Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cells ; 10(7)2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34359888

RESUMO

Organoids retain the morphological and molecular patterns of their tissue of origin, are self-organizing, relatively simple to handle and accessible to genetic engineering. Thus, they represent an optimal tool for studying the mechanisms of tissue maintenance and aging. Long-term expansion under standard growth conditions, however, is accompanied by changes in the growth pattern and kinetics. As a potential explanation of these alterations, epigenetic drifts in organoid culture have been suggested. Here, we studied histone tri-methylation at lysine 4 (H3K4me3) and 27 (H3K27me3) and transcriptome profiles of intestinal organoids derived from mismatch repair (MMR)-deficient and control mice and cultured for 3 and 20 weeks and compared them with data on their tissue of origin. We found that, besides the expected changes in short-term culture, the organoids showed profound changes in their epigenomes also during the long-term culture. The most prominent were epigenetic gene activation by H3K4me3 recruitment to previously unmodified genes and by H3K27me3 loss from originally bivalent genes. We showed that a long-term culture is linked to broad transcriptional changes that indicate an ongoing maturation and metabolic adaptation process. This process was disturbed in MMR-deficient mice, resulting in endoplasmic reticulum (ER) stress and Wnt activation. Our results can be explained in terms of a mathematical model assuming that epigenetic changes during a long-term culture involve DNA demethylation that ceases if the metabolic adaptation is disturbed.


Assuntos
Epigênese Genética , Técnicas de Cultura de Órgãos , Organoides/metabolismo , Adaptação Fisiológica/genética , Animais , Histonas/metabolismo , Camundongos , Fatores de Tempo , Transcrição Gênica
2.
Int J Mol Sci ; 21(6)2020 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-32178409

RESUMO

Aberrant DNA methylation in stem cells is a hallmark of aging and tumor development. Recently, we have suggested that promoter DNA hyper-methylation originates in DNA repair and that even successful DNA repair might confer this kind of epigenetic long-term change. Here, we ask for interrelations between promoter DNA methylation and histone modification changes observed in the intestine weeks after irradiation and/or following Msh2 loss. We focus on H3K4me3 recruitment to the promoter of H3K27me3 target genes. By RNA- and histone ChIP-sequencing, we demonstrate that this recruitment occurs without changes of the average gene transcription and does not involve H3K9me3. Applying a mathematical model of epigenetic regulation of transcription, we show that the recruitment can be explained by stronger DNA binding of H3K4me3 and H3K27me3 histone methyl-transferases as a consequence of lower DNA methylation. This scenario implicates stable transcription despite of H3K4me3 recruitment, in agreement with our RNA-seq data. Following several kinds of stress, including moderate irradiation, stress-sensitive intestinal stem cell (ISCs) are known to become replaced by more resistant populations. Our simulation results suggest that the stress-resistant ISCs are largely protected against promoter hyper-methylation of H3K27me3 target genes.


Assuntos
Metilação de DNA/genética , DNA/genética , Histonas/genética , Intestinos/fisiologia , Regiões Promotoras Genéticas/genética , Células-Tronco/fisiologia , Animais , Epigênese Genética/genética , Código das Histonas/genética , Camundongos
3.
Clin Epigenetics ; 11(1): 65, 2019 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-31029155

RESUMO

BACKGROUND: Mismatch repair (MMR)-deficiency increases the risk of colorectal tumorigenesis. To determine whether the tumors develop on a normal or disturbed epigenetic background and how radiation affects this, we quantified genome-wide histone H3 methylation profiles in macroscopic normal intestinal tissue of young radiated and untreated MMR-deficient VCMsh2LoxP/LoxP (Msh2-/-) mice months before tumor onset. RESULTS: Histone H3 methylation increases in Msh2-/- compared to control Msh2+/+ mice. Activating H3K4me3 and H3K36me3 histone marks frequently accumulate at genes that are H3K27me3 or H3K4me3 modified in Msh2+/+ mice, respectively. The genes recruiting H3K36me3 enrich in gene sets associated with DNA repair, RNA processing, and ribosome biogenesis that become transcriptionally upregulated in the developing tumors. A similar epigenetic effect is present in Msh2+/+ mice 4 weeks after a single-radiation hit, whereas radiation of Msh2-/- mice left their histone methylation profiles almost unchanged. CONCLUSIONS: MMR deficiency results in genome-wide changes in histone H3 methylation profiles preceding tumor development. Similar changes constitute a persistent epigenetic signature of radiation-induced DNA damage.


Assuntos
Redes Reguladoras de Genes/efeitos da radiação , Histonas/metabolismo , Neoplasias Intestinais/etiologia , Intestinos/efeitos da radiação , Proteína 2 Homóloga a MutS/genética , Idoso , Animais , Estudos de Casos e Controles , Sequenciamento de Cromatina por Imunoprecipitação , Modelos Animais de Doenças , Epigênese Genética/efeitos da radiação , Feminino , Humanos , Neoplasias Intestinais/genética , Intestinos/química , Masculino , Camundongos , Sequenciamento Completo do Genoma
4.
BMC Cardiovasc Disord ; 18(1): 65, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29653511

RESUMO

BACKGROUND: Chemerin is an adipokine which plays a crucial role in atherosclerosis. Here, we examined whether circulating chemerin is enhanced in patients with advanced carotid stenosis. METHODS: Chemerin was quantified in 178 patients prior to carotid end arterectomy (CEA) and in age- and gender-matched controls (n = 163). Chemerin levels were related to anthropometric, clinical and metabolic characteristics of the patients. RESULTS: Chemerin levels were higher in patients compared to controls (p <  0.001). Chemerin correlated to parameters associated with inflammation such as C-reactive protein (CRP, p <  0.001), leukocyte blood count (p <  0.001) and circulating TNF-α (p = 0.004) in the patients. Chemerin levels did not differ between asymptomatic (n = 93) and symptomatic patients who experienced an ischemic event within 6 months prior to CEA (n = 85). However, in the case of high-grade carotid stenosis (≥ 90%), chemerin levels were higher in symptomatic (n = 44) compared to asymptomatic patients (n = 41, p = 0.014). Chemerin was increased in patients with (n = 50) compared to patients without (n = 128) coronary artery disease (CAD, p = 0.002). A high level of chemerin increases the risk for CAD in patients (p = 0.0013). CONCLUSIONS: Circulating chemerin is increased and correlates to inflammatory parameters in patients with advanced carotid stenosis.


Assuntos
Estenose das Carótidas/sangue , Quimiocinas/sangue , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Idoso , Biomarcadores/sangue , Estenose das Carótidas/diagnóstico por imagem , Estudos de Casos e Controles , Feminino , Humanos , Mediadores da Inflamação/sangue , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Regulação para Cima
5.
Genes (Basel) ; 9(1)2018 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-29303998

RESUMO

Aberrant DNA methylation in stem cells is a hallmark of aging and tumor development. Here, we explore whether and how DNA damage repair might impact on these time-dependent changes, in particular in proliferative intestinal stem cells. We introduce a 3D multiscale computer model of intestinal crypts enabling simulation of aberrant DNA and histone methylation of gene promoters during aging. We assume histone state-dependent activity of de novo DNA methyltransferases (DNMTs) and methylation-dependent binding of maintenance DNMTs to CpGs. We simulate aging with and without repeated DNA repair. Motivated by recent findings on the histone demethylase KDM2b, we consider that DNA repair is associated with chromatin opening and improved recruitment of de novo DNMTs. Our results suggest that methylation-dependent binding of maintenance DNMTs to CpGs, establishing bistable DNA methylation states, is a prerequisite to promoter hyper-methylation following DNA repair. With this, the transient increase in de novo DNMT activity during repair can induce switches from low to high methylation states. These states remain stable after repair, leading to an epigenetic drift. The switches are most frequent in genes with H3K27me3 modified promoters. Our model provides a mechanistic explanation on how even successful DNA repair might confer long term changes of the epigenome.

6.
Dev Biol ; 433(2): 254-261, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29198564

RESUMO

Intestinal stem cells (ISCs) require well-defined signals from their environment in order to carry out their specific functions. Most of these signals are provided by neighboring cells that form a stem cell niche, whose shape and cellular composition self-organize. Major features of this self-organization can be studied in ISC-derived organoid culture. In this system, manipulation of essential pathways of stem cell maintenance and differentiation results in well-described growth phenotypes. We here provide an individual cell-based model of intestinal organoids that enables a mechanistic explanation of the observed growth phenotypes. In simulation studies of the 3D structure of expanding organoids, we investigate interdependences between Wnt- and Notch-signaling which control the shape of the stem cell niche and, thus, the growth pattern of the organoids. Similar to in vitro experiments, changes of pathway activities alter the cellular composition of the organoids and, thereby, affect their shape. Exogenous Wnt enforces transitions from branched into a cyst-like growth pattern; known to occur spontaneously during long term organoid expansion. Based on our simulation results, we predict that the cyst-like pattern is associated with biomechanical changes of the cells which assign them a growth advantage. The results suggest ongoing stem cell adaptation to in vitro conditions during long term expansion by stabilizing Wnt-activity. Our study exemplifies the potential of individual cell-based modeling in unraveling links between molecular stem cell regulation and 3D growth of tissues. This kind of modeling combines experimental results in the fields of stem cell biology and cell biomechanics constituting a prerequisite for a better understanding of tissue regeneration as well as developmental processes.


Assuntos
Simulação por Computador , Intestinos/citologia , Modelos Biológicos , Organoides/crescimento & desenvolvimento , Células-Tronco/fisiologia , Animais , Apoptose , Fenômenos Biomecânicos , Adesão Celular , Forma Celular , Camundongos , Camundongos Endogâmicos C57BL , Polímeros , Receptores Notch/fisiologia , Regeneração/fisiologia , Nicho de Células-Tronco , Técnicas de Cultura de Tecidos , Proteínas Wnt/fisiologia , Via de Sinalização Wnt
7.
J Pathol ; 243(2): 242-254, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28727142

RESUMO

Colorectal cancer (CRC) arising in Lynch syndrome (LS) comprises tumours with constitutional mutations in DNA mismatch repair genes. There is still a lack of whole-genome and transcriptome studies of LS-CRC to address questions about similarities and differences in mutation and gene expression characteristics between LS-CRC and sporadic CRC, about the molecular heterogeneity of LS-CRC, and about specific mechanisms of LS-CRC genesis linked to dysfunctional mismatch repair in LS colonic mucosa and the possible role of immune editing. Here, we provide a first molecular characterization of LS tumours and of matched tumour-distant reference colonic mucosa based on whole-genome DNA-sequencing and RNA-sequencing analyses. Our data support two subgroups of LS-CRCs, G1 and G2, whereby G1 tumours show a higher number of somatic mutations, a higher amount of microsatellite slippage, and a different mutation spectrum. The gene expression phenotypes support this difference. Reference mucosa of G1 shows a strong immune response associated with the expression of HLA and immune checkpoint genes and the invasion of CD4+ T cells. Such an immune response is not observed in LS tumours, G2 reference and normal (non-Lynch) mucosa, and sporadic CRC. We hypothesize that G1 tumours are edited for escape from a highly immunogenic microenvironment via loss of HLA presentation and T-cell exhaustion. In contrast, G2 tumours seem to develop in a less immunogenic microenvironment where tumour-promoting inflammation parallels tumourigenesis. Larger studies on non-neoplastic mucosa tissue of mutation carriers are required to better understand the early phases of emerging tumours. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Neoplasias Colorretais/genética , Mutação/genética , Antígenos de Neoplasias/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais Hereditárias sem Polipose/imunologia , Expressão Gênica/genética , Genes Neoplásicos/genética , Genoma Humano/genética , Humanos , Imunidade Celular , Fenótipo , Recidiva , Transcriptoma/genética , Evasão Tumoral/genética , Evasão Tumoral/imunologia
8.
Int J Mol Sci ; 18(5)2017 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-28513551

RESUMO

Bivalent genes are frequently associated with developmental and lineage specification processes. Resolving their bivalency enables fast changes in their expression, which potentially can trigger cell fate decisions. Here, we provide a theoretical model of bivalency that allows for predictions on the occurrence, stability and regulatory capacity of this prominent modification state. We suggest that bivalency enables balanced gene expression heterogeneity that constitutes a prerequisite of robust lineage priming in somatic stem cells. Moreover, we demonstrate that interactions between the histone and DNA methylation machineries together with the proliferation activity control the stability of the bivalent state and can turn it into an unmodified state. We suggest that deregulation of these interactions underlies cell transformation processes as associated with acute myeloid leukemia (AML) and provide a model of AML blast formation following deregulation of the Ten-eleven Translocation (TET) pathway.


Assuntos
Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes , Modelos Teóricos , Algoritmos , Simulação por Computador , Metilação de DNA , Epigênese Genética , Histonas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Modelos Biológicos , Transcrição Gênica
9.
J R Soc Interface ; 13(117)2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27097654

RESUMO

Pluripotent mouse embryonic stem cells (mESCs) show heterogeneous expression levels of transcription factors (TFs) involved in pluripotency regulation, among them Nanog and Rex1. The expression of both TFs can change dynamically between states of high and low activity, correlating with the cells' capacity for self-renewal. Stochastic fluctuations as well as sustained oscillations in gene expression are possible mechanisms to explain this behaviour, but the lack of suitable data hampered their clear distinction. Here, we present a systems biology approach in which novel experimental data on TF heterogeneity is complemented by an agent-based model of mESC self-renewal. Because the model accounts for intracellular interactions, cell divisions and heredity structures, it allows for evaluating the consistency of the proposed mechanisms with data on population growth and on TF dynamics after cell sorting. Our model-based analysis revealed that a bistable, noise-driven network model fulfils the minimal requirements to consistently explain Nanog and Rex1 expression dynamics in heterogeneous and sorted mESC populations. Moreover, we studied the impact of TF-related proliferation capacities on the frequency of state transitions and demonstrate that cellular genealogies can provide insights into the heredity structures of mESCs.


Assuntos
Regulação da Expressão Gênica/fisiologia , Modelos Biológicos , Células-Tronco Embrionárias Murinas , Proteína Homeobox Nanog/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia , Animais , Linhagem Celular , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo
10.
Development ; 142(13): 2250-60, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26130756

RESUMO

The maintenance of pluripotency in embryonic stem cells (ESCs), its loss during lineage specification or its re-induction to generate induced pluripotent stem cells are central topics in stem cell biology. To uncover the molecular basis and the design principles of pluripotency control, a multitude of experimental, but also an increasing number of computational, studies have been published. Here, we consider recent reports that apply computational or mathematical modelling approaches to describe the regulatory processes that underlie cell fate decisions in mouse ESCs. We summarise the principles, the strengths and potentials but also the limitations of different computational strategies.


Assuntos
Linhagem da Célula , Simulação por Computador , Células-Tronco Embrionárias/citologia , Modelos Biológicos , Animais , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Humanos , Células-Tronco Pluripotentes
11.
Cytometry A ; 87(6): 481-90, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25605123

RESUMO

Pluripotent embryonic stem cells (ESCs) have the potential to differentiate into cells of all three germ layers. This unique property has been extensively studied on the intracellular, transcriptional level. However, ESCs typically form clusters of cells with distinct size and shape, and establish spatial structures that are vital for the maintenance of pluripotency. Even though it is recognized that the cells' arrangement and local interactions play a role in fate decision processes, the relations between transcriptional and spatial patterns have not yet been studied. We present a systems biology approach which combines live-cell imaging, quantitative image analysis, and multiscale, mathematical modeling of ESC growth. In particular, we develop quantitative measures of the morphology and of the spatial clustering of ESCs with different expression levels and apply them to images of both in vitro and in silico cultures. Using the same measures, we are able to compare model scenarios with different assumptions on cell-cell adhesions and intercellular feedback mechanisms directly with experimental data. Applying our methodology to microscopy images of cultured ESCs, we demonstrate that the emerging colonies are highly variable regarding both morphological and spatial fluorescence patterns. Moreover, we can show that most ESC colonies contain only one cluster of cells with high self-renewing capacity. These cells are preferentially located in the interior of a colony structure. The integrated approach combining image analysis with mathematical modeling allows us to reveal potential transcription factor related cellular and intercellular mechanisms behind the emergence of observed patterns that cannot be derived from images directly.


Assuntos
Movimento Celular/fisiologia , Células-Tronco Embrionárias/citologia , Processamento de Imagem Assistida por Computador/métodos , Modelos Teóricos , Células-Tronco Pluripotentes/citologia , Animais , Adesão Celular/fisiologia , Diferenciação Celular , Células Cultivadas , Biologia Computacional/métodos , Simulação por Computador , Meios de Cultura/farmacologia , Fator Inibidor de Leucemia/farmacologia , Camundongos , Microscopia de Fluorescência , Biologia de Sistemas/métodos
12.
PLoS One ; 9(3): e92496, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24643025

RESUMO

Mouse embryonic stem cells (mESCs) can be maintained in a proliferative and undifferentiated state over many passages (self-renewal) while retaining the potential to give rise to every cell type of the organism (pluripotency). Autocrine FGF4/Erk signalling has been identified as a major stimulus for fate decisions and lineage commitment in these cells. Recent findings on serum-free culture conditions with specific inhibitors (known as 2i) demonstrate that the inhibition of this pathway reduces transcription factor heterogeneity and is vital to maintain ground state pluripotency of mESCs. We suggest a novel mathematical model to explicitly integrate FGF4/Erk signalling into an interaction network of key pluripotency factors (namely Oct4, Sox2, Nanog and Rex1). The envisaged model allows to explore whether and how proposed mechanisms and feedback regulations can account for different expression patterns in mESC cultures. We demonstrate that an FGF4/Erk-mediated negative feedback is sufficient to induce molecular heterogeneity with respect to Nanog and Rex1 expression and thus critically regulates the propensity for differentiation and the loss of pluripotency. Furthermore, we compare simulation results on the transcription factor dynamics in different self-renewing states and during differentiation with experimental data on a Rex1GFPd2 reporter cell line using flow cytometry and qRT-PCR measurements. Concluding from our results we argue that interaction between FGF4/Erk signalling and Nanog expression qualifies as a key mechanism to manipulate mESC pluripotency. In particular, we infer that ground state pluripotency under 2i is achieved by shifting stable expression pattern of Nanog from a bistable into a monostable regulation impeding stochastic state transitions. Furthermore, we derive testable predictions on altering the degree of Nanog heterogeneity and on the frequency of state transitions in LIF/serum conditions to challenge our model assumptions.


Assuntos
Células-Tronco Embrionárias/fisiologia , Modelos Biológicos , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Simulação por Computador , Meios de Cultura Livres de Soro , Proteínas de Homeodomínio/metabolismo , Fator Inibidor de Leucemia/fisiologia , Camundongos , Proteína Homeobox Nanog , Fenótipo , Estabilidade Proteica , Transdução de Sinais
13.
Ann Clin Biochem ; 50(Pt 1): 62-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23065528

RESUMO

BACKGROUND: Measurements of plasma normetanephrine and metanephrine provide a useful diagnostic test for phaeochromocytoma, but this depends on appropriate reference intervals. Upper cut-offs set too high compromise diagnostic sensitivity, whereas set too low, false-positives are a problem. This study aimed to establish optimal reference intervals for plasma normetanephrine and metanephrine. METHODS: Blood samples were collected in the supine position from 1226 subjects, aged 5-84 y, including 116 children, 575 normotensive and hypertensive adults and 535 patients in whom phaeochromocytoma was ruled out. Reference intervals were examined according to age and gender. Various models were examined to optimize upper cut-offs according to estimates of diagnostic sensitivity and specificity in a separate validation group of 3888 patients tested for phaeochromocytoma, including 558 with confirmed disease. RESULTS: Plasma metanephrine, but not normetanephrine, was higher (P < 0.001) in men than in women, but reference intervals did not differ. Age showed a positive relationship (P < 0.0001) with plasma normetanephrine and a weaker relationship (P = 0.021) with metanephrine. Upper cut-offs of reference intervals for normetanephrine increased from 0.47 nmol/L in children to 1.05 nmol/L in subjects over 60 y. A curvilinear model for age-adjusted compared with fixed upper cut-offs for normetanephrine, together with a higher cut-off for metanephrine (0.45 versus 0.32 nmol/L), resulted in a substantial gain in diagnostic specificity from 88.3% to 96.0% with minimal loss in diagnostic sensitivity from 93.9% to 93.6%. CONCLUSIONS: These data establish age-adjusted cut-offs of reference intervals for plasma normetanephrine and optimized cut-offs for metanephrine useful for minimizing false-positive results.


Assuntos
Neoplasias das Glândulas Suprarrenais/sangue , Hipertensão/sangue , Metanefrina/sangue , Normetanefrina/sangue , Feocromocitoma/sangue , Adolescente , Neoplasias das Glândulas Suprarrenais/complicações , Neoplasias das Glândulas Suprarrenais/diagnóstico , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Criança , Pré-Escolar , Reações Falso-Positivas , Feminino , Humanos , Hipertensão/complicações , Hipertensão/diagnóstico , Masculino , Pessoa de Meia-Idade , Feocromocitoma/complicações , Feocromocitoma/diagnóstico , Valores de Referência , Sensibilidade e Especificidade , Fatores Sexuais
15.
PLoS One ; 5(6): e11238, 2010 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-20574542

RESUMO

The expression of the transcription factors Oct4, Sox2, and Nanog is commonly associated with pluripotency of mouse embryonic stem (ES) cells. However, recent observations suggest that ES cell populations are heterogeneous with respect to the expression of Nanog and that individual ES cells reversibly change their Nanog expression level. Furthermore, it has been shown that cells exhibiting a low Nanog level are more likely to undergo differentiation. Applying a novel mathematical transcription factor network model we explore mechanisms and feedback regulations to describe the observed variation of the Nanog levels in mouse ES cells. In particular we show that these variations can occur under different assumptions yielding similar experimental characteristics. Based on model predictions we propose experimental strategies to distinguish between these explanations. Concluding from our results we argue that the heterogeneity with respect to the Nanog concentrations is most likely a functional element to control the differentiation propensity of an ES cell population. Furthermore, we provide a conceptual framework that consistently explains Nanog variability and a potential "gate-keeper" function of Nanog expression with respect to the control of ES cell differentiation.


Assuntos
Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/metabolismo , Modelos Biológicos , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Retroalimentação Fisiológica , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição SOXB1/metabolismo , Processos Estocásticos
16.
Bull Math Biol ; 71(3): 602-26, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19101772

RESUMO

Previously, we have modeled hematopoietic stem cell organization by a stochastic, single cell-based approach. Applications to different experimental systems demonstrated that this model consistently explains a broad variety of in vivo and in vitro data. A major advantage of the agent-based model (ABM) is the representation of heterogeneity within the hematopoietic stem cell population. However, this advantage comes at the price of time-consuming simulations if the systems become large. One example in this respect is the modeling of disease and treatment dynamics in patients with chronic myeloid leukemia (CML), where the realistic number of individual cells to be considered exceeds 10(6). To overcome this deficiency, without losing the representation of the inherent heterogeneity of the stem cell population, we here propose to approximate the ABM by a system of partial differential equations (PDEs). The major benefit of such an approach is its independence from the size of the system. Although this mean field approach includes a number of simplifying assumptions compared to the ABM, it retains the key structure of the model including the "age"-structure of stem cells. We show that the PDE model qualitatively and quantitatively reproduces the results of the agent-based approach.


Assuntos
Células-Tronco Hematopoéticas/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Modelos Biológicos , Humanos , Processos Estocásticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA