Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 20(4): e1011975, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38557892

ABSTRACT

Arboviruses can emerge rapidly and cause explosive epidemics of severe disease. Some of the most epidemiologically important arboviruses, including dengue virus (DENV), Zika virus (ZIKV), Chikungunya (CHIKV) and yellow fever virus (YFV), are transmitted by Aedes mosquitoes, most notably Aedes aegypti and Aedes albopictus. After a mosquito blood feeds on an infected host, virus enters the midgut and infects the midgut epithelium. The virus must then overcome a series of barriers before reaching the mosquito saliva and being transmitted to a new host. The virus must escape from the midgut (known as the midgut escape barrier; MEB), which is thought to be mediated by transient changes in the permeability of the midgut-surrounding basal lamina layer (BL) following blood feeding. Here, we present a mathematical model of the within-mosquito population dynamics of DENV (as a model system for mosquito-borne viruses more generally) that includes the interaction of the midgut and BL which can account for the MEB. Our results indicate a dose-dependency of midgut establishment of infection as well as rate of escape from the midgut: collectively, these suggest that the extrinsic incubation period (EIP)-the time taken for DENV virus to be transmissible after infection-is shortened when mosquitoes imbibe more virus. Additionally, our experimental data indicate that multiple blood feeding events, which more closely mimic mosquito-feeding behavior in the wild, can hasten the course of infections, and our model predicts that this effect is sensitive to the amount of virus imbibed. Our model indicates that mutations to the virus which impact its replication rate in the midgut could lead to even shorter EIPs when double-feeding occurs. Mechanistic models of within-vector viral infection dynamics provide a quantitative understanding of infection dynamics and could be used to evaluate novel interventions that target the mosquito stages of the infection.


Subject(s)
Aedes , Dengue Virus , Dengue , Zika Virus Infection , Zika Virus , Animals , Gastrointestinal Tract , Mosquito Vectors
2.
PLoS Comput Biol ; 20(2): e1011252, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38363799

ABSTRACT

Tumour angiogenesis leads to the formation of blood vessels that are structurally and spatially heterogeneous. Poor blood perfusion, in conjunction with increased hypoxia and oxygen heterogeneity, impairs a tumour's response to radiotherapy. The optimal strategy for enhancing tumour perfusion remains unclear, preventing its regular deployment in combination therapies. In this work, we first identify vascular architectural features that correlate with enhanced perfusion following radiotherapy, using in vivo imaging data from vascular tumours. Then, we present a novel computational model to determine the relationship between these architectural features and blood perfusion in silico. If perfusion is defined to be the proportion of vessels that support blood flow, we find that vascular networks with small mean diameters and large numbers of angiogenic sprouts show the largest increases in perfusion post-irradiation for both biological and synthetic tumours. We also identify cases where perfusion increases due to the pruning of hypoperfused vessels, rather than blood being rerouted. These results indicate the importance of considering network composition when determining the optimal irradiation strategy. In the future, we aim to use our findings to identify tumours that are good candidates for perfusion enhancement and to improve the efficacy of combination therapies.


Subject(s)
Hypoxia , Neoplasms , Humans , Perfusion , Combined Modality Therapy , Oxygen , Neoplasms/radiotherapy
3.
Biophys J ; 123(7): 799-813, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38414238

ABSTRACT

Interstitial fluid flow is a feature of many solid tumors. In vitro experiments have shown that such fluid flow can direct tumor cell movement upstream or downstream depending on the balance between the competing mechanisms of tensotaxis (cell migration up stress gradients) and autologous chemotaxis (downstream cell movement in response to flow-induced gradients of self-secreted chemoattractants). In this work we develop a probabilistic-continuum, two-phase model for cell migration in response to interstitial flow. We use a kinetic description for the cell velocity probability density function, and model the flow-dependent mechanical and chemical stimuli as forcing terms that bias cell migration upstream and downstream. Using velocity-space averaging, we reformulate the model as a system of continuum equations for the spatiotemporal evolution of the cell volume fraction and flux in response to forcing terms that depend on the local direction and magnitude of the mechanochemical cues. We specialize our model to describe a one-dimensional cell layer subject to fluid flow. Using a combination of numerical simulations and asymptotic analysis, we delineate the parameter regime where transitions from downstream to upstream cell migration occur. As has been observed experimentally, the model predicts downstream-oriented chemotactic migration at low cell volume fractions, and upstream-oriented tensotactic migration at larger volume fractions. We show that the locus of the critical volume fraction, at which the system transitions from downstream to upstream migration, is dominated by the ratio of the rate of chemokine secretion and advection. Our model also predicts that, because the tensotactic stimulus depends strongly on the cell volume fraction, upstream, tensotaxis-dominated migration occurs only transiently when the cells are initially seeded, and transitions to downstream, chemotaxis-dominated migration occur at later times due to the dispersive effect of cell diffusion.


Subject(s)
Chemotaxis , Neoplasms , Humans , Cell Movement/physiology , Diffusion , Models, Biological
4.
PLoS Comput Biol ; 19(3): e1010994, 2023 03.
Article in English | MEDLINE | ID: mdl-36972297

ABSTRACT

We introduce a new spatial statistic, the weighted pair correlation function (wPCF). The wPCF extends the existing pair correlation function (PCF) and cross-PCF to describe spatial relationships between points marked with combinations of discrete and continuous labels. We validate its use through application to a new agent-based model (ABM) which simulates interactions between macrophages and tumour cells. These interactions are influenced by the spatial positions of the cells and by macrophage phenotype, a continuous variable that ranges from anti-tumour to pro-tumour. By varying model parameters that regulate macrophage phenotype, we show that the ABM exhibits behaviours which resemble the 'three Es of cancer immunoediting': Equilibrium, Escape, and Elimination. We use the wPCF to analyse synthetic images generated by the ABM. We show that the wPCF generates a 'human readable' statistical summary of where macrophages with different phenotypes are located relative to both blood vessels and tumour cells. We also define a distinct 'PCF signature' that characterises each of the three Es of immunoediting, by combining wPCF measurements with the cross-PCF describing interactions between vessels and tumour cells. By applying dimension reduction techniques to this signature, we identify its key features and train a support vector machine classifier to distinguish between simulation outputs based on their PCF signature. This proof-of-concept study shows how multiple spatial statistics can be combined to analyse the complex spatial features that the ABM generates, and to partition them into interpretable groups. The intricate spatial features produced by the ABM are similar to those generated by state-of-the-art multiplex imaging techniques which distinguish the spatial distribution and intensity of multiple biomarkers in biological tissue regions. Applying methods such as the wPCF to multiplex imaging data would exploit the continuous variation in biomarker intensities and generate more detailed characterisation of the spatial and phenotypic heterogeneity in tissue samples.


Subject(s)
Neoplasms , Humans , Neoplasms/pathology , Computer Simulation , Phenotype , Macrophages/pathology
5.
PLoS Comput Biol ; 19(8): e1011329, 2023 08.
Article in English | MEDLINE | ID: mdl-37578973

ABSTRACT

Although children and adolescents with acute lymphoblastic leukaemia (ALL) have high survival rates, approximately 15-20% of patients relapse. Risk of relapse is routinely estimated at diagnosis by biological factors, including flow cytometry data. This high-dimensional data is typically manually assessed by projecting it onto a subset of biomarkers. Cell density and "empty spaces" in 2D projections of the data, i.e. regions devoid of cells, are then used for qualitative assessment. Here, we use topological data analysis (TDA), which quantifies shapes, including empty spaces, in data, to analyse pre-treatment ALL datasets with known patient outcomes. We combine these fully unsupervised analyses with Machine Learning (ML) to identify significant shape characteristics and demonstrate that they accurately predict risk of relapse, particularly for patients previously classified as 'low risk'. We independently confirm the predictive power of CD10, CD20, CD38, and CD45 as biomarkers for ALL diagnosis. Based on our analyses, we propose three increasingly detailed prognostic pipelines for analysing flow cytometry data from ALL patients depending on technical and technological availability: 1. Visual inspection of specific biological features in biparametric projections of the data; 2. Computation of quantitative topological descriptors of such projections; 3. A combined analysis, using TDA and ML, in the four-parameter space defined by CD10, CD20, CD38 and CD45. Our analyses readily extend to other haematological malignancies.


Subject(s)
Hematologic Neoplasms , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Adolescent , Humans , Neoplasm Recurrence, Local , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Flow Cytometry , Immunophenotyping , Recurrence
6.
Bull Math Biol ; 86(2): 19, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38238433

ABSTRACT

Longitudinal tumour volume data from head-and-neck cancer patients show that tumours of comparable pre-treatment size and stage may respond very differently to the same radiotherapy fractionation protocol. Mathematical models are often proposed to predict treatment outcome in this context, and have the potential to guide clinical decision-making and inform personalised fractionation protocols. Hindering effective use of models in this context is the sparsity of clinical measurements juxtaposed with the model complexity required to produce the full range of possible patient responses. In this work, we present a compartment model of tumour volume and tumour composition, which, despite relative simplicity, is capable of producing a wide range of patient responses. We then develop novel statistical methodology and leverage a cohort of existing clinical data to produce a predictive model of both tumour volume progression and the associated level of uncertainty that evolves throughout a patient's course of treatment. To capture inter-patient variability, all model parameters are patient specific, with a bootstrap particle filter-like Bayesian approach developed to model a set of training data as prior knowledge. We validate our approach against a subset of unseen data, and demonstrate both the predictive ability of our trained model and its limitations.


Subject(s)
Models, Biological , Neoplasms , Humans , Bayes Theorem , Mathematical Concepts , Models, Theoretical , Neoplasms/radiotherapy
7.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34625491

ABSTRACT

Highly resolved spatial data of complex systems encode rich and nonlinear information. Quantification of heterogeneous and noisy data-often with outliers, artifacts, and mislabeled points-such as those from tissues, remains a challenge. The mathematical field that extracts information from the shape of data, topological data analysis (TDA), has expanded its capability for analyzing real-world datasets in recent years by extending theory, statistics, and computation. An extension to the standard theory to handle heterogeneous data is multiparameter persistent homology (MPH). Here we provide an application of MPH landscapes, a statistical tool with theoretical underpinnings. MPH landscapes, computed for (noisy) data from agent-based model simulations of immune cells infiltrating into a spheroid, are shown to surpass existing spatial statistics and one-parameter persistent homology. We then apply MPH landscapes to study immune cell location in digital histology images from head and neck cancer. We quantify intratumoral immune cells and find that infiltrating regulatory T cells have more prominent voids in their spatial patterns than macrophages. Finally, we consider how TDA can integrate and interrogate data of different types and scales, e.g., immune cell locations and regions with differing levels of oxygenation. This work highlights the power of MPH landscapes for quantifying, characterizing, and comparing features within the tumor microenvironment in synthetic and real datasets.


Subject(s)
Head and Neck Neoplasms/diagnostic imaging , Macrophages/cytology , T-Lymphocytes, Regulatory/cytology , Tumor Hypoxia/physiology , Tumor Microenvironment/immunology , Cell Count/methods , Computational Biology/methods , Computer Simulation , Data Analysis , Head and Neck Neoplasms/immunology , Humans , Macrophages/immunology , Spheroids, Cellular , T-Lymphocytes, Regulatory/immunology
8.
Microcirculation ; 30(4): e12799, 2023 05.
Article in English | MEDLINE | ID: mdl-36635617

ABSTRACT

OBJECTIVE: Disease complications can alter vascular network morphology and disrupt tissue functioning. Microvascular diseases of the retina are assessed by visual inspection of retinal images, but this can be challenging when diseases exhibit silent symptoms or patients cannot attend in-person meetings. We examine the performance of machine learning algorithms in detecting microvascular disease when trained on statistical and topological summaries of segmented retinal vascular images. METHODS: We compute 13 separate descriptor vectors (5 statistical, 8 topological) to summarize the morphology of retinal vessel segmentation images and train support vector machines to predict each image's disease classification from the summary vectors. We assess the performance of each descriptor vector, using five-fold cross validation to estimate their accuracy. We apply these methods to four datasets that were assembled from four existing data repositories; three datasets contain segmented retinal vascular images from one of the repositories, whereas the fourth "All" dataset combines images from four repositories. RESULTS: Among the 13 total descriptor vectors considered, either a statistical Box-counting descriptor vector or a topological Flooding descriptor vector achieves the highest accuracy levels. On the combined "All" dataset, the Box-counting vector outperforms all other descriptors, including the topological Flooding vector which is sensitive to differences in the annotation styles between the different datasets. CONCLUSION: Our work represents a first step to establishing which computational methods are most suitable for identifying microvascular disease and assessing their current limitations. These methods could be incorporated into automated disease assessment tools.


Subject(s)
Retina , Retinal Vessels , Humans , Retina/diagnostic imaging , Retinal Vessels/diagnostic imaging , Algorithms
9.
J Theor Biol ; 559: 111377, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36470468

ABSTRACT

The Lotka-Volterra model is widely used to model interactions between two species. Here, we generate synthetic data mimicking competitive, mutualistic and antagonistic interactions between two tumor cell lines, and then use the Lotka-Volterra model to infer the interaction type. Structural identifiability of the Lotka-Volterra model is confirmed, and practical identifiability is assessed for three experimental designs: (a) use of a single data set, with a mixture of both cell lines observed over time, (b) a sequential design where growth rates and carrying capacities are estimated using data from experiments in which each cell line is grown in isolation, and then interaction parameters are estimated from an experiment involving a mixture of both cell lines, and (c) a parallel experimental design where all model parameters are fitted to data from two mixtures (containing both cell lines but with different initial ratios) simultaneously. Each design is tested on data generated from the Lotka-Volterra model with noise added, to determine efficacy in an ideal sense. In addition to assessing each design for practical identifiability, we investigate how the predictive power of the model - i.e., its ability to fit data for initial ratios other than those to which it was calibrated - is affected by the choice of experimental design. The parallel calibration procedure is found to be optimal and is further tested on in silico data generated from a spatially-resolved cellular automaton model, which accounts for oxygen consumption and allows for variation in the intensity level of the interaction between the two cell lines. We use this study to highlight the care that must be taken when interpreting parameter estimates for the spatially-averaged Lotka-Volterra model when it is calibrated against data produced by the spatially-resolved cellular automaton model, since baseline competition for space and resources in the CA model may contribute to a discrepancy between the type of interaction used to generate the CA data and the type of interaction inferred by the LV model.


Subject(s)
Models, Biological , Symbiosis , Cell Line, Tumor
10.
J Theor Biol ; 556: 111248, 2023 01 07.
Article in English | MEDLINE | ID: mdl-36150537

ABSTRACT

We present a mathematical model that describes how tumour heterogeneity evolves in a tissue slice that is oxygenated by a single blood vessel. Phenotype is identified with the stemness level of a cell and determines its proliferative capacity, apoptosis propensity and response to treatment. Our study is based on numerical bifurcation analysis and dynamical simulations of a system of coupled, non-local (in phenotypic "space") partial differential equations that link the phenotypic evolution of the tumour cells to local tissue oxygen levels. In our formulation, we consider a 1D geometry where oxygen is supplied by a blood vessel located on the domain boundary and consumed by the tumour cells as it diffuses through the tissue. For biologically relevant parameter values, the system exhibits multiple steady states; in particular, depending on the initial conditions, the tumour is either eliminated ("tumour-extinction") or it persists ("tumour-invasion"). We conclude by using the model to investigate tumour responses to radiotherapy, and focus on identifying radiotherapy strategies which can eliminate the tumour. Numerical simulations reveal how phenotypic heterogeneity evolves during treatment and highlight the critical role of tissue oxygen levels on the efficacy of radiation protocols that are commonly used in the clinic.


Subject(s)
Neoplasms , Humans , Neoplasms/pathology , Models, Theoretical , Phenotype , Oxygen
11.
J Theor Biol ; 567: 111491, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37044357

ABSTRACT

We consider a hierarchy of ordinary differential equation models that describe the within-host viral kinetics of influenza infections: the IR model explicitly accounts for an immune response to the virus, while the simpler, target-cell limited TEIV and TV models do not. We show that when the IR model is fitted to pooled experimental murine data of the viral load, fraction of dead cells, and immune response levels, its parameters values can be determined. However, if, as is common, only viral load data are available, we can estimate parameters of the TEIV and TV models but not the IR model. These results are substantiated by a structural and practical identifiability analysis. We then use the IR model to generate synthetic data representing infections in hosts whose immune responses differ. We fit the TV model to these synthetic datasets and show that it can reproduce the characteristic exponential increase and decay of viral load generated by the IR model. Furthermore, the values of the fitted parameters of the TV model can be mapped from the immune response parameters in the IR model. We conclude that, if only viral load data are available, a simple target-cell limited model can reproduce influenza infection dynamics and distinguish between hosts with differing immune responses.


Subject(s)
Influenza, Human , Animals , Mice , Humans , Immunity, Innate
12.
Bull Math Biol ; 85(8): 74, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37378740

ABSTRACT

Cancer is a heterogeneous disease and tumours of the same type can differ greatly at the genetic and phenotypic levels. Understanding how these differences impact sensitivity to treatment is an essential step towards patient-specific treatment design. In this paper, we investigate how two different mechanisms for growth control may affect tumour cell responses to fractionated radiotherapy (RT) by extending an existing ordinary differential equation model of tumour growth. In the absence of treatment, this model distinguishes between growth arrest due to nutrient insufficiency and competition for space and exhibits three growth regimes: nutrient limited, space limited (SL) and bistable (BS), where both mechanisms for growth arrest coexist. We study the effect of RT for tumours in each regime, finding that tumours in the SL regime typically respond best to RT, while tumours in the BS regime typically respond worst to RT. For tumours in each regime, we also identify the biological processes that may explain positive and negative treatment outcomes and the dosing regimen which maximises the reduction in tumour burden.


Subject(s)
Models, Biological , Neoplasms , Humans , Mathematical Concepts , Neoplasms/radiotherapy , Neoplasms/pathology
13.
Bull Math Biol ; 85(5): 37, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36991234

ABSTRACT

Atherosclerosis is an inflammatory disease characterised by the formation of plaques, which are deposits of lipids and cholesterol-laden macrophages that form in the artery wall. The inflammation is often non-resolving, due in large part to changes in normal macrophage anti-inflammatory behaviour that are induced by the toxic plaque microenvironment. These changes include higher death rates, defective efferocytic uptake of dead cells, and reduced rates of emigration. We develop a free boundary multiphase model for early atherosclerotic plaques, and we use it to investigate the effects of impaired macrophage anti-inflammatory behaviour on plaque structure and growth. We find that high rates of cell death relative to efferocytic uptake results in a plaque populated mostly by dead cells. We also find that emigration can potentially slow or halt plaque growth by allowing material to exit the plaque, but this is contingent on the availability of live macrophage foam cells in the deep plaque. Finally, we introduce an additional bead species to model macrophage tagging via microspheres, and we use the extended model to explore how high rates of cell death and low rates of efferocytosis and emigration prevent the clearance of macrophages from the plaque.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Plaque, Atherosclerotic/metabolism , Models, Biological , Mathematical Concepts , Macrophages/physiology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
14.
Bull Math Biol ; 85(5): 38, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36991173

ABSTRACT

Tumour spheroids have been the focus of a variety of mathematical models, ranging from Greenspan's classical study of the 1970 s through to contemporary agent-based models. Of the many factors that regulate spheroid growth, mechanical effects are perhaps some of the least studied, both theoretically and experimentally, though experimental enquiry has established their significance to tumour growth dynamics. In this tutorial, we formulate a hierarchy of mathematical models of increasing complexity to explore the role of mechanics in spheroid growth, all the while seeking to retain desirable simplicity and analytical tractability. Beginning with the theory of morphoelasticity, which combines solid mechanics and growth, we successively refine our assumptions to develop a somewhat minimal model of mechanically regulated spheroid growth that is free from many unphysical and undesirable behaviours. In doing so, we will see how iterating upon simple models can provide rigorous guarantees of emergent behaviour, which are often precluded by existing, more complex modelling approaches. Perhaps surprisingly, we also demonstrate that the final model considered in this tutorial agrees favourably with classical experimental results, highlighting the potential for simple models to provide mechanistic insight whilst also serving as mathematical examples.


Subject(s)
Neoplasms , Spheroids, Cellular , Humans , Models, Biological , Mathematical Concepts , Models, Theoretical
15.
Proc Natl Acad Sci U S A ; 117(45): 27811-27819, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33109723

ABSTRACT

Oxygen heterogeneity in solid tumors is recognized as a limiting factor for therapeutic efficacy. This heterogeneity arises from the abnormal vascular structure of the tumor, but the precise mechanisms linking abnormal structure and compromised oxygen transport are only partially understood. In this paper, we investigate the role that red blood cell (RBC) transport plays in establishing oxygen heterogeneity in tumor tissue. We focus on heterogeneity driven by network effects, which are challenging to observe experimentally due to the reduced fields of view typically considered. Motivated by our findings of abnormal vascular patterns linked to deviations from current RBC transport theory, we calculated average vessel lengths [Formula: see text] and diameters [Formula: see text] from tumor allografts of three cancer cell lines and observed a substantial reduction in the ratio [Formula: see text] compared to physiological conditions. Mathematical modeling reveals that small values of the ratio λ (i.e., [Formula: see text]) can bias hematocrit distribution in tumor vascular networks and drive heterogeneous oxygenation of tumor tissue. Finally, we show an increase in the value of λ in tumor vascular networks following treatment with the antiangiogenic cancer agent DC101. Based on our findings, we propose λ as an effective way of monitoring the efficacy of antiangiogenic agents and as a proxy measure of perfusion and oxygenation in tumor tissue undergoing antiangiogenic treatment.


Subject(s)
Blood Circulation/physiology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Angiogenesis Inhibitors/therapeutic use , Animals , Biomarkers, Tumor/physiology , Cell Line, Tumor , Erythrocytes/metabolism , Genetic Heterogeneity , Hematocrit , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Models, Theoretical , Neoplasms/drug therapy , Oxygen/metabolism , Perfusion
16.
J Theor Biol ; 545: 111104, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35337794

ABSTRACT

New experimental data have shown how the periodic exposure of cells to low oxygen levels (i.e., cyclic hypoxia) impacts their progress through the cell-cycle. Cyclic hypoxia has been detected in tumours and linked to poor prognosis and treatment failure. While fluctuating oxygen environments can be reproduced in vitro, the range of oxygen cycles that can be tested is limited. By contrast, mathematical models can be used to predict the response to a wide range of cyclic dynamics. Accordingly, in this paper we develop a mechanistic model of the cell-cycle that can be combined with in vitro experiments to better understand the link between cyclic hypoxia and cell-cycle dysregulation. A distinguishing feature of our model is the inclusion of impaired DNA synthesis and cell-cycle arrest due to periodic exposure to severely low oxygen levels. Our model decomposes the cell population into five compartments and a time-dependent delay accounts for the variability in the duration of the S phase which increases in severe hypoxia due to reduced rates of DNA synthesis. We calibrate our model against experimental data and show that it recapitulates the observed cell-cycle dynamics. We use the calibrated model to investigate the response of cells to oxygen cycles not yet tested experimentally. When the re-oxygenation phase is sufficiently long, our model predicts that cyclic hypoxia simply slows cell proliferation since cells spend more time in the S phase. On the contrary, cycles with short periods of re-oxygenation are predicted to lead to inhibition of proliferation, with cells arresting from the cell-cycle in the G2 phase. While model predictions on short time scales (about a day) are fairly accurate (i.e, confidence intervals are small), the predictions become more uncertain over longer periods. Hence, we use our model to inform experimental design that can lead to improved model parameter estimates and validate model predictions.


Subject(s)
Hypoxia , Oxygen , Cell Hypoxia/physiology , DNA/metabolism , Humans , Models, Theoretical , Oxygen/metabolism
17.
PLoS Comput Biol ; 17(1): e1008055, 2021 01.
Article in English | MEDLINE | ID: mdl-33411727

ABSTRACT

We introduce a hybrid two-dimensional multiscale model of angiogenesis, the process by which endothelial cells (ECs) migrate from a pre-existing vascular bed in response to local environmental cues and cell-cell interactions, to create a new vascular network. Recent experimental studies have highlighted a central role of cell rearrangements in the formation of angiogenic networks. Our model accounts for this phenomenon via the heterogeneous response of ECs to their microenvironment. These cell rearrangements, in turn, dynamically remodel the local environment. The model reproduces characteristic features of angiogenic sprouting that include branching, chemotactic sensitivity, the brush border effect, and cell mixing. These properties, rather than being hardwired into the model, emerge naturally from the gene expression patterns of individual cells. After calibrating and validating our model against experimental data, we use it to predict how the structure of the vascular network changes as the baseline gene expression levels of the VEGF-Delta-Notch pathway, and the composition of the extracellular environment, vary. In order to investigate the impact of cell rearrangements on the vascular network structure, we introduce the mixing measure, a scalar metric that quantifies cell mixing as the vascular network grows. We calculate the mixing measure for the simulated vascular networks generated by ECs of different lineages (wild type cells and mutant cells with impaired expression of a specific receptor). Our results show that the time evolution of the mixing measure is directly correlated to the generic features of the vascular branching pattern, thus, supporting the hypothesis that cell rearrangements play an essential role in sprouting angiogenesis. Furthermore, we predict that lower cell rearrangement leads to an imbalance between branching and sprout elongation. Since the computation of this statistic requires only individual cell trajectories, it can be computed for networks generated in biological experiments, making it a potential biomarker for pathological angiogenesis.


Subject(s)
Endothelial Cells , Models, Biological , Neovascularization, Physiologic/physiology , Animals , Cell Differentiation/physiology , Cell Line , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/physiology , Mice , Neovascularization, Pathologic/physiopathology , Signal Transduction/physiology , Transcriptome/physiology
18.
PLoS Comput Biol ; 17(6): e1009094, 2021 06.
Article in English | MEDLINE | ID: mdl-34181657

ABSTRACT

Angiogenesis is the process by which blood vessels form from pre-existing vessels. It plays a key role in many biological processes, including embryonic development and wound healing, and contributes to many diseases including cancer and rheumatoid arthritis. The structure of the resulting vessel networks determines their ability to deliver nutrients and remove waste products from biological tissues. Here we simulate the Anderson-Chaplain model of angiogenesis at different parameter values and quantify the vessel architectures of the resulting synthetic data. Specifically, we propose a topological data analysis (TDA) pipeline for systematic analysis of the model. TDA is a vibrant and relatively new field of computational mathematics for studying the shape of data. We compute topological and standard descriptors of model simulations generated by different parameter values. We show that TDA of model simulation data stratifies parameter space into regions with similar vessel morphology. The methodologies proposed here are widely applicable to other synthetic and experimental data including wound healing, development, and plant biology.


Subject(s)
Models, Cardiovascular , Neovascularization, Pathologic , Neovascularization, Physiologic , Algorithms , Animals , Blood Vessels/anatomy & histology , Blood Vessels/growth & development , Blood Vessels/physiology , Chemotaxis , Computational Biology , Computer Simulation , Humans , Neoplasms/blood supply , Spatio-Temporal Analysis
19.
Bull Math Biol ; 84(8): 80, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35773547

ABSTRACT

The processes underpinning solid tumour growth involve the interactions between various healthy and tumour tissue components and the vasculature, and can be affected in different ways by cancer treatment. In particular, the growth-limiting mechanisms at play may influence tumour responses to treatment. In this paper, we propose a simple ordinary differential equation model of solid tumour growth to investigate how tumour-specific mechanisms of growth arrest may affect tumour response to different combination cancer therapies. We consider the interactions of tumour cells with the physical space in which they proliferate and a nutrient supplied by the tumour vasculature, with the aim of representing two distinct growth arrest mechanisms. More specifically, we wish to consider growth arrest due to (1) nutrient deficiency, which corresponds to balancing cell proliferation and death rates, and (2) competition for space, which corresponds to cessation of proliferation without cell death. We perform numerical simulations of the model and a steady-state analysis to determine the possible tumour growth scenarios described by the model. We find that there are three distinct growth regimes: the nutrient- and spatially limited regimes and a bi-stable regime, in which both growth arrest mechanisms are simultaneously active. Thus, the proposed model has the features required to investigate and distinguish tumour responses to different cancer treatments.


Subject(s)
Models, Biological , Neoplasms , Computer Simulation , Humans , Mathematical Concepts , Mathematics , Neoplasms/pathology
20.
Bull Math Biol ; 84(12): 137, 2022 10 23.
Article in English | MEDLINE | ID: mdl-36273372

ABSTRACT

The MEK/ERK signalling pathway is involved in cell division, cell specialisation, survival and cell death (Shaul and Seger in Biochim Biophys Acta (BBA)-Mol Cell Res 1773(8):1213-1226, 2007). Here we study a polynomial dynamical system describing the dynamics of MEK/ERK proposed by Yeung et al. (Curr Biol 2019, https://doi.org/10.1016/j.cub.2019.12.052 ) with their experimental setup, data and known biological information. The experimental dataset is a time-course of ERK measurements in different phosphorylation states following activation of either wild-type MEK or MEK mutations associated with cancer or developmental defects. We demonstrate how methods from computational algebraic geometry, differential algebra, Bayesian statistics and computational algebraic topology can inform the model reduction, identification and parameter inference of MEK variants, respectively. Throughout, we show how this algebraic viewpoint offers a rigorous and systematic analysis of such models.


Subject(s)
Mathematical Concepts , Models, Biological , Kinetics , Bayes Theorem , Phosphorylation , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL