. We also compare different stochastic models for the movement of endothelial tip cells which have the same macroscopic, deterministic behaviour, but lead to markedly different behaviour in terms of production of new vessel cells.


Subject(s)
Models, Cardiovascular , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Arteriovenous Anastomosis/cytology , Cell Movement , Cell Proliferation , Chemotaxis , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Mathematical Concepts , Stochastic Processes
17.
Bull Math Biol ; 76(8): 1953-80, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25053556

ABSTRACT

There is growing interest in inflammation due to its involvement in many diverse medical conditions, including Alzheimer's disease, cancer, arthritis and asthma. The traditional view that resolution of inflammation is a passive process is now being superceded by an alternative hypothesis whereby its resolution is an active, anti-inflammatory process that can be manipulated therapeutically. This shift in mindset has stimulated a resurgence of interest in the biological mechanisms by which inflammation resolves. The anti-inflammatory processes central to the resolution of inflammation revolve around macrophages and are closely related to pro-inflammatory processes mediated by neutrophils and their ability to damage healthy tissue. We develop a spatially averaged model of inflammation centring on its resolution, accounting for populations of neutrophils and macrophages and incorporating both pro- and anti-inflammatory processes. Our ordinary differential equation model exhibits two outcomes that we relate to healthy and unhealthy states. We use bifurcation analysis to investigate how variation in the system parameters affects its outcome. We find that therapeutic manipulation of the rate of macrophage phagocytosis can aid in resolving inflammation but success is critically dependent on the rate of neutrophil apoptosis. Indeed our model predicts that an effective treatment protocol would take a dual approach, targeting macrophage phagocytosis alongside neutrophil apoptosis.


Subject(s)
Apoptosis/immunology , Inflammation/immunology , Macrophages/immunology , Models, Immunological , Neutrophils/immunology , Phagocytosis/immunology , Humans
18.
J Theor Biol ; 361: 87-100, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25017724

ABSTRACT

Wound healing is a complex process in which a sequence of interrelated phases contributes to a reduction in wound size. For diabetic patients, many of these processes are compromised, so that wound healing slows down. In this paper we present a simple ordinary differential equation model for wound healing in which attention focusses on the dominant processes that contribute to closure of a full thickness wound. Asymptotic analysis of the resulting model reveals that normal healing occurs in stages: the initial and rapid elastic recoil of the wound is followed by a longer proliferative phase during which growth in the dermis dominates healing. At longer times, fibroblasts exert contractile forces on the dermal tissue, the resulting tension stimulating further dermal tissue growth and enhancing wound closure. By fitting the model to experimental data we find that the major difference between normal and diabetic healing is a marked reduction in the rate of dermal tissue growth for diabetic patients. The model is used to estimate the breakdown of dermal healing into two processes: tissue growth and contraction, the proportions of which provide information about the quality of the healed wound. We show further that increasing dermal tissue growth in the diabetic wound produces closure times similar to those associated with normal healing and we discuss the clinical implications of this hypothesised treatment.


Subject(s)
Diabetes Mellitus , Models, Biological , Wound Healing , Wounds and Injuries , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Humans , Wounds and Injuries/metabolism , Wounds and Injuries/pathology , Wounds and Injuries/physiopathology
19.
Article in English | MEDLINE | ID: mdl-23679466

ABSTRACT

The development of new models of biological tissues that consider cells in a discrete manner is becoming increasingly popular as an alternative to continuum methods based on partial differential equations, although formal relationships between the discrete and continuum frameworks remain to be established. For crystal mechanics, the discrete-to-continuum bridge is often made by assuming that local atom displacements can be mapped homogeneously from the mesoscale deformation gradient, an assumption known as the Cauchy-Born rule (CBR). Although the CBR does not hold exactly for noncrystalline materials, it may still be used as a first-order approximation for analytic calculations of effective stresses or strain energies. In this work, our goal is to investigate numerically the applicability of the CBR to two-dimensional cellular-scale models by assessing the mechanical behavior of model biological tissues, including crystalline (honeycomb) and noncrystalline reference states. The numerical procedure involves applying an affine deformation to the boundary cells and computing the quasistatic position of internal cells. The position of internal cells is then compared with the prediction of the CBR and an average deviation is calculated in the strain domain. For center-based cell models, we show that the CBR holds exactly when the deformation gradient is relatively small and the reference stress-free configuration is defined by a honeycomb lattice. We show further that the CBR may be used approximately when the reference state is perturbed from the honeycomb configuration. By contrast, for vertex-based cell models, a similar analysis reveals that the CBR does not provide a good representation of the tissue mechanics, even when the reference configuration is defined by a honeycomb lattice. The paper concludes with a discussion of the implications of these results for concurrent discrete and continuous modeling, adaptation of atom-to-continuum techniques to biological tissues, and model classification.


Subject(s)
Cells , Models, Biological , Reproducibility of Results
20.
Integr Biol (Camb) ; 5(3): 597-605, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23388834

ABSTRACT

A critical step in the dissemination of ovarian cancer is the formation of multicellular spheroids from cells shed from the primary tumour. The objectives of this study were to apply bioengineered three-dimensional (3D) microenvironments for culturing ovarian cancer spheroids in vitro and simultaneously to build on a mathematical model describing the growth of multicellular spheroids in these biomimetic matrices. Cancer cells derived from human epithelial ovarian carcinoma were embedded within biomimetic hydrogels of varying stiffness and grown for up to 4 weeks. Immunohistochemistry, imaging and growth analyses were used to quantify the dependence of cell proliferation and apoptosis on matrix stiffness, long-term culture and treatment with the anti-cancer drug paclitaxel. The mathematical model was formulated as a free boundary problem in which each spheroid was treated as an incompressible porous medium. The functional forms used to describe the rates of cell proliferation and apoptosis were motivated by the experimental work and predictions of the mathematical model compared with the experimental output. This work aimed to establish whether it is possible to simulate solid tumour growth on the basis of data on spheroid size, cell proliferation and cell death within these spheroids. The mathematical model predictions were in agreement with the experimental data set and simulated how the growth of cancer spheroids was influenced by mechanical and biochemical stimuli including matrix stiffness, culture duration and administration of a chemotherapeutic drug. Our computational model provides new perspectives on experimental results and has informed the design of new 3D studies of chemoresistance of multicellular cancer spheroids.


Subject(s)
Hydrogels/chemistry , Ovarian Neoplasms/pathology , Spheroids, Cellular/cytology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomimetics , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Models, Theoretical , Paclitaxel/pharmacology , Pressure , Stress, Mechanical , Time Factors , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...