Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
EMBO J ; 42(21): e113891, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37743763

ABSTRACT

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.


Subject(s)
Cilia , Organelles , Cilia/metabolism , Cell Differentiation
2.
Dev Cell ; 58(21): 2292-2308.e6, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37591246

ABSTRACT

Basic helix-loop-helix genes, particularly proneural genes, are well-described triggers of cell differentiation, yet information on their dynamics is limited, notably in human development. Here, we focus on Neurogenin 3 (NEUROG3), which is crucial for pancreatic endocrine lineage initiation. By monitoring both NEUROG3 gene expression and protein in single cells using a knockin dual reporter in 2D and 3D models of human pancreas development, we show an approximately 2-fold slower expression of human NEUROG3 than that of the mouse. We observe heterogeneous peak levels of NEUROG3 expression and reveal through long-term live imaging that both low and high NEUROG3 peak levels can trigger differentiation into hormone-expressing cells. Based on fluorescence intensity, we statistically integrate single-cell transcriptome with dynamic behaviors of live cells and propose a data-mapping methodology applicable to other contexts. Using this methodology, we identify a role for KLK12 in motility at the onset of NEUROG3 expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Nerve Tissue Proteins , Humans , Animals , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas/metabolism , Cell Differentiation/genetics , Endocrine System/metabolism
3.
Nat Rev Endocrinol ; 19(4): 232-248, 2023 04.
Article in English | MEDLINE | ID: mdl-36670309

ABSTRACT

Our understanding of diabetes mellitus has benefited from a combination of clinical investigations and work in model organisms and cell lines. Organoid models for a wide range of tissues are emerging as an additional tool enabling the study of diabetes mellitus. The applications for organoid models include studying human pancreatic cell development, pancreatic physiology, the response of target organs to pancreatic hormones and how glucose toxicity can affect tissues such as the blood vessels, retina, kidney and nerves. Organoids can be derived from human tissue cells or pluripotent stem cells and enable the production of human cell assemblies mimicking human organs. Many organ mimics relevant to diabetes mellitus are already available, but only a few relevant studies have been performed. We discuss the models that have been developed for the pancreas, liver, kidney, nerves and vasculature, how they complement other models, and their limitations. In addition, as diabetes mellitus is a multi-organ disease, we highlight how a merger between the organoid and bioengineering fields will provide integrative models.


Subject(s)
Diabetes Mellitus , Pluripotent Stem Cells , Humans , Organoids/metabolism , Diabetes Mellitus/metabolism , Pancreas , Liver
4.
Semin Cell Dev Biol ; 141: 23-32, 2023 05 30.
Article in English | MEDLINE | ID: mdl-35466054

ABSTRACT

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.


Subject(s)
Organoids , Synthetic Biology , Cell Differentiation , Signal Transduction
5.
Nat Commun ; 13(1): 6255, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36271049

ABSTRACT

Diabetes is a multifactorial disorder characterized by loss or dysfunction of pancreatic ß-cells. ß-cells are heterogeneous, exhibiting different glucose sensing, insulin secretion and gene expression. They communicate with other endocrine cell types via paracrine signals and between ß-cells via gap junctions. Here, we identify the importance of signaling between ß-cells via the extracellular signal WNT4. We show heterogeneity in Wnt4 expression, most strikingly in the postnatal maturation period, Wnt4-positive cells, being more mature while Wnt4-negative cells are more proliferative. Knock-out in adult ß-cells shows that WNT4 controls the activation of calcium signaling in response to a glucose challenge, as well as metabolic pathways converging to lower ATP/ADP ratios, thereby reducing insulin secretion. These results reveal that paracrine signaling between ß-cells is important in addition to gap junctions in controling insulin secretion. Together with previous reports of WNT4 up-regulation in obesity our observations suggest an adaptive insulin response coordinating ß-cells.


Subject(s)
Calcium Signaling , Insulins , Glucose/metabolism , Adenosine Triphosphate/metabolism , Insulins/metabolism , Adenosine Diphosphate/metabolism
6.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-36314540

ABSTRACT

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.


Subject(s)
Biomedical Research , Pluripotent Stem Cells , Organoids , Organogenesis , Pancreas
7.
Annu Rev Cell Dev Biol ; 38: 447-466, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35767871

ABSTRACT

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.


Subject(s)
Organoids , Stem Cells , Cell Differentiation
8.
Stem Cell Reports ; 17(5): 1215-1228, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35452596

ABSTRACT

With the aim of producing ß cells for replacement therapies to treat diabetes, several protocols have been developed to differentiate human pluripotent stem cells to ß cells via pancreatic progenitors. While in vivo pancreatic progenitors expand throughout development, the in vitro protocols have been designed to make these cells progress as fast as possible to ß cells. Here, we report on a protocol enabling a long-term expansion of human pancreatic progenitors in a defined medium on fibronectin, in the absence of feeder layers. Moreover, through a screening of a polymer library we identify a polymer that can replace fibronectin. Our experiments, comparing expanded progenitors to directly differentiated progenitors, show that the expanded progenitors differentiate more efficiently into glucose-responsive ß cells and produce fewer glucagon-expressing cells. The ability to expand progenitors under defined conditions and cryopreserve them will provide flexibility in research and therapeutic production.


Subject(s)
Insulin-Secreting Cells , Pluripotent Stem Cells , Cell Differentiation , Fibronectins/pharmacology , Humans , Pancreas , Polymers
9.
Cell Stem Cell ; 29(4): 500-502, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35395185

ABSTRACT

Du et al. transplanted ß cells derived from pluripotent stem cells in diabetic monkeys for the first time, as an intermediate stage toward clinical translation. They observed benefits unfolding over months but also observed immune rejection of the grafts by 5-6 months.


Subject(s)
Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Pluripotent Stem Cells , Animals , Haplorhini
10.
Stem Cell Rev Rep ; 18(1): 360-377, 2022 01.
Article in English | MEDLINE | ID: mdl-34586606

ABSTRACT

Mesenchymal cells of diverse origins differ in gene and protein expression besides producing varying effects on their organ-matched epithelial cells' maintenance and differentiation capacity. Co-culture with rodent's tissue-specific pancreatic mesenchyme accelerates proliferation, self-renewal, and differentiation of pancreatic epithelial progenitors. Therefore, in our study, the impact of three-dimensional (3D) co-culture of human fetal pancreatic-derived mesenchymal cells (hFP-MCs) with human embryonic stem cell-derived pancreatic progenitors (hESC-PPs) development towards endocrine and beta cells was assessed. Besides, the ability to maintain scalable cultures combining hFP-MCs and hESC-PPs was investigated. hFP-MCs expressed many markers in common with bone marrow-derived mesenchymal stem cells (BM-MSCs). However, they showed higher expression of DESMIN compared to BM-MSCs. After co-culture of hESC-PPs with hFP-MCs, the pancreatic progenitor (PP) spheroids generated in Matrigel had higher expression of NGN3 and INSULIN than BM-MSCs co-culture group, which shows an inductive impact of pancreatic mesenchyme on hESC-PPs beta-cells maturation. Pancreatic aggregates generated by forced aggregation through scalable AggreWell system showed similar features compared to the spheroids. These aggregates, a combination of hFP-MCs and hESC-PPs, can be applied as an appropriate tool for assessing endocrine-niche interactions and developmental processes by mimicking the pancreatic tissue.


Subject(s)
Human Embryonic Stem Cells , Mesenchymal Stem Cells , Cell Differentiation , Coculture Techniques , Human Embryonic Stem Cells/metabolism , Humans , Pancreas
11.
Curr Opin Genet Dev ; 72: 30-37, 2022 02.
Article in English | MEDLINE | ID: mdl-34794006

ABSTRACT

Epithelial organoids can recapitulate many processes reminiscent of morphogenesis in vivo including lumen and multilayer formation, folding, branching, delamination and elongation. While being noisier in vitro than in vivo, these processes can be monitored live and subjected to interferences, a field that is emerging. We elaborate on the signalling molecules controlling morphogenesis, from the medium and their emergence as signalling centers in the organoids. Further, we discuss how organoid shape is controlled by mechanical cues within the organoid and their interplay with the material properties of the environment.


Subject(s)
Organoids , Morphogenesis/genetics
12.
Nat Commun ; 12(1): 3144, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035279

ABSTRACT

Human organogenesis remains relatively unexplored for ethical and practical reasons. Here, we report the establishment of a single-cell transcriptome atlas of the human fetal pancreas between 7 and 10 post-conceptional weeks of development. To interrogate cell-cell interactions, we describe InterCom, an R-Package we developed for identifying receptor-ligand pairs and their downstream effects. We further report the establishment of a human pancreas culture system starting from fetal tissue or human pluripotent stem cells, enabling the long-term maintenance of pancreas progenitors in a minimal, defined medium in three-dimensions. Benchmarking the cells produced in 2-dimensions and those expanded in 3-dimensions to fetal tissue identifies that progenitors expanded in 3-dimensions are transcriptionally closer to the fetal pancreas. We further demonstrate the potential of this system as a screening platform and identify the importance of the EGF and FGF pathways controlling human pancreas progenitor expansion.


Subject(s)
Cell Culture Techniques/methods , Organogenesis , Pancreas/embryology , Pluripotent Stem Cells/physiology , Tissue Culture Techniques/methods , Aborted Fetus , Animals , Cell Communication , Cell Differentiation , Cell Line , Datasets as Topic , Embryo, Mammalian , Epidermal Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Pancreas/cytology , RNA-Seq , Signal Transduction/physiology , Single-Cell Analysis , Spheroids, Cellular , Transcriptome
13.
Curr Top Dev Biol ; 143: 75-110, 2021.
Article in English | MEDLINE | ID: mdl-33820626

ABSTRACT

The pancreas of adult mammals displays a branched structure which transports digestive enzymes produced in the distal acini through a tree-like network of ducts into the duodenum. In contrast to several other branched organs, its branching patterns are not stereotypic. Moreover, the branches do not grow from dichotomic splitting of an initial stem but rather from the formation of microlumen in a mass of cells. These lumen progressively assemble into a hyperconnected network that refines into a tree by the time of birth. We review the cell remodeling events and the molecular mechanisms governing pancreas branching, as well as the role of the surrounding tissues in this process. Furthermore, we draw parallels with other branched organs such as the salivary and mammary gland.


Subject(s)
Organogenesis , Pancreas , Animals , Mammals , Morphogenesis
14.
J Mol Med (Berl) ; 99(4): 449-462, 2021 04.
Article in English | MEDLINE | ID: mdl-33221939

ABSTRACT

Organoids constitute biological systems which are used to model organ development, homeostasis, regeneration, and disease in vitro and hold promise for use in therapy. Reflecting in vivo development, organoids form from tissue cells or pluripotent stem cells. Cues provided from the media and individual cells promote self-organization of these uniform starting cells into a structure, with emergent differentiated cells, morphology, and often functionality that resemble the tissue of origin. Therefore, organoids provide a complement to two-dimensional in vitro culture and in vivo animal models of development, providing the experimental control and flexibility of in vitro methods with the three-dimensional context of in vivo models, with fewer ethical restraints than human or animal work. However, using organoids, we are only just beginning to understand on the cellular level how the external conditions and signaling between individual cells promote the emergence of cells and structures. In this review, we focus specifically on organoids derived from endodermal tissues: the starting conditions of the cells, signaling mechanisms, and external media that allow the emergence of higher order self-organization.


Subject(s)
Endoderm/cytology , Organoids/cytology , Adult Stem Cells/cytology , Animals , Cell Communication , Cell Culture Techniques/methods , Cell Differentiation , Feedback, Physiological , Humans , Induced Pluripotent Stem Cells/cytology , Intestines/cytology , Mice , Morphogenesis , Organ Specificity , Organogenesis , Organoids/ultrastructure , Signal Transduction
15.
Curr Opin Genet Dev ; 64: 84-93, 2020 10.
Article in English | MEDLINE | ID: mdl-32721583

ABSTRACT

The most common pancreas-related disorders are diabetes, pancreatitis and different types of pancreatic cancers. Diabetes is a chronic condition which results from insufficient functional ß-cell mass, either as a result of an autoimmune destruction of insulin producing ß-cells, or as their death or de-differentiation following years of hyperactivity to compensate for insulin resistance. Chronic pancreatitis leads to cell death and can develop into diabetes or pancreatic cancer. To stimulate regeneration in such pathologies, it is of high importance to evaluate the endogenous regeneration capacity of the pancreas, to understand the conditions needed to trigger it, and to investigate the cellular and molecular regenerative responses. This short review focuses on observations made in the last 2 years on the mechanisms enhancing pancreatic cell proliferation, notably new combinations of pharmacological agents, as well as those triggering cellular conversion.


Subject(s)
Cell Differentiation , Cell Proliferation , Insulin-Secreting Cells/cytology , Pancreatic Diseases/therapy , Regeneration , Animals , Humans , Insulin-Secreting Cells/physiology
16.
Dev Cell ; 54(2): 140-141, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32693054

ABSTRACT

A major trigger of adult ß-cell insulin secretion is glucose. In a recent issue of Cell Metabolism, Helman and colleagues show that in fetuses insulin secretion depends on the activation of mTOR by amino acids and that reducing amino acids promotes maturation of ß-cells derived from pluripotent stem cells.


Subject(s)
Glucose , Insulin-Secreting Cells , Adult , Glucose/metabolism , Humans , Infant, Newborn , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Nutrients
17.
Cell Rep ; 31(8): 107677, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32460029

ABSTRACT

Cell polarity is essential for the architecture and function of numerous epithelial tissues. Here, we show that apical restriction of planar cell polarity (PCP) components is necessary for the maintenance of epithelial integrity. Using the mammalian pancreas as a model, we find that components of the core PCP pathway, such as the transmembrane protein Van Gogh-like (VANGL), become apically restricted over a period of several days. Expansion of VANGL localization to the basolateral membranes of progenitors leads to their death and disruption of the epithelial integrity. VANGL basolateral expansion does not affect apico-basal polarity but acts in the cells where Vangl is mislocalized by reducing Dishevelled and its downstream target ROCK. This reduction in ROCK activity culminates in progenitor cell egression, death, and eventually pancreatic hypoplasia. Thus, precise spatiotemporal modulation of VANGL-dependent PCP signaling is crucial for proper pancreatic morphogenesis.


Subject(s)
Membrane Proteins/metabolism , Pancreatic Ducts/ultrastructure , Animals , Cell Membrane , Cell Polarity , Epithelium , Mice
18.
Diabetes ; 68(4): 761-773, 2019 04.
Article in English | MEDLINE | ID: mdl-30655386

ABSTRACT

Bromodomain and extraterminal (BET) proteins are epigenetic readers that interact with acetylated lysines of histone tails. Recent studies have demonstrated their role in cancer progression because they recruit key components of the transcriptional machinery to modulate gene expression. However, their role during embryonic development of the pancreas has never been studied. Using mouse embryonic pancreatic explants and human induced pluripotent stem cells (hiPSCs), we show that BET protein inhibition with I-BET151 or JQ1 enhances the number of neurogenin3 (NEUROG3) endocrine progenitors. In mouse explants, BET protein inhibition further led to increased expression of ß-cell markers but in the meantime, strongly downregulated Ins1 expression. Similarly, although acinar markers, such as Cpa1 and CelA, were upregulated, Amy expression was repressed. In hiPSCs, BET inhibitors strongly repressed C-peptide and glucagon during endocrine differentiation. Explants and hiPSCs were then pulsed with BET inhibitors to increase NEUROG3 expression and further chased without inhibitors. Endocrine development was enhanced in explants with higher expression of insulin and maturation markers, such as UCN3 and MAFA. In hiPSCs, the outcome was different because C-peptide expression remained lower than in controls, but ghrelin expression was increased. Altogether, by using two independent models of pancreatic development, we show that BET proteins regulate multiple aspects of pancreatic development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Nerve Tissue Proteins/metabolism , Proteins/antagonists & inhibitors , Animals , Azepines/pharmacology , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Induced Pluripotent Stem Cells , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Signal Transduction/drug effects , Signal Transduction/physiology , Triazoles/pharmacology
19.
Development ; 145(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-30042179

ABSTRACT

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.


Subject(s)
Fetus/embryology , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental/physiology , Pancreas/embryology , Transcription, Genetic/physiology , Fetus/cytology , Humans , Pancreas/cytology , Pluripotent Stem Cells/metabolism
20.
PLoS Biol ; 16(7): e2002842, 2018 07.
Article in English | MEDLINE | ID: mdl-30048442

ABSTRACT

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.


Subject(s)
Pancreatic Ducts/embryology , Animals , Body Fluids/metabolism , Colforsin/pharmacology , Computer Simulation , Embryonic Development , Female , Image Processing, Computer-Assisted , Mice, Inbred ICR , Pancreatic Ducts/anatomy & histology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...