Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Theor Biol ; 592: 111882, 2024 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-38944379

RESUMO

Regulation of cell proliferation is a crucial aspect of tissue development and homeostasis and plays a major role in morphogenesis, wound healing, and tumor invasion. A phenomenon of such regulation is contact inhibition, which describes the dramatic slowing of proliferation, cell migration and individual cell growth when multiple cells are in contact with each other. While many physiological, molecular and genetic factors are known, the mechanism of contact inhibition is still not fully understood. In particular, the relevance of cellular signaling due to interfacial contact for contact inhibition is still debated. Cellular automata (CA) have been employed in the past as numerically efficient mathematical models to study the dynamics of cell ensembles, but they are not suitable to explore the origins of contact inhibition as such agent-based models assume fixed cell sizes. We develop a minimal, data-driven model to simulate the dynamics of planar cell cultures by extending a probabilistic CA to incorporate size changes of individual cells during growth and cell division. We successfully apply this model to previous in-vitro experiments on contact inhibition in epithelial tissue: After a systematic calibration of the model parameters to measurements of single-cell dynamics, our CA model quantitatively reproduces independent measurements of emergent, culture-wide features, like colony size, cell density and collective cell migration. In particular, the dynamics of the CA model also exhibit the transition from a low-density confluent regime to a stationary postconfluent regime with a rapid decrease in cell size and motion. This implies that the volume exclusion principle, a mechanical constraint which is the only inter-cellular interaction incorporated in the model, paired with a size-dependent proliferation rate is sufficient to generate the observed contact inhibition. We discuss how our approach enables the introduction of effective bio-mechanical interactions in a CA framework for future studies.


Assuntos
Proliferação de Células , Tamanho Celular , Células Epiteliais , Modelos Biológicos , Proliferação de Células/fisiologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Inibição de Contato/fisiologia , Humanos , Animais , Movimento Celular/fisiologia
2.
Cancers (Basel) ; 15(23)2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38067348

RESUMO

Understanding the complex dynamics of tumor growth to develop more efficient therapeutic strategies is one of the most challenging problems in biomedicine. Three-dimensional (3D) tumor spheroids, reflecting avascular microregions within a tumor, are an advanced in vitro model system to assess the curative effect of combinatorial radio(chemo)therapy. Tumor spheroids exhibit particular crucial pathophysiological characteristics such as a radial oxygen gradient that critically affect the sensitivity of the malignant cell population to treatment. However, spheroid experiments remain laborious, and determining long-term radio(chemo)therapy outcomes is challenging. Mathematical models of spheroid dynamics have the potential to enhance the informative value of experimental data, and can support study design; however, they typically face one of two limitations: while non-spatial models are computationally cheap, they lack the spatial resolution to predict oxygen-dependent radioresponse, whereas models that describe spatial cell dynamics are computationally expensive and often heavily parameterized, impeding the required calibration to experimental data. Here, we present an effectively one-dimensional mathematical model based on the cell dynamics within and across radial spheres which fully incorporates the 3D dynamics of tumor spheroids by exploiting their approximate rotational symmetry. We demonstrate that this radial-shell (RS) model reproduces experimental spheroid growth curves of several cell lines with and without radiotherapy, showing equal or better performance than published models such as 3D agent-based models. Notably, the RS model is sufficiently efficient to enable multi-parametric optimization within previously reported and/or physiologically reasonable ranges based on experimental data. Analysis of the model reveals that the characteristic change of dynamics observed in experiments at small spheroid volume originates from the spatial scale of cell interactions. Based on the calibrated parameters, we predict the spheroid volumes at which this behavior should be observable. Finally, we demonstrate how the generic parameterization of the model allows direct parameter transfer to 3D agent-based models.

3.
PLoS Comput Biol ; 18(9): e1010460, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36121837

RESUMO

Understanding the segregation of cells is crucial to answer questions about tissue formation in embryos or tumor progression. Steinberg proposed that separation of cells can be compared to the separation of two liquids. Such a separation is well described by the Cahn-Hilliard (CH) equations and the segregation indices exhibit an algebraic decay with exponent 1/3 with respect to time. Similar exponents are also observed in cell-based models. However, the scaling behavior in these numerical models is usually only examined in the asymptotic regime and these models have not been directly applied to actual cell segregation data. In contrast, experimental data also reveals other scaling exponents and even slow logarithmic scaling laws. These discrepancies are commonly attributed to the effects of collective motion or velocity-dependent interactions. By calibrating a 2D cellular automaton (CA) model which efficiently implements a dynamic variant of the differential adhesion hypothesis to 2D experimental data from Méhes et al., we reproduce the biological cell segregation experiments with just adhesive forces. The segregation in the cellular automaton model follows a logarithmic scaling initially, which is in contrast to the proposed algebraic scaling with exponent 1/3. However, within the less than two orders of magnitudes in time which are observable in the experiments, a logarithmic scaling may appear as a pseudo-algebraic scaling. In particular, we demonstrate that the cellular automaton model can exhibit a range of exponents ≤1/3 for such a pseudo-algebraic scaling. Moreover, the time span of the experiment falls into the transitory regime of the cellular automaton rather than the asymptotic one. We additionally develop a method for the calibration of the 2D Cahn-Hilliard model and find a match with experimental data within the transitory regime of the Cahn-Hilliard model with exponent 1/4. On the one hand this demonstrates that the transitory behavior is relevant for the experiment rather than the asymptotic one. On the other hand this corroborates the ambiguity of the scaling behavior, when segregation processes can be only observed on short time spans.


Assuntos
Movimento (Física) , Separação Celular
4.
PLoS Comput Biol ; 18(8): e1010403, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35984850

RESUMO

Cancer development is a multistep process often starting with a single cell in which a number of epigenetic and genetic alterations have accumulated thus transforming it into a tumor cell. The progeny of such a single benign tumor cell expands in the tissue and can at some point progress to malignant tumor cells until a detectable tumor is formed. The dynamics from the early phase of a single cell to a detectable tumor with billions of tumor cells are complex and still not fully resolved, not even for the well-known prototype of multistage carcinogenesis, the adenoma-adenocarcinoma sequence of colorectal cancer. Mathematical models of such carcinogenesis are frequently tested and calibrated based on reported age-specific incidence rates of cancer, but they usually require calibration of four or more parameters due to the wide range of processes these models aim to reflect. We present a cell-based model, which focuses on the competition between wild-type and tumor cells in colonic crypts, with which we are able reproduce epidemiological incidence rates of colon cancer. Additionally, the fraction of cancerous tumors with precancerous lesions predicted by the model agree with clinical estimates. The correspondence between model and reported data suggests that the fate of tumor development is majorly determined by the early phase of tumor growth and progression long before a tumor becomes detectable. Due to the focus on the early phase of tumor development, the model has only a single fit parameter, the time scale set by an effective replacement rate of stem cells in the crypt. We find this effective rate to be considerable smaller than the actual replacement rate, which implies that the time scale is limited by the processes succeeding clonal conversion of crypts.


Assuntos
Neoplasias do Colo , Fatores Etários , Carcinogênese , Colo , Neoplasias do Colo/epidemiologia , Neoplasias do Colo/genética , Humanos , Incidência
5.
Mol Cancer Res ; 20(5): 794-809, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35135863

RESUMO

Tumor heterogeneity and cellular plasticity are key determinants of tumor progression, metastatic spread, and therapy response driven by the cancer stem cell (CSC) population. Within the current study, we analyzed irradiation-induced plasticity within the aldehyde dehydrogenase (ALDH)-positive (ALDH+) population in prostate cancer. The radiosensitivity of xenograft tumors derived from ALDH+ and ALDH-negative (ALDH-) cells was determined with local tumor control analyses and demonstrated different dose-response profiles, time to relapse, and focal adhesion signaling. The transcriptional heterogeneity was analyzed in pools of 10 DU145 and PC3 cells with multiplex gene expression analyses and illustrated a higher degree of heterogeneity within the ALDH+ population that even increases upon irradiation in comparison with ALDH- cells. Phenotypic conversion and clonal competition were analyzed with fluorescence protein-labeled cells to distinguish cellular origins in competitive three-dimensional cultures and xenograft tumors. We found that the ALDH+ population outcompetes ALDH- cells and drives tumor growth, in particular upon irradiation. The observed dynamics of the cellular state compositions between ALDH+ and ALDH- cells in vivo before and after tumor irradiation was reproduced by a probabilistic Markov compartment model that incorporates cellular plasticity, clonal competition, and phenotype-specific radiosensitivities. Transcriptional analyses indicate that the cellular conversion from ALDH- into ALDH+ cells within xenograft tumors under therapeutic pressure was partially mediated through induction of the transcriptional repressor SNAI2. In summary, irradiation-induced cellular conversion events are present in xenograft tumors derived from prostate cancer cells and may be responsible for radiotherapy failure. IMPLICATIONS: The increase of ALDH+ cells with stem-like features in prostate xenograft tumors after local irradiation represents a putative cellular escape mechanism inducing tumor radioresistance.


Assuntos
Aldeído Desidrogenase , Neoplasias da Próstata , Aldeído Desidrogenase/genética , Humanos , Masculino , Recidiva Local de Neoplasia , Células PC-3 , Neoplasias da Próstata/genética , Neoplasias da Próstata/radioterapia , Tolerância a Radiação
6.
Entropy (Basel) ; 23(11)2021 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-34828077

RESUMO

The process of cell-sorting is essential for development and maintenance of tissues. Mathematical modeling can provide the means to analyze the consequences of different hypotheses about the underlying mechanisms. With the Differential Adhesion Hypothesis, Steinberg proposed that cell-sorting is determined by quantitative differences in cell-type-specific intercellular adhesion strengths. An implementation of the Differential Adhesion Hypothesis is the Differential Migration Model by Voss-Böhme and Deutsch. There, an effective adhesion parameter was derived analytically for systems with two cell types, which predicts the asymptotic sorting pattern. However, the existence and form of such a parameter for more than two cell types is unclear. Here, we generalize analytically the concept of an effective adhesion parameter to three and more cell types and demonstrate its existence numerically for three cell types based on in silico time-series data that is produced by a cellular-automaton implementation of the Differential Migration Model. Additionally, we classify the segregation behavior using statistical learning methods and show that the estimated effective adhesion parameter for three cell types matches our analytical prediction. Finally, we demonstrate that the effective adhesion parameter can resolve a recent dispute about the impact of interfacial adhesion, cortical tension and heterotypic repulsion on cell segregation.

7.
Philos Trans R Soc Lond B Biol Sci ; 375(1807): 20190378, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32713300

RESUMO

Biological processes, such as embryonic development, wound repair and cancer invasion, or bacterial swarming and fruiting body formation, involve collective motion of cells as a coordinated group. Collective cell motion of eukaryotic cells often includes interactions that result in polar alignment of cell velocities, while bacterial patterns typically show features of apolar velocity alignment. For analysing the population-scale effects of these different alignment mechanisms, various on- and off-lattice agent-based models have been introduced. However, discriminating model-specific artefacts from general features of collective cell motion is challenging. In this work, we focus on equivalence criteria at the population level to compare on- and off-lattice models. In particular, we define prototypic off- and on-lattice models of polar and apolar alignment, and show how to obtain an on-lattice from an off-lattice model of velocity alignment. By characterizing the behaviour and dynamical description of collective migration models at the macroscopic level, we suggest the type of phase transitions and possible patterns in the approximative macroscopic partial differential equation descriptions as informative equivalence criteria between on- and off-lattice models. This article is part of the theme issue 'Multi-scale analysis and modelling of collective migration in biological systems'.


Assuntos
Movimento Celular , Modelos Biológicos
8.
Nat Commun ; 10(1): 1787, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30992437

RESUMO

The identity and unique capacity of cancer stem cells (CSC) to drive tumor growth and resistance have been challenged in brain tumors. Here we report that cells expressing CSC-associated cell membrane markers in Glioblastoma (GBM) do not represent a clonal entity defined by distinct functional properties and transcriptomic profiles, but rather a plastic state that most cancer cells can adopt. We show that phenotypic heterogeneity arises from non-hierarchical, reversible state transitions, instructed by the microenvironment and is predictable by mathematical modeling. Although functional stem cell properties were similar in vitro, accelerated reconstitution of heterogeneity provides a growth advantage in vivo, suggesting that tumorigenic potential is linked to intrinsic plasticity rather than CSC multipotency. The capacity of any given cancer cell to reconstitute tumor heterogeneity cautions against therapies targeting CSC-associated membrane epitopes. Instead inherent cancer cell plasticity emerges as a novel relevant target for treatment.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/genética , Plasticidade Celular/efeitos dos fármacos , Glioblastoma/genética , Microambiente Tumoral/efeitos dos fármacos , Animais , Antineoplásicos Alquilantes/uso terapêutico , Biópsia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Plasticidade Celular/genética , Estudos de Coortes , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Resultado do Tratamento , Microambiente Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Front Oncol ; 8: 668, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687642

RESUMO

The development of cancer is a multistep process in which cells increase in malignancy through progressive alterations. Such altered cells compete with wild-type cells and have to establish within a tissue in order to induce tumor formation. The range of this competition and the tumor-originating cell type which acquires the first alteration is unknown for most human tissues, mainly because the involved processes are hardly observable, aggravating an understanding of early tumor development. On the tissue scale, one observes different progression types, namely with and without detectable benign precursor stages. Human epidemiological data on the ratios of the two progression types exhibit large differences between cancers. The idea of this study is to utilize data of the ratios of progression types in human cancers to estimate the homeostatic range of competition in human tissues. This homeostatic competition range can be interpreted as necessary numbers of altered cells to induce tumor formation on the tissue scale. For this purpose, we develop a cell-based stochastic model which is calibrated with newly-interpreted human epidemiological data. We find that the number of tumor cells which inevitably leads to later tumor formation is surprisingly small compared to the overall tumor and largely depends on the human tissue type. This result points toward the existence of a tissue-specific tumor-originating niche in which the fate of tumor development is decided early and long before a tumor becomes detectable. Moreover, our results suggest that the fixation of tumor cells in the tumor-originating niche triggers new processes which accelerate tumor growth after normal tissue homeostasis is voided. Our estimate for the human colon agrees well with the size of the stem cell niche in colonic crypts. For other tissues, our results might aid to identify the tumor-originating cell type. For instance, data on primary and secondary glioblastoma suggest that the tumors originate from a cell type competing in a range of 300 - 1,900 cells.

10.
J R Soc Interface ; 14(135)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28978744

RESUMO

The polarization of cells and tissues is fundamental for tissue morphogenesis during biological development and regeneration. A deeper understanding of biological polarity pattern formation can be gained from the consideration of pattern reorganization in response to an opposing instructive cue, which we here consider using the example of experimentally inducible body axis inversions in planarian flatworms. We define a dynamically diluted alignment model linking three processes: entrainment of cell polarity by a global signal, local cell-cell coupling aligning polarity among neighbours, and cell turnover replacing polarized cells by initially unpolarized cells. We show that a persistent global orienting signal determines the final mean polarity orientation in this stochastic model. Combining numerical and analytical approaches, we find that neighbour coupling retards polarity pattern reorganization, whereas cell turnover accelerates it. We derive a formula for an effective neighbour coupling strength integrating both effects and find that the time of polarity reorganization depends linearly on this effective parameter and no abrupt transitions are observed. This allows us to determine neighbour coupling strengths from experimental observations. Our model is related to a dynamic 8-Potts model with annealed site-dilution and makes testable predictions regarding the polarization of dynamic systems, such as the planarian epithelium.


Assuntos
Polaridade Celular/fisiologia , Modelos Biológicos , Planárias/fisiologia , Animais , Epitélio/fisiologia , Planárias/citologia
11.
Sci Rep ; 7(1): 9237, 2017 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-28835679

RESUMO

During tissue invasion individual tumor cells exhibit two interconvertible migration modes, namely mesenchymal and amoeboid migration. The cellular microenvironment triggers the switch between both modes, thereby allowing adaptation to dynamic conditions. It is, however, unclear if this amoeboid-mesenchymal migration plasticity contributes to a more effective tumor invasion. We address this question with a mathematical model, where the amoeboid-mesenchymal migration plasticity is regulated in response to local extracellular matrix resistance. Our numerical analysis reveals that extracellular matrix structure and presence of a chemotactic gradient are key determinants of the model behavior. Only in complex microenvironments, if the extracellular matrix is highly heterogeneous and a chemotactic gradient directs migration, the amoeboid-mesenchymal migration plasticity allows a more widespread invasion compared to the non-switching amoeboid and mesenchymal modes. Importantly, these specific conditions are characteristic for in vivo tumor invasion. Thus, our study suggests that in vitro systems aiming at unraveling the underlying molecular mechanisms of tumor invasion should take into account the complexity of the microenvironment by considering the combined effects of structural heterogeneities and chemical gradients on cell migration.


Assuntos
Modelos Biológicos , Neoplasias/patologia , Microambiente Tumoral , Algoritmos , Linhagem Celular Tumoral , Movimento Celular , Quimiotaxia , Simulação por Computador , Matriz Extracelular/metabolismo , Humanos , Invasividade Neoplásica , Neoplasias/metabolismo , Fenótipo
12.
Biol Direct ; 12(1): 18, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28800767

RESUMO

BACKGROUND: Cancer cell invasion, dissemination, and metastasis have been linked to an epithelial-mesenchymal transition (EMT) of individual tumour cells. During EMT, adhesion molecules like E-cadherin are downregulated and the decrease of cell-cell adhesion allows tumour cells to dissociate from the primary tumour mass. This complex process depends on intracellular cues that are subject to genetic and epigenetic variability, as well as extrinsic cues from the local environment resulting in a spatial heterogeneity in the adhesive phenotype of individual tumour cells. Here, we use a novel mathematical model to study how adhesion heterogeneity, influenced by intrinsic and extrinsic factors, affects the dissemination of tumour cells from an epithelial cell population. The model is a multiscale cellular automaton that couples intracellular adhesion receptor regulation with cell-cell adhesion. RESULTS: Simulations of our mathematical model indicate profound effects of adhesion heterogeneity on tumour cell dissemination. In particular, we show that a large variation of intracellular adhesion receptor concentrations in a cell population reinforces cell dissemination, regardless of extrinsic cues mediated through the local cell density. However, additional control of adhesion receptor concentration through the local cell density, which can be assumed in healthy cells, weakens the effect. Furthermore, we provide evidence that adhesion heterogeneity can explain the remarkable differences in adhesion receptor concentrations of epithelial and mesenchymal phenotypes observed during EMT and might drive early dissemination of tumour cells. CONCLUSIONS: Our results suggest that adhesion heterogeneity may be a universal trigger to reinforce cell dissemination in epithelial cell populations. This effect can be at least partially compensated by a control of adhesion receptor regulation through neighbouring cells. Accordingly, our findings explain how both an increase in intra-tumour adhesion heterogeneity and the loss of control through the local environment can promote tumour cell dissemination. REVIEWERS: This article was reviewed by Hanspeter Herzel, Thomas Dandekar and Marek Kimmel.


Assuntos
Adesão Celular , Modelos Teóricos , Movimento Celular , Simulação por Computador , Transição Epitelial-Mesenquimal , Invasividade Neoplásica , Metástase Neoplásica/patologia , Neoplasias/patologia , Transdução de Sinais
13.
Bioinform Biol Insights ; 11: 1177932217712241, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28659714

RESUMO

Many normal and cancerous cell lines exhibit a stable composition of cells in distinct states which can, e.g., be defined on the basis of cell surface markers. There is evidence that such an equilibrium is associated with stochastic transitions between distinct states. Quantifying these transitions has the potential to better understand cell lineage compositions. We introduce CellTrans, an R package to quantify stochastic cell state transitions from cell state proportion data from fluorescence-activated cell sorting and flow cytometry experiments. The R package is based on a mathematical model in which cell state alterations occur due to stochastic transitions between distinct cell states whose rates only depend on the current state of a cell. CellTrans is an automated tool for estimating the underlying transition probabilities from appropriately prepared data. We point out potential analytical challenges in the quantification of these cell transitions and explain how CellTrans handles them. The applicability of CellTrans is demonstrated on publicly available data on the evolution of cell state compositions in cancer cell lines. We show that CellTrans can be used to (1) infer the transition probabilities between different cell states, (2) predict cell line compositions at a certain time, (3) predict equilibrium cell state compositions, and (4) estimate the time needed to reach this equilibrium. We provide an implementation of CellTrans in R, freely available via GitHub (https://github.com/tbuder/CellTrans).

14.
PLoS Comput Biol ; 11(12): e1004662, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26658166

RESUMO

Pilocytic astrocytoma (PA) is the most common brain tumor in children. This tumor is usually benign and has a good prognosis. Total resection is the treatment of choice and will cure the majority of patients. However, often only partial resection is possible due to the location of the tumor. In that case, spontaneous regression, regrowth, or progression to a more aggressive form have been observed. The dependency between the residual tumor size and spontaneous regression is not understood yet. Therefore, the prognosis is largely unpredictable and there is controversy regarding the management of patients for whom complete resection cannot be achieved. Strategies span from pure observation (wait and see) to combinations of surgery, adjuvant chemotherapy, and radiotherapy. Here, we introduce a mathematical model to investigate the growth and progression behavior of PA. In particular, we propose a Markov chain model incorporating cell proliferation and death as well as mutations. Our model analysis shows that the tumor behavior after partial resection is essentially determined by a risk coefficient γ, which can be deduced from epidemiological data about PA. Our results quantitatively predict the regression probability of a partially resected benign PA given the residual tumor size and lead to the hypothesis that this dependency is linear, implying that removing any amount of tumor mass will improve prognosis. This finding stands in contrast to diffuse malignant glioma where an extent of resection threshold has been experimentally shown, below which no benefit for survival is expected. These results have important implications for future therapeutic studies in PA that should include residual tumor volume as a prognostic factor.


Assuntos
Astrocitoma/fisiopatologia , Neoplasias Encefálicas/fisiopatologia , Modelos Biológicos , Modelos Estatísticos , Regressão Neoplásica Espontânea/fisiopatologia , Apoptose/genética , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Proliferação de Células/genética , Simulação por Computador , Humanos , Mutação/genética , Regressão Neoplásica Espontânea/patologia , Neoplasia Residual
15.
PLoS Comput Biol ; 11(9): e1004366, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26335202

RESUMO

Tumor cells develop different strategies to cope with changing microenvironmental conditions. A prominent example is the adaptive phenotypic switching between cell migration and proliferation. While it has been shown that the migration-proliferation plasticity influences tumor spread, it remains unclear how this particular phenotypic plasticity affects overall tumor growth, in particular initiation and persistence. To address this problem, we formulate and study a mathematical model of spatio-temporal tumor dynamics which incorporates the microenvironmental influence through a local cell density dependence. Our analysis reveals that two dynamic regimes can be distinguished. If cell motility is allowed to increase with local cell density, any tumor cell population will persist in time, irrespective of its initial size. On the contrary, if cell motility is assumed to decrease with respect to local cell density, any tumor population below a certain size threshold will eventually extinguish, a fact usually termed as Allee effect in ecology. These results suggest that strategies aimed at modulating migration are worth to be explored as alternatives to those mainly focused at keeping tumor proliferation under control.


Assuntos
Modelos Biológicos , Processos Neoplásicos , Microambiente Tumoral/fisiologia , Contagem de Células , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Biologia Computacional , Simulação por Computador
16.
Bull Math Biol ; 77(4): 660-97, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25894920

RESUMO

Collective dynamics of migrating cell populations drive key processes in tissue formation and maintenance under normal and diseased conditions. Collective cell behavior at the tissue level is typically characterized by considering cell density patterns such as clusters and moving cell fronts. However, there are also important observables of collective dynamics related to individual cell behavior. In particular, individual cell trajectories are footprints of emergent behavior in populations of migrating cells. Lattice-gas cellular automata (LGCA) have proven successful to model and analyze collective behavior arising from interactions of migrating cells. There are well-established methods to analyze cell density patterns in LGCA models. Although LGCA dynamics are defined by cell-based rules, individual cells are not distinguished. Therefore, individual cell trajectories cannot be analyzed in LGCA so far. Here, we extend the classical LGCA framework to allow labeling and tracking of individual cells. We consider cell number conserving LGCA models of migrating cell populations where cell interactions are regulated by local cell density and derive stochastic differential equations approximating individual cell trajectories in LGCA. This result allows the prediction of complex individual cell trajectories emerging in LGCA models and is a basis for model-experiment comparisons at the individual cell level.


Assuntos
Movimento Celular/fisiologia , Modelos Biológicos , Animais , Contagem de Células , Microambiente Celular , Simulação por Computador , Humanos , Conceitos Matemáticos , Processos Estocásticos
17.
PLoS One ; 9(4): e93207, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24690888

RESUMO

The liver is a multi-functional organ that regulates major physiological processes and that possesses a remarkable regeneration capacity. After loss of functional liver mass the liver grows back to its original, individual size through hepatocyte proliferation and apoptosis. How does a single hepatocyte 'know' when the organ has grown to its final size? This work considers the initial growth phase of liver regeneration after partial hepatectomy in which the mass is restored. There are strong and valid arguments that the trigger of proliferation after partial hepatectomy is mediated through the portal blood flow. It remains unclear, if either or both the concentration of metabolites in the blood or the shear stress are crucial to hepatocyte proliferation and liver size control. A cell-based mathematical model is developed that helps discriminate the effects of these two potential triggers. Analysis of the mathematical model shows that a metabolic load and a hemodynamical hypothesis imply different feedback mechanisms at the cellular scale. The predictions of the developed mathematical model are compared to experimental data in rats. The assumption that hepatocytes are able to buffer the metabolic load leads to a robustness against short-term fluctuations of the trigger which can not be achieved with a purely hemodynamical trigger.


Assuntos
Hepatócitos/fisiologia , Regeneração Hepática/fisiologia , Fígado/anatomia & histologia , Fígado/fisiologia , Algoritmos , Animais , Apoptose , Proliferação de Células , Simulação por Computador , Retroalimentação Fisiológica , Hepatectomia , Modelos Animais , Modelos Teóricos , Tamanho do Órgão , Ratos
18.
PLoS One ; 7(9): e42852, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984409

RESUMO

Cellular Potts models (CPMs) are used as a modeling framework to elucidate mechanisms of biological development. They allow a spatial resolution below the cellular scale and are applied particularly when problems are studied where multiple spatial and temporal scales are involved. Despite the increasing usage of CPMs in theoretical biology, this model class has received little attention from mathematical theory. To narrow this gap, the CPMs are subjected to a theoretical study here. It is asked to which extent the updating rules establish an appropriate dynamical model of intercellular interactions and what the principal behavior at different time scales characterizes. It is shown that the longtime behavior of a CPM is degenerate in the sense that the cells consecutively die out, independent of the specific interdependence structure that characterizes the model. While CPMs are naturally defined on finite, spatially bounded lattices, possible extensions to spatially unbounded systems are explored to assess to which extent spatio-temporal limit procedures can be applied to describe the emergent behavior at the tissue scale. To elucidate the mechanistic structure of CPMs, the model class is integrated into a general multiscale framework. It is shown that the central role of the surface fluctuations, which subsume several cellular and intercellular factors, entails substantial limitations for a CPM's exploitation both as a mechanistic and as a phenomenological model.


Assuntos
Células/metabolismo , Modelos Biológicos , Morfogênese , Termodinâmica , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...