Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioengineering (Basel) ; 10(1)2022 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-36671623

RESUMO

The implementation of stem-cell-based organoid culture more than ten years ago started a development that created new avenues for diagnostic analyses and regenerative medicine. In parallel, computational modelling groups realized the potential of this culture system to support their theoretical approaches to study tissues in silico. These groups developed computational organoid models (COMs) that enabled testing consistency between cell biological data and developing theories of tissue self-organization. The models supported a mechanistic understanding of organoid growth and maturation and helped linking cell mechanics and tissue shape in general. What comes next? Can we use COMs as tools to complement the equipment of our biological and medical research? While these models already support experimental design, can they also quantitatively predict tissue behavior? Here, we review the current state of the art of COMs and discuss perspectives for their application.

2.
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
3.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34298973

RESUMO

Intestinal cylindrical growth peaks in mice a few weeks after birth, simultaneously with crypt fission activity. It nearly stops after weaning and cannot be reactivated later. Transgenic mice expressing Cd97/Adgre5 in the intestinal epithelium develop a mega-intestine with normal microscopic morphology in adult mice. Here, we demonstrate premature intestinal differentiation in Cd97/Adgre5 transgenic mice at both the cellular and molecular levels until postnatal day 14. Subsequently, the growth of the intestinal epithelium becomes activated and its maturation suppressed. These changes are paralleled by postnatal regulation of growth factors and by an increased expression of secretory cell markers, suggesting growth activation of non-epithelial tissue layers as the origin of enforced tissue growth. To understand postnatal intestinal growth mechanistically, we study epithelial fate decisions during this period with the use of a 3D individual cell-based computer model. In the model, the expansion of the intestinal stem cell (SC) population, a prerequisite for crypt fission, is largely independent of the tissue growth rate and is therefore not spontaneously adaptive. Accordingly, the model suggests that, besides the growth activation of non-epithelial tissue layers, the formation of a mega-intestine requires a released growth control in the epithelium, enabling accelerated SC expansion. The similar intestinal morphology in Cd97/Adgre5 transgenic and wild type mice indicates a synchronization of tissue growth and SC expansion, likely by a crypt density-controlled contact inhibition of growth of intestinal SC proliferation. The formation of a mega-intestine with normal microscopic morphology turns out to originate in changes of autonomous and conditional specification of the intestinal cell fate induced by the activation of Cd97/Adgre5.


Assuntos
Simulação por Computador , Mucosa Intestinal/crescimento & desenvolvimento , Intestino Delgado/crescimento & desenvolvimento , Modelos Biológicos , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo , Animais , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Receptores Acoplados a Proteínas G/genética
4.
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
5.
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
6.
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.

7.
Nat Cell Biol ; 20(1): 69-80, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29230016

RESUMO

To establish and maintain organ structure and function, tissues need to balance stem cell proliferation and differentiation rates and coordinate cell fate with position. By quantifying and modelling tissue stress and deformation in the mammalian epidermis, we find that this balance is coordinated through local mechanical forces generated by cell division and delamination. Proliferation within the basal stem/progenitor layer, which displays features of a jammed, solid-like state, leads to crowding, thereby locally distorting cell shape and stress distribution. The resulting decrease in cortical tension and increased cell-cell adhesion trigger differentiation and subsequent delamination, reinstating basal cell layer density. After delamination, cells establish a high-tension state as they increase myosin II activity and convert to E-cadherin-dominated adhesion, thereby reinforcing the boundary between basal and suprabasal layers. Our results uncover how biomechanical signalling integrates single-cell behaviours to couple proliferation, cell fate and positioning to generate a multilayered tissue.


Assuntos
Caderinas/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento , Mecanotransdução Celular , Miosina Tipo II/genética , Animais , Fenômenos Biomecânicos , Caderinas/metabolismo , Adesão Celular , Divisão Celular , Forma Celular , Embrião de Mamíferos , Células Epidérmicas/citologia , Células Epidérmicas/metabolismo , Epiderme/embriologia , Epiderme/metabolismo , Humanos , Microscopia Intravital , Camundongos , Camundongos Endogâmicos C57BL , Miosina Tipo II/metabolismo , Cultura Primária de Células
8.
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
9.
Epigenetics ; 12(10): 886-896, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28758855

RESUMO

In acute myeloid leukemia (AML) DNA hypermethylation of gene promoters is frequently observed and often correlates with a block of differentiation. Treatment of AML patients with DNA methyltransferase inhibitors results in global hypomethylation of genes and, thereby, can lead to a reactivation of the differentiation capability. Unfortunately, after termination of treatment both hypermethylation and differentiation block return in most cases. Here, we apply, for the first time, a computational model of epigenetic regulation of transcription to: i) provide a mechanistic understanding of the DNA (de-) methylation process in AML and; ii) improve DNA demethylation treatment strategies. By in silico simulation, we analyze promoter hypermethylation scenarios referring to DNMT dysfunction, decreased H3K4me3 and increased H3K27me3 modification activity, and accelerated cell proliferation. We quantify differences between these scenarios with respect to gene repression and activation. Moreover, we compare the scenarios regarding their response to DNMT inhibitor treatment alone and in combination with inhibitors of H3K27me3 histone methyltransferases and of H3K4me3 histone demethylases. We find that the different hypermethylation scenarios respond specifically to therapy, suggesting that failure of remission originates in patient-specific deregulation. We observe that inappropriate demethylation therapy can result even in enforced deregulation. As an example, our results suggest that application of high DNMT inhibitor concentration can induce unwanted global gene activation if hypermethylation originates in increased H3K27me3 modification. Our results underline the importance of a personalized therapy requiring knowledge about the patient-specific mechanism of epigenetic deregulation.


Assuntos
Desmetilação do DNA/efeitos dos fármacos , Metilação de DNA/genética , Inibidores Enzimáticos/uso terapêutico , Leucemia Mieloide Aguda/genética , Azacitidina/química , Azacitidina/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Biologia Computacional , Metilases de Modificação do DNA/antagonistas & inibidores , Metilases de Modificação do DNA/genética , Inibidores Enzimáticos/química , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/química , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/química , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Modelos Moleculares , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética
10.
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
11.
Stem Cells ; 35(3): 694-704, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27734598

RESUMO

The molecular mechanisms by which heterogeneity, a major characteristic of stem cells, is achieved are yet unclear. We here study the expression of the membrane stem cell antigen-1 (Sca-1) in mouse bone marrow mesenchymal stem cell (MSC) clones. We show that subpopulations with varying Sca-1 expression profiles regenerate the Sca-1 profile of the mother population within a few days. However, after extensive replication in vitro, the expression profiles shift to lower values and the regeneration time increases. Study of the promoter of Ly6a unravels that the expression level of Sca-1 is related to the promoter occupancy by the activating histone mark H3K4me3. We demonstrate that these findings can be consistently explained by a computational model that considers positive feedback between promoter H3K4me3 modification and gene transcription. This feedback implicates bistable epigenetic states which the cells occupy with an age-dependent frequency due to persistent histone (de-)modification. Our results provide evidence that MSC heterogeneity, and presumably that of other stem cells, is associated with bistable epigenetic states and suggest that MSCs are subject to permanent state fluctuations. Stem Cells 2017;35:694-704.


Assuntos
Envelhecimento/genética , Epigênese Genética , Células-Tronco Mesenquimais/metabolismo , Animais , Antígenos Ly/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular/genética , Proliferação de Células , Células Clonais , Perfilação da Expressão Gênica , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Modelos Genéticos , Regiões Promotoras Genéticas
12.
J R Soc Interface ; 13(121)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27534699

RESUMO

Three-dimensional (3D) computational tissue models can provide a comprehensive description of tissue dynamics at the molecular, cellular and tissue level. Moreover, they can support the development of hypotheses about cellular interactions and about synergies between major signalling pathways. We exemplify these capabilities by simulation of a 3D single-cell-based model of mouse small intestinal crypts. We analyse the impact of lineage specification, distribution and cellular lifespan on clonal competition and study effects of Notch- and Wnt activation on fixation of mutations within the tissue. Based on these results, we predict that experimentally observed synergistic effects between autonomous Notch- and Wnt signalling in triggering intestinal tumourigenesis originate in the suppression of Wnt-dependent secretory lineage specification by Notch, giving rise to an increased fixation probability of Wnt-activating mutations. Our study demonstrates that 3D computational tissue models can support a mechanistic understanding of long-term tissue dynamics under homeostasis and during transformation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/metabolismo , Modelos Biológicos , Células-Tronco/metabolismo , Via de Sinalização Wnt , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Humanos , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Intestinos/patologia , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Células-Tronco/patologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
13.
Biochem Soc Trans ; 42(3): 671-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24849236

RESUMO

The intestinal epithelium is permanently renewed during homoeostasis. Stable function of its stem cells is ensured by interaction with a specific tissue compartment, the so-called 'intestinal stem cell niche'. The essential regulatory principles of this niche are still under debate. In order to approach this question, we have introduced several single cell-based models of the spatiotemporal stem cell organization in murine intestinal crypts and organoids. In the present article, we provide a brief review of these models. Starting with pedigree models reproducing cell kinetics, over the last few years, we have successively improved these models by refining the biomechanical representation of the system and introducing environmentally controlled lineage specification. Our current models of the intestinal crypt are capable of linking a broad spectrum of experimental observations encompassing spatially confined cell proliferation, directed cell migration, multiple cell lineage decisions and clonal competition. Our model of intestinal organoids provides for the first time a description of a self-organizing intestinal stem cell niche. It suggests that this niche is established by secretory activity of specified cells and in addition requires a defined spatial organization, which sensitively depends on tissue biomechanics.


Assuntos
Biologia Computacional , Intestinos/citologia , Nicho de Células-Tronco , Animais , Mucosa Intestinal/citologia , Camundongos
14.
Aging Cell ; 13(2): 320-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24428552

RESUMO

During aging, a decline in stem cell function is observed in many tissues. This decline is accompanied by complex changes of the chromatin structure among them changes in histone modifications and DNA methylation which both affect transcription of a tissue-specific subset of genes. A mechanistic understanding of these age-associated processes, their interrelations and environmental dependence is currently lacking. Here, we discuss related questions on the molecular, cellular, and population level. We combine an individual cell-based model of stem cell populations with a model of epigenetic regulation of transcription. The novel model enables to simulate age-related changes of trimethylation of lysine 4 at histone H3 and of DNA methylation. These changes entail expression changes of genes that induce age-related phenotypes (ARPs) of cells. We compare age-related changes of regulatory states in quiescent stem cells occupying a niche with those observed in proliferating cells. Moreover, we analyze the impact of the activity of the involved epigenetic modifiers on these changes. We find that epigenetic aging strongly affects stem cell heterogeneity and that homing at stem cell niches retards epigenetic aging. Our model provides a mechanistic explanation how increased stem cell proliferation can lead to progeroid phenotypes. Adapting our model to properties observed for aged hematopoietic stem cell (HSC) clones, we predict that the hematopoietic ARP activates young HSCs and thereby retards aging of the entire HSC population. In addition, our model suggests that the experimentally observed high interindividual variance in HSC numbers originates in a variance of histone methyltransferase activity.


Assuntos
Senescência Celular/genética , Biologia Computacional/métodos , Epigênese Genética , Células-Tronco Hematopoéticas/metabolismo , Modelos Biológicos , Simulação por Computador , Metilação de DNA/genética , Retroalimentação Fisiológica , Células-Tronco Hematopoéticas/citologia , Humanos , Fenótipo , Biologia de Sistemas
15.
Cell Rep ; 5(2): 349-56, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-24209744

RESUMO

The pyloric epithelium continuously self-renews throughout life, driven by limited reservoirs of resident Lgr5+ adult stem cells. Here, we characterize the population dynamics of these stem cells during epithelial homeostasis. Using a clonal fate-mapping strategy, we demonstrate that multiple Lgr5+ cells routinely contribute to epithelial renewal in the pyloric gland and, similar to what was previously observed in the intestine, a balanced homeostasis of the glandular epithelium and stem cell pools is predominantly achieved via neutral competition between symmetrically dividing Lgr5+ stem cells. Additionally, we document a lateral expansion of stem cell clones via gland fission under nondamage conditions. These findings represent a major advance in our basic understanding of tissue homeostasis in the stomach and form the foundation for identifying altered stem cell behavior during gastric disease.


Assuntos
Células-Tronco Adultas/metabolismo , Piloro/citologia , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco Adultas/citologia , Animais , Linhagem da Célula , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Homeostase , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética
16.
Bioessays ; 34(10): 841-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22821708

RESUMO

Epigenetic control of gene expression by chromatin remodeling is critical for adult stem cell function. A decline in stem cell function is observed during aging, which is accompanied by changes in the chromatin structure that are currently unexplained. Here, we hypothesize that these epigenetic changes originate from the limited cellular capability to inherit epigenetic information. We suggest that spontaneous loss of histone modification, due to fluctuations over short time scales, gives rise to long-term changes in DNA methylation and, accordingly, in gene expression. These changes are assumed to impair stem cell function and, thus, to contribute to aging. We discuss cell replication as a major source of fluctuations in histone modification patterns. Gene silencing by our proposed mechanism can be interpreted as a manifestation of the conflict between the stem cell plasticity required for tissue regeneration and the permanent silencing of potentially deleterious genomic sequences.


Assuntos
Células-Tronco Adultas/fisiologia , Senescência Celular/genética , Montagem e Desmontagem da Cromatina , Células-Tronco Adultas/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Metilação de DNA , Epigênese Genética , Histonas/metabolismo , Humanos , Modelos Biológicos , Especificidade de Órgãos , Processamento de Proteína Pós-Traducional
17.
FEBS J ; 279(18): 3475-87, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22632461

RESUMO

In vitro culture of intestinal tissue has been attempted for decades. Only recently did Sato et al. [Sato, T., Vries, R. G., Snippert, H. J., van de Wetering, M., Barker, N., Stange, D. E., van Es, J. H., Abo, A., Kujala, P., Peters, P. J., et al. (2009) Nature 459, 262-265] succeed in establishing long-term intestinal culture, demonstrating that cells expressing the Lgr5 gene can give rise to organoids with crypt-like domains similar to those found in vivo. In these cultures, Paneth cells provide essential signals supporting stem cell function. We have recently developed an individual cell-based computational model of the intestinal tissue [Buske, P., Galle, J., Barker, N., Aust, G., Clevers, H. & Loeffler, M. (2011) PLoS Comput Biol 7, e1001045]. The model is capable of quantitatively reproducing a comprehensive set of experimental data on intestinal cell organization. Here, we present a significant extension of this model that allows simulation of intestinal organoid formation in silico. For this purpose, we introduce a flexible basal membrane that assigns a bending modulus to the organoid surface. This membrane may be re-organized by cells attached to it depending on their differentiation status. Accordingly, the morphology of the epithelium is self-organized. We hypothesize that local tissue curvature is a key regulatory factor in stem cell organization in the intestinal tissue by controlling Paneth cell specification. In simulation studies, our model closely resembles the spatio-temporal organization of intestinal organoids. According to our results, proliferation-induced shape fluctuations are sufficient to induce crypt-like domains, and spontaneous tissue curvature induced by Paneth cells can control cell number ratios. Thus, stem cell expansion in an organoid depends sensitively on its biomechanics. We suggest a number of experiments that will enable new insights into mechano-transduction in the intestine, and suggest model extensions in the field of gland formation.


Assuntos
Intestinos/citologia , Mecanotransdução Celular/fisiologia , Organoides/metabolismo , Nicho de Células-Tronco , Fenômenos Biomecânicos , Contagem de Células , Diferenciação Celular , Proliferação de Células , Simulação por Computador , Células Epiteliais/fisiologia , Modelos Biológicos , Celulas de Paneth/fisiologia , Células-Tronco/citologia
18.
PLoS One ; 6(7): e21960, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21760935

RESUMO

Therapeutic application of mesenchymal stem cells (MSC) requires their extensive in vitro expansion. MSC in culture typically grow to confluence within a few weeks. They show spindle-shaped fibroblastoid morphology and align to each other in characteristic spatial patterns at high cell density. We present an individual cell-based model (IBM) that is able to quantitatively describe the spatio-temporal organization of MSC in culture. Our model substantially improves on previous models by explicitly representing cell podia and their dynamics. It employs podia-generated forces for cell movement and adjusts cell behavior in response to cell density. At the same time, it is simple enough to simulate thousands of cells with reasonable computational effort. Experimental sheep MSC cultures were monitored under standard conditions. Automated image analysis was used to determine the location and orientation of individual cells. Our simulations quantitatively reproduced the observed growth dynamics and cell-cell alignment assuming cell density-dependent proliferation, migration, and morphology. In addition to cell growth on plain substrates our model captured cell alignment on micro-structured surfaces. We propose a specific surface micro-structure that according to our simulations can substantially enlarge cell culture harvest. The 'tool box' of cell migratory behavior newly introduced in this study significantly enhances the bandwidth of IBM. Our approach is capable of accommodating individual cell behavior and collective cell dynamics of a variety of cell types and tissues in computational systems biology.


Assuntos
Células-Tronco Mesenquimais/citologia , Modelos Biológicos , Pseudópodes/metabolismo , Animais , Contagem de Células , Divisão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Células-Tronco Mesenquimais/metabolismo , Ovinos
19.
BMC Syst Biol ; 4: 73, 2010 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-20507570

RESUMO

BACKGROUND: In vitro cultivated stem cell populations are in general heterogeneous with respect to their expression of differentiation markers. In hematopoietic progenitor populations, this heterogeneity has been shown to regenerate within days from isolated subpopulations defined by high or low marker expression. This kind of plasticity has been suggested to be a fundamental feature of mesenchymal stem cells (MSCs) as well. Here, we study MSC plasticity on the level of individual cells applying a multi-scale computer model that is based on the concept of noise-driven stem cell differentiation. RESULTS: By simulation studies, we provide detailed insight into the kinetics of MSC organisation. Monitoring the fates of individual cells in high and low oxygen culture, we calculated the average transition times of individual cells into stem cell and differentiated states. We predict that at low oxygen the heterogeneity of a MSC population with respect to differentiation regenerates from any selected subpopulation in about two days. At high oxygen, regeneration becomes substantially slowed down. Simulation results on the composition of the functional stem cell pool of MSC populations suggest that most of the cells that constitute this pool originate from more differentiated cells. CONCLUSIONS: Individual cell-based models are well-suited to provide quantitative predictions on essential features of the spatio-temporal organisation of MSC in vitro. Our predictions on MSC plasticity and its dependence on the environment motivate a number of in vitro experiments for validation. They may contribute to a better understanding of MSC organisation in vitro, including features of clonal expansion, environmental adaptation and stem cell ageing.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Modelos Biológicos , Simulação por Computador , Técnicas In Vitro
20.
PLoS One ; 3(8): e2922, 2008 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-18698344

RESUMO

BACKGROUND: The balance between maintenance of the stem cell state and terminal differentiation is influenced by the cellular environment. The switching between these states has long been understood as a transition between attractor states of a molecular network. Herein, stochastic fluctuations are either suppressed or can trigger the transition, but they do not actually determine the attractor states. METHODOLOGY/PRINCIPAL FINDINGS: We present a novel mathematical concept in which stem cell and progenitor population dynamics are described as a probabilistic process that arises from cell proliferation and small fluctuations in the state of differentiation. These state fluctuations reflect random transitions between different activation patterns of the underlying regulatory network. Importantly, the associated noise amplitudes are state-dependent and set by the environment. Their variability determines the attractor states, and thus actually governs population dynamics. This model quantitatively reproduces the observed dynamics of differentiation and dedifferentiation in promyelocytic precursor cells. CONCLUSIONS/SIGNIFICANCE: Consequently, state-specific noise modulation by external signals can be instrumental in controlling stem cell and progenitor population dynamics. We propose follow-up experiments for quantifying the imprinting influence of the environment on cellular noise regulation.


Assuntos
Diferenciação Celular/fisiologia , Ruído , Células-Tronco/citologia , Células-Tronco/fisiologia , Aclimatação , Animais , Divisão Celular , Meio Ambiente , Cinética , Modelos Biológicos , Dinâmica Populacional , Regeneração/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...