Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 38: 447-466, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35767871

RESUMEN

Organoids are miniaturized and simplified versions of an organ produced in vitro from stem or progenitor cells. They are used as a model system consisting of multiple cell types forming an architecture relevant to the organ and carrying out the function of the organ. They are a useful tool to study development, homeostasis, regeneration, and disease. The imaging of organoids has become a pivotal method to visualize and understand their self-organization, symmetry breaking, growth, differentiation, and function. In this review, we discuss imaging methods, how to analyze these images, and challenges in organoid research.


Asunto(s)
Organoides , Células Madre , Diferenciación Celular
2.
EMBO J ; 42(21): e113891, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37743763

RESUMEN

Primary cilia project from the surface of most vertebrate cells and are key in sensing extracellular signals and locally transducing this information into a cellular response. Recent findings show that primary cilia are not merely static organelles with a distinct lipid and protein composition. Instead, the function of primary cilia relies on the dynamic composition of molecules within the cilium, the context-dependent sensing and processing of extracellular stimuli, and cycles of assembly and disassembly in a cell- and tissue-specific manner. Thereby, primary cilia dynamically integrate different cellular inputs and control cell fate and function during tissue development. Here, we review the recently emerging concept of primary cilia dynamics in tissue development, organization, remodeling, and function.


Asunto(s)
Cilios , Orgánulos , Cilios/metabolismo , Diferenciación Celular
3.
Semin Cell Dev Biol ; 141: 23-32, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-35466054

RESUMEN

The functional relevance of an organoid is dependent on the differentiation, morphology, cell arrangement and biophysical properties, which collectively define the state of an organoid. For an organoid culture, an individual organoid or the cells that compose it, these state variables can be characterised, most easily by transcriptomics and by high-content image analysis. Their states can be compared to their in vivo counterparts. Current evidence suggests that organoids explore a wider state space than organs in vivo due to the lack of niche signalling and the variability of boundary conditions in vitro. Using data-driven state inference and in silico modelling, phase diagrams can be constructed to systematically sort organoids along biochemical or biophysical axes. These phase diagrams allow us to identify control strategies to modulate organoid state. To do so, the biochemical and biophysical environment, as well as the cells that seed organoids, can be manipulated.


Asunto(s)
Organoides , Biología Sintética , Diferenciación Celular , Transducción de Señal
4.
Development ; 149(20)2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36314540

RESUMEN

Organoids have become one of the fastest progressing and applied models in biological and medical research, and various organoids have now been developed for most of the organs of the body. Here, we review the methods developed to generate pancreas organoids in vitro from embryonic, fetal and adult cells, as well as pluripotent stem cells. We discuss how these systems have been used to learn new aspects of pancreas development, regeneration and disease, as well as their limitations and potential for future discoveries.


Asunto(s)
Investigación Biomédica , Células Madre Pluripotentes , Organoides , Organogénesis , Páncreas
5.
Development ; 145(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30042179

RESUMEN

To decipher the populations of cells present in the human fetal pancreas and their lineage relationships, we developed strategies to isolate pancreatic progenitors, endocrine progenitors and endocrine cells. Transcriptome analysis of the individual populations revealed a large degree of conservation among vertebrates in the drivers of gene expression changes that occur at different steps of differentiation, although notably, sometimes, different members of the same gene family are expressed. The transcriptome analysis establishes a resource to identify novel genes and pathways involved in human pancreas development. Single-cell profiling further captured intermediate stages of differentiation and enabled us to decipher the sequence of transcriptional events occurring during human endocrine differentiation. Furthermore, we evaluate how well individual pancreatic cells derived in vitro from human pluripotent stem cells mirror the natural process occurring in human fetuses. This comparison uncovers a few differences at the progenitor steps, a convergence at the steps of endocrine induction, and the current inability to fully resolve endocrine cell subtypes in vitro.


Asunto(s)
Feto/embriología , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/fisiología , Páncreas/embriología , Transcripción Genética/fisiología , Feto/citología , Humanos , Páncreas/citología , Células Madre Pluripotentes/metabolismo
6.
PLoS Biol ; 16(7): e2002842, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30048442

RESUMEN

The mammalian pancreas is a branched organ that does not exhibit stereotypic branching patterns, similarly to most other glands. Inside branches, it contains a network of ducts that undergo a transition from unconnected microlumen to a mesh of interconnected ducts and finally to a treelike structure. This ductal remodeling is poorly understood, both on a microscopic and macroscopic level. In this article, we quantify the network properties at different developmental stages. We find that the pancreatic network exhibits stereotypic traits at each stage and that the network properties change with time toward the most economical and optimized delivery of exocrine products into the duodenum. Using in silico modeling, we show how steps of pancreatic network development can be deconstructed into two simple rules likely to be conserved for many other glands. The early stage of the network is explained by noisy, redundant duct connection as new microlumens form. The later transition is attributed to pruning of the network based on the flux of fluid running through the pancreatic network into the duodenum.


Asunto(s)
Conductos Pancreáticos/embriología , Animales , Líquidos Corporales/metabolismo , Colforsina/farmacología , Simulación por Computador , Desarrollo Embrionario , Femenino , Procesamiento de Imagen Asistido por Computador , Ratones Endogámicos ICR , Conductos Pancreáticos/anatomía & histología , Factores de Tiempo
7.
Development ; 144(6): 946-951, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28292839

RESUMEN

Organoids representing a diversity of tissues have recently been created, bridging the gap between cell culture and experiments performed in vivo Being small and amenable to continuous monitoring, they offer the opportunity to scrutinize the dynamics of organ development, including the exciting prospect of observing aspects of human embryo development live. From a physicist's perspective, their ability to self-organize - to differentiate and organize cells in space - calls for the identification of the simple rules that underlie this capacity. Organoids provide tractable conditions to investigate the effects of the growth environment, including its molecular composition and mechanical properties, along with the initial conditions such as cell number and type(s). From a theoretical standpoint, different types of in silico modeling can complement the measurements performed in organoids to understand the role of chemical diffusion, contact signaling, differential cell adhesion and mechanical controls. Here, we discuss what it means to take a biophysical approach to understanding organogenesis in vitro and how we might expect such approaches to develop in the future.


Asunto(s)
Fenómenos Biofísicos , Organogénesis , Organoides/fisiología , Animales , Biología Computacional , Humanos , Modelos Biológicos
8.
Semin Cell Dev Biol ; 66: 51-68, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28089869

RESUMEN

The pancreas is an essential endoderm-derived organ that ensures nutrient metabolism via its endocrine and exocrine functions. Here we review the essential processes governing the embryonic and early postnatal development of the pancreas discussing both the mechanisms and molecules controlling progenitor specification, expansion and differentiation. We elaborate on how these processes are orchestrated in space and coordinated with morphogenesis. We draw mainly from experiments conducted in the mouse model but also from investigations in other model organisms, complementing a recent comprehensive review of human pancreas development (Jennings et al., 2015) [1]. The understanding of pancreas development in model organisms provides a framework to interpret how human mutations lead to neonatal diabetes and may contribute to other forms of diabetes and to guide the production of desired pancreatic cell types from pluripotent stem cells for therapeutic purposes.


Asunto(s)
Morfogénesis/genética , Organogénesis/genética , Páncreas/crecimiento & desarrollo , Animales , Humanos
9.
Gastroenterology ; 155(4): 1164-1176.e2, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29935151

RESUMEN

BACKGROUND & AIMS: Glucagon-like peptide 1 (GLP1) is produced by L cells in the intestine, and agonists of the GLP1 receptor are effective in the treatment of diabetes. Levels of GLP1 increase with numbers of L cells. Therefore, agents that increase numbers of L cell might be developed for treatment of diabetes. Ras homologue family member A (RhoA) signaling through Rho-associated coiled-coil-containing protein kinases 1 and 2 (ROCK1 and ROCK2) controls cell differentiation, but it is not clear whether this pathway regulates enteroendocrine differentiation in the intestinal epithelium. We investigated the effects of Y-27632, an inhibitor of ROCK1 and ROCK2, on L-cell differentiation. METHODS: We collected intestinal tissues from GLU-Venus, GPR41-RFP, and Neurog3-RFP mice, in which the endocrine lineage is fluorescently labeled, for in vitro culture and histologic analysis. Small intestine organoids derived from these mice were cultured with Y-27632 and we measured percentages of L cells, expression of intestinal cell-specific markers, and secretion of GLP1 in medium. Mice were fed a normal chow or a high-fat diet and given Y-27632 or saline (control) and blood samples were collected for measurement of GLP1, insulin, and glucose. RESULTS: Incubation of intestinal organoids with Y-27632 increased numbers of L cells and secretion of GLP1. These increases were associated with upregulated expression of genes encoding intestinal hormones, neurogenin 3, neurogenic differentiation factor 1, forkhead box A1 and A2, and additional markers of secretory cells. Mice fed the normal chow diet and given Y-27632 had increased numbers of L cells in intestinal tissues, increased plasma levels of GLP1 and insulin, and lower blood levels of glucose compared with mice fed the normal chow diet and given saline. In mice with insulin resistance induced by the high-fat diet, administration of Y-27632 increased secretion of GLP1 and glucose tolerance compared with administration of saline. CONCLUSIONS: In mouse intestinal organoids, an inhibitor of RhoA signaling increased the differentiation of the secretory lineage and the development of enteroendocrine cells. Inhibitors of RhoA signaling or other strategies to increase numbers of L cells might be developed for treatment of patients with type 2 diabetes or for increasing glucose tolerance.


Asunto(s)
Amidas/farmacología , Glucemia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Enteroendocrinas/efectos de los fármacos , Intolerancia a la Glucosa/tratamiento farmacológico , Hipoglucemiantes/farmacología , Íleon/efectos de los fármacos , Resistencia a la Insulina , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Células Madre/efectos de los fármacos , Proteínas de Unión al GTP rho/metabolismo , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/sangre , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/fisiopatología , Íleon/metabolismo , Insulina/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Organoides/efectos de los fármacos , Organoides/metabolismo , Fenotipo , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
10.
Development ; 142(11): 1912-7, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26015535

RESUMEN

The endoderm gives rise to diverse tissues and organs that are essential for the homeostasis and metabolism of the organism: the thymus, thyroid, lungs, liver and pancreas, and the functionally diverse domains of the digestive tract. Classically, the endoderm, the 'innermost germ layer', was in the shadow of the ectoderm and mesoderm. However, at a recent Keystone meeting it took center stage, revealing astonishing progress in dissecting the mechanisms underlying the development and malfunction of the endodermal organs. In vitro cultures of stem and progenitor cells have become widespread, with remarkable success in differentiating three-dimensional organoids, which - in a new turn for the field - can be used as disease models.


Asunto(s)
Linaje de la Célula , Enfermedad , Desarrollo Embrionario , Endodermo/citología , Animales , Tipificación del Cuerpo , Diferenciación Celular , Modelos Animales de Enfermedad , Humanos , Organoides/embriología
11.
Development ; 142(5): 858-70, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25715394

RESUMEN

In human, mutations in bicaudal C1 (BICC1), an RNA binding protein, have been identified in patients with kidney dysplasia. Deletion of Bicc1 in mouse leads to left-right asymmetry randomization and renal cysts. Here, we show that BICC1 is also expressed in both the pancreatic progenitor cells that line the ducts during development, and in the ducts after birth, but not in differentiated endocrine or acinar cells. Genetic inactivation of Bicc1 leads to ductal cell over-proliferation and cyst formation. Transcriptome comparison between WT and Bicc1 KO pancreata, before the phenotype onset, reveals that PKD2 functions downstream of BICC1 in preventing cyst formation in the pancreas. Moreover, the analysis highlights immune cell infiltration and stromal reaction developing early in the pancreas of Bicc1 knockout mice. In addition to these functions in duct morphogenesis, BICC1 regulates NEUROG3(+) endocrine progenitor production. Its deletion leads to a late but sustained endocrine progenitor decrease, resulting in a 50% reduction of endocrine cells. We show that BICC1 functions downstream of ONECUT1 in the pathway controlling both NEUROG3(+) endocrine cell production and ductal morphogenesis, and suggest a new candidate gene for syndromes associating kidney dysplasia with pancreatic disorders, including diabetes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Técnica del Anticuerpo Fluorescente , Genotipo , Factor Nuclear 6 del Hepatocito/genética , Etiquetado Corte-Fin in Situ , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Células Madre/citología , Células Madre/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
12.
PLoS Biol ; 13(3): e1002111, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25786211

RESUMEN

Organogenesis relies on the spatiotemporal balancing of differentiation and proliferation driven by an expanding pool of progenitor cells. In the mouse pancreas, lineage tracing at the population level has shown that the expanding pancreas progenitors can initially give rise to all endocrine, ductal, and acinar cells but become bipotent by embryonic day 13.5, giving rise to endocrine cells and ductal cells. However, the dynamics of individual progenitors balancing self-renewal and lineage-specific differentiation has never been described. Using three-dimensional live imaging and in vivo clonal analysis, we reveal the contribution of individual cells to the global behaviour and demonstrate three modes of progenitor divisions: symmetric renewing, symmetric endocrinogenic, and asymmetric generating a progenitor and an endocrine progenitor. Quantitative analysis shows that the endocrine differentiation process is consistent with a simple model of cell cycle-dependent stochastic priming of progenitors to endocrine fate. The findings provide insights to define control parameters to optimize the generation of ß-cells in vitro.


Asunto(s)
Células Acinares/citología , Ciclo Celular/genética , Linaje de la Célula/genética , Células Secretoras de Insulina/citología , Células Madre/citología , Células Acinares/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Proliferación Celular , Rastreo Celular , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Procesamiento de Imagen Asistido por Computador , Células Secretoras de Insulina/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Organogénesis/genética , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Células Madre/metabolismo , Técnicas de Cultivo de Tejidos , Transactivadores/genética , Transactivadores/metabolismo , Proteína Fluorescente Roja
13.
EMBO J ; 32(10): 1393-408, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23584530

RESUMEN

Stem cell differentiation depends on transcriptional activation driven by lineage-specific regulators as well as changes in chromatin organization. However, the coordination of these events is poorly understood. Here, we show that T-box proteins team up with chromatin modifying enzymes to drive the expression of the key lineage regulator, Eomes during endodermal differentiation of embryonic stem (ES) cells. The Eomes locus is maintained in a transcriptionally poised configuration in ES cells. During early differentiation steps, the ES cell factor Tbx3 associates with the histone demethylase Jmjd3 at the enhancer element of the Eomes locus to allow enhancer-promoter interactions. This spatial reorganization of the chromatin primes the cells to respond to Activin signalling, which promotes the binding of Jmjd3 and Eomes to its own bivalent promoter region to further stimulate Eomes expression in a positive feedback loop. In addition, Eomes activates a transcriptional network of core regulators of endodermal differentiation. Our results demonstrate that Jmjd3 sequentially associates with two T-box factors, Tbx3 and Eomes to drive stem cell differentiation towards the definitive endoderm lineage.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Endodermo/citología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas de Dominio T Box/metabolismo , Activinas/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Células Madre Embrionarias/citología , Endodermo/embriología , Endodermo/metabolismo , Elementos de Facilitación Genéticos , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Serina/metabolismo , Proteína Smad2/metabolismo , Proteínas de Dominio T Box/genética
14.
Development ; 141(22): 4385-94, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25371369

RESUMEN

The timing and gene regulatory logic of organ-fate commitment from within the posterior foregut of the mammalian endoderm is largely unexplored. Transient misexpression of a presumed pancreatic-commitment transcription factor, Ptf1a, in embryonic mouse endoderm (Ptf1a(EDD)) dramatically expanded the pancreatic gene regulatory network within the foregut. Ptf1a(EDD) temporarily suppressed Sox2 broadly over the anterior endoderm. Pancreas-proximal organ territories underwent full tissue conversion. Early-stage Ptf1a(EDD) rapidly expanded the endogenous endodermal Pdx1-positive domain and recruited other pancreas-fate-instructive genes, thereby spatially enlarging the potential for pancreatic multipotency. Early Ptf1a(EDD) converted essentially the entire glandular stomach, rostral duodenum and extrahepatic biliary system to pancreas, with formation of many endocrine cell clusters of the type found in normal islets of Langerhans. Sliding the Ptf1a(EDD) expression window through embryogenesis revealed differential temporal competencies for stomach-pancreas respecification. The response to later-stage Ptf1a(EDD) changed radically towards unipotent, acinar-restricted conversion. We provide strong evidence, beyond previous Ptf1a inactivation or misexpression experiments in frog embryos, for spatiotemporally context-dependent activity of Ptf1a as a potent gain-of-function trigger of pro-pancreatic commitment.


Asunto(s)
Endodermo/embriología , Tracto Gastrointestinal/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Organogénesis/fisiología , Páncreas/embriología , Factores de Transcripción/metabolismo , Animales , Endodermo/metabolismo , Técnica del Anticuerpo Fluorescente , Tracto Gastrointestinal/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Técnicas Histológicas , Ratones , Microscopía Confocal , Organogénesis/genética , Factores de Transcripción SOXB1/metabolismo
15.
Diabetologia ; 59(3): 550-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26613896

RESUMEN

AIMS/HYPOTHESIS: It is generally accepted that structural and functional quantitative imaging of individual islets would be beneficial to elucidate the pathogenesis of type 1 diabetes. We here introduce functional optical coherence imaging (FOCI) for fast, label-free monitoring of beta cell destruction and associated alterations of islet vascularisation. METHODS: NOD mouse and human islets transplanted into the anterior chamber of the eye (ACE) were imaged with FOCI, in which the optical contrast of FOCI is based on intrinsic variations of the index of refraction resulting in a faster tomographic acquisition. In addition, the phase sensitivity allows simultaneous label-free acquisition of vascularisation. RESULTS: We demonstrate that FOCI allows longitudinal quantification of progressive autoimmune insulitis, including the three-dimensional quantification of beta cell volume, inflammation and vascularisation. The substantially increased backscattering of islets is dominated by the insulin-zinc nanocrystals in the beta cell granules. This translates into a high specificity for the functional beta cell volume of islets. Applying FOCI to a spontaneous mouse model of type 1 diabetes, we quantify the modifications of the pancreatic microvasculature accompanying the progression of diabetes and reveal a strong correlation between increasing insulitis and density of the vascular network of the islet. CONCLUSIONS/INTERPRETATION: FOCI provides a novel imaging technique for investigating functional and structural diabetes-induced alterations of the islets. The label-free detection of beta cell volume and infiltration together with vascularisation offers a unique extension to study ACE-transplanted human islets. These results are contributing to a deeper understanding of human islet transplant rejection and label-free in vivo monitoring of drug efficacy.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Animales , Modelos Animales de Enfermedad , Genotipo , Humanos , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados
16.
Dev Biol ; 405(2): 316-27, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26156633

RESUMEN

To contribute to devise successful beta-cell differentiation strategies for the cure of Type 1 diabetes we sought to uncover barriers that restrict endocrine fate acquisition by studying the role of the transcriptional repressor REST in the developing pancreas. Rest expression is prevented in neurons and in endocrine cells, which is necessary for their normal function. During development, REST represses a subset of genes in the neuronal differentiation program and Rest is down-regulated as neurons differentiate. Here, we investigate the role of REST in the differentiation of pancreatic endocrine cells, which are molecularly close to neurons. We show that Rest is widely expressed in pancreas progenitors and that it is down-regulated in differentiated endocrine cells. Sustained expression of REST in Pdx1(+) progenitors impairs the differentiation of endocrine-committed Neurog3(+) progenitors, decreases beta and alpha cell mass by E18.5, and triggers diabetes in adulthood. Conditional inactivation of Rest in Pdx1(+) progenitors is not sufficient to trigger endocrine differentiation but up-regulates a subset of differentiation genes. Our results show that the transcriptional repressor REST is active in pancreas progenitors where it gates the activation of part of the beta cell differentiation program.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Páncreas/metabolismo , Proteínas Represoras/fisiología , Animales , Glucemia/metabolismo , Regulación hacia Abajo , Células Endocrinas/citología , Células Endocrinas/metabolismo , Sistema Endocrino/metabolismo , Eliminación de Gen , Proteínas de Homeodominio/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Páncreas/embriología , Células Madre/citología , Transactivadores/metabolismo , Transgenes
17.
Development ; 140(21): 4452-62, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24130330

RESUMEN

In the context of a cellular therapy for diabetes, methods for pancreatic progenitor expansion and subsequent differentiation into insulin-producing beta cells would be extremely valuable. Here we establish three-dimensional culture conditions in Matrigel that enable the efficient expansion of dissociated mouse embryonic pancreatic progenitors. By manipulating the medium composition we generate either hollow spheres, which are mainly composed of pancreatic progenitors, or complex organoids that spontaneously undergo pancreatic morphogenesis and differentiation. The in vitro maintenance and expansion of pancreatic progenitors require active Notch and FGF signaling, thus recapitulating in vivo niche signaling interactions. Our experiments reveal new aspects of pancreas development, such as a community effect by which small groups of cells better maintain progenitor properties and expand more efficiently than isolated cells, as well as the requirement for three-dimensionality. Finally, growth conditions in chemically defined biomaterials pave the way for testing the biophysical and biochemical properties of the niche that sustains pancreatic progenitors.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Morfogénesis/fisiología , Páncreas/citología , Páncreas/crecimiento & desarrollo , Células Madre/citología , Animales , Colágeno , Combinación de Medicamentos , Hidrogel de Polietilenoglicol-Dimetacrilato , Inmunohistoquímica , Laminina , Ratones , Microscopía Fluorescente , Proteoglicanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Estadísticas no Paramétricas , Imagen de Lapso de Tiempo
18.
Phys Biol ; 13(1): 016007, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26906913

RESUMEN

Many organs such as the vasculature, kidney, lungs, pancreas and several other glands form ramified networks of tubes that either maximize exchange surfaces between two compartments or minimize the volume of an organ dedicated to the production and local delivery of a cell-derived product. The structure of these tubular networks can be stereotyped, as in the lungs, or stochastic with large variations between individuals, as in the pancreas. The principles driving stereotyped branching have attracted much attention and several models have been proposed and refined. Here we focus on the pancreas, as a model of non-stereotyped branching. In many ramified tubular organs, an important role of the mesenchyme as a source of branching signals has been proposed, including in the pancreas. However, our previous work has shown that in the absence of mesenchyme, epithelial cells seeded in vitro in Matrigel form heavily branched organoids. Here we experimentally show that pancreatic organoids grow primarily at the tips. Furthermore, in contrast to classical 'depletion of activator' mechanisms, organoids growing in close vicinity seem not to affect each other's growth before they get in contact. We recapitulate these observations in an in silico model of branching assuming a 'local inhibitor' is secreted by the epithelium. Remarkably this simple mechanism is sufficient to generate branched organoids similar to those observed in vitro, including their transition from filled spheres to a tree like structure. Quantifying the similarity between in silico and in vitro development through a normalized surface to volume ratio, our in silico model predicts that inhibition is likely to be cooperative and that the diffusing inhibitor decays within a length scale of 10-20 µm.


Asunto(s)
Células Epiteliales/fisiología , Inhibidores de Crecimiento/metabolismo , Páncreas/crecimiento & desarrollo , Transducción de Señal , Animales , Epitelio/fisiología , Humanos , Modelos Químicos
19.
Stem Cells ; 33(1): 8-14, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25185771

RESUMEN

Three-dimensional models of organ biogenesis have recently flourished. They promote a balance between stem/progenitor cell expansion and differentiation without the constraints of flat tissue culture vessels, allowing for autonomous self-organization of cells. Such models allow the formation of miniature organs in a dish and are emerging for the pancreas, starting from embryonic progenitors and adult cells. This review focuses on the currently available systems and how these allow new types of questions to be addressed. We discuss the expected advancements including their potential to study human pancreas development and function as well as to develop diabetes models and therapeutic cells.


Asunto(s)
Organogénesis/fisiología , Páncreas/crecimiento & desarrollo , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Diabetes Mellitus/patología , Humanos , Técnicas In Vitro , Páncreas/citología , Células Madre/fisiología
20.
Diabetologia ; 58(5): 859-61, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25733380

RESUMEN

The transcription factor SOX9 is regarded as a crucial player in pancreas development, both maintaining progenitors and later being required for beta cell differentiation. However, very little is known about the possible involvement of other SOX family members in such processes. In this issue, the work of Xu et al (DOI: 10.1007/s00125-015-3507-x ) shines a spotlight on SOX4, revealing this factor to be a major player in the beta cell program. Using conditional inactivation in mice, they show that SOX4 shares some functions in progenitors with SOX9, but also plays a distinct role at a later stage of development, during the maturation of endocrine cells. This information is timely as this final maturation process is currently the most challenging to reproduce in vitro when coaxing pluripotent stem cells to convert into beta cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Islotes Pancreáticos/embriología , Proteínas del Tejido Nervioso/metabolismo , Organogénesis/genética , Factores de Transcripción SOXC/metabolismo , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA