Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
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
2.
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
3.
PLoS Comput Biol ; 7(1): e1001045, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-21253562

RESUMO

We introduce a novel dynamic model of stem cell and tissue organisation in murine intestinal crypts. Integrating the molecular, cellular and tissue level of description, this model links a broad spectrum of experimental observations encompassing spatially confined cell proliferation, directed cell migration, multiple cell lineage decisions and clonal competition.Using computational simulations we demonstrate that the model is capable of quantitatively describing and predicting the dynamic behaviour of the intestinal tissue during steady state as well as after cell damage and following selective gain or loss of gene function manipulations affecting Wnt- and Notch-signalling. Our simulation results suggest that reversibility and flexibility of cellular decisions are key elements of robust tissue organisation of the intestine. We predict that the tissue should be able to fully recover after complete elimination of cellular subpopulations including subpopulations deemed to be functional stem cells. This challenges current views of tissue stem cell organisation.


Assuntos
Intestinos/citologia , Modelos Biológicos , Células-Tronco/citologia , Apoptose , Proliferação de Células , Mucosa Intestinal/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo
4.
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
5.
Mol Biol Cell ; 24(14): 2256-68, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23676664

RESUMO

Postnatal enlargement of the mammalian intestine comprises cylindrical and luminal growth, associated with crypt fission and crypt/villus hyperplasia, respectively, which subsequently predominate before and after weaning. The bipartite adhesion G protein-coupled receptor CD97 shows an expression gradient along the crypt-villus axis in the normal human intestine. We here report that transgenic mice overexpressing CD97 in intestinal epithelial cells develop an upper megaintestine. Intestinal enlargement involves an increase in length and diameter but does not affect microscopic morphology, as typical for cylindrical growth. The megaintestine is acquired after birth and before weaning, independent of the genotype of the mother, excluding altered availability of milk constituents as driving factor. CD97 overexpression does not regulate intestinal growth factors, stem cell markers, and Wnt signaling, which contribute to epithelial differentiation and renewal, nor does it affect suckling-to-weaning transition. Consistent with augmented cylindrical growth, suckling but not adult transgenic mice show enlarged crypts and thus more crypt fissions caused by a transient increase of the crypt transit-amplifying zone. Intestinal enlargement by CD97 requires its seven-span transmembrane/cytoplasmic C-terminal fragment but not the N-terminal fragment binding partner CD55. In summary, ectopic expression of CD97 in intestinal epithelial cells provides a unique model for intestinal cylindrical growth occurring in breast-fed infants.


Assuntos
Células Epiteliais/citologia , Expressão Gênica , Mucosa Intestinal/citologia , Intestino Delgado/citologia , Glicoproteínas de Membrana/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Animais Lactentes/fisiologia , Antígenos CD55/genética , Antígenos CD55/metabolismo , Proliferação de Células , Células Epiteliais/metabolismo , Feminino , Humanos , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores Acoplados a Proteínas G , Desmame
6.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA