Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Proc Natl Acad Sci U S A ; 119(15): e2112892119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35412853

ABSTRACT

During early Drosophila embryogenesis, a network of gene regulatory interactions orchestrates terminal patterning, playing a critical role in the subsequent formation of the gut. We utilized CRISPR gene editing at endogenous loci to create live reporters of transcription and light-sheet microscopy to monitor the individual components of the posterior gut patterning network across 90 min prior to gastrulation. We developed a computational approach for fusing imaging datasets of the individual components into a common multivariable trajectory. Data fusion revealed low intrinsic dimensionality of posterior patterning and cell fate specification in wild-type embryos. The simple structure that we uncovered allowed us to construct a model of interactions within the posterior patterning regulatory network and make testable predictions about its dynamics at the protein level. The presented data fusion strategy is a step toward establishing a unified framework that would explore how stochastic spatiotemporal signals give rise to highly reproducible morphogenetic outcomes.


Subject(s)
Body Patterning , Drosophila Proteins , Drosophila melanogaster , Endoderm , Gene Regulatory Networks , Animals , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Endoderm/growth & development , Gene Expression Regulation, Developmental
2.
Science ; 375(6580): 574-578, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35113719

ABSTRACT

The mammalian blastocyst consists of three distinct cell types: epiblast, trophoblast (TB), and primitive endoderm (PrE). Although embryonic stem cells (ESCs) and trophoblast stem cells (TSCs) retain the functional properties of epiblast and TB, respectively, stem cells that fully recapitulate the developmental potential of PrE have not been established. Here, we report derivation of primitive endoderm stem cells (PrESCs) in mice. PrESCs recapitulate properties of embryonic day 4.5 founder PrE, are efficiently incorporated into PrE upon blastocyst injection, generate functionally competent PrE-derived tissues, and support fetal development of PrE-depleted blastocysts in chimeras. Furthermore, PrESCs can establish interactions with ESCs and TSCs and generate descendants with yolk sac-like structures in utero. Establishment of PrESCs will enable the elucidation of the mechanisms for PrE specification and subsequent pre- and postimplantation development.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Endoderm/cytology , Endoderm/embryology , Animals , Blastocyst/cytology , Blastocyst/physiology , Cell Differentiation , Cell Line , Cell Lineage , Chimera , Embryonic Development , Endoderm/growth & development , Fetal Development , Germ Layers/cytology , Germ Layers/embryology , Mice , Mice, Inbred C57BL , Trophoblasts/cytology , Trophoblasts/physiology
3.
Nat Commun ; 12(1): 3277, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34078907

ABSTRACT

Generating properly differentiated embryonic structures in vitro from pluripotent stem cells remains a challenge. Here we show that instruction of aggregates of mouse embryonic stem cells with an experimentally engineered morphogen signalling centre, that functions as an organizer, results in the development of embryo-like entities (embryoids). In situ hybridization, immunolabelling, cell tracking and transcriptomic analyses show that these embryoids form the three germ layers through a gastrulation process and that they exhibit a wide range of developmental structures, highly similar to neurula-stage mouse embryos. Embryoids are organized around an axial chordamesoderm, with a dorsal neural plate that displays histological properties similar to the murine embryo neuroepithelium and that folds into a neural tube patterned antero-posteriorly from the posterior midbrain to the tip of the tail. Lateral to the chordamesoderm, embryoids display somitic and intermediate mesoderm, with beating cardiac tissue anteriorly and formation of a vasculature network. Ventrally, embryoids differentiate a primitive gut tube, which is patterned both antero-posteriorly and dorso-ventrally. Altogether, embryoids provide an in vitro model of mammalian embryo that displays extensive development of germ layer derivatives and that promises to be a powerful tool for in vitro studies and disease modelling.


Subject(s)
Body Patterning/genetics , Embryoid Bodies/metabolism , Embryonic Development/genetics , Mouse Embryonic Stem Cells/metabolism , Signal Transduction/genetics , Animals , Ectoderm/cytology , Ectoderm/growth & development , Ectoderm/metabolism , Embryo, Mammalian , Embryoid Bodies/cytology , Endoderm/cytology , Endoderm/growth & development , Endoderm/metabolism , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gastrula/cytology , Gastrula/growth & development , Gastrula/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , HMGB Proteins/genetics , HMGB Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neural Tube/cytology , Neural Tube/growth & development , Neural Tube/metabolism , Notochord/cytology , Notochord/growth & development , Notochord/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
4.
In Vitro Cell Dev Biol Anim ; 57(5): 493-500, 2021 May.
Article in English | MEDLINE | ID: mdl-33977398

ABSTRACT

LIM kinases are involved in various cellular events such as migration, cycle, and differentiation, but whether they have a role in the specification of mammalian early endoderm remains unclear. In the present study, we found that depletion of LIMK2 severely inhibited the generation of definitive endoderm (DE) from human embryonic stem cells (hESCs) and promoted an early neuroectodermal fate. Upon the silencing of LIMK2 during the endodermal differentiation, the assembly of actin stress fibers was disturbed, and the phosphorylation of cofilin was decreased. In addition, knockdown of LIMK2 during DE differentiation also interfered the upregulation of epithelial-to-mesenchymal transition (EMT)-related genes and cell migration. Collectively, the results highlight that the serine/threonine kinase LIMK2, acting as a key regulator in actin remodeling, plays a critical role in endodermal lineage determination.


Subject(s)
Actin Cytoskeleton/metabolism , Endoderm/growth & development , Lim Kinases/metabolism , Blotting, Western , Cell Movement , Epithelial-Mesenchymal Transition , Fluorescent Antibody Technique , Gene Knockdown Techniques , Human Embryonic Stem Cells , Humans , Reverse Transcriptase Polymerase Chain Reaction
5.
Nat Protoc ; 16(3): 1581-1599, 2021 03.
Article in English | MEDLINE | ID: mdl-33580232

ABSTRACT

The endodermal germ layer gives rise to respiratory epithelium, hepatocytes, pancreatic cells and intestinal lineages, among other cell types. These lineages can be differentiated from human pluripotent stem cells (hPSCs) via a common definitive endoderm (DE) intermediate that is characterized by the co-expression of the cell surface markers CXCR4, c-KIT and EPCAM and the transcription factors SOX17 and FOXA2. Here we provide a detailed protocol for mass production of DE from hPSCs in scalable and easy-to-handle suspension culture using a rotating Erlenmeyer flask or a sophisticated, fully controllable, 150-ml stirred tank bioreactor. This protocol uses two different media formulations that are chemically defined and xeno free and therefore good manufacturing practice ready. Our protocol allows for efficient hPSC-derived DE specification in multicellular aggregates within 3 days and generates up to 1 × 108 DE cells with >92% purity in one differentiation batch when using the bioreactor. The hPSC-derived DE cells that are generated can be cryopreserved for later downstream differentiation into various endodermal lineages. This protocol should facilitate the flexible production of mature DE derivatives for physiologically relevant disease models, high-throughput drug screening, toxicology testing and cellular therapies.


Subject(s)
Cell Culture Techniques/methods , Endoderm/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Cryopreservation/methods , Culture Media , Endoderm/growth & development , Endoderm/metabolism , Hepatocytes/cytology , Humans , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology
7.
Development ; 147(24)2020 12 23.
Article in English | MEDLINE | ID: mdl-33158927

ABSTRACT

Pharyngeal arches (PAs) are segmented by endodermal outpocketings called pharyngeal pouches (PPs). Anterior and posterior PAs appear to be generated by different mechanisms, but it is unclear how the anterior and posterior PAs combine. Here, we addressed this issue with precise live imaging of PP development and cell tracing of pharyngeal endoderm in zebrafish embryos. We found that two endodermal bulges are initially generated in the future second PP (PP2) region, which separates anterior and posterior PAs. Subsequently, epithelial remodeling causes contact between these two bulges, resulting in the formation of mature PP2 with a bilayered morphology. The rostral and caudal bulges develop into the operculum and gill, respectively. Development of the caudal PP2 and more posterior PPs is affected by impaired retinoic acid signaling or pax1a/b dysfunction, suggesting that the rostral front of posterior PA development corresponds to the caudal PP2. Our study clarifies an aspect of PA development that is essential for generation of a seamless array of PAs in zebrafish.


Subject(s)
Branchial Region/growth & development , Embryonic Development/genetics , Endoderm/growth & development , Paired Box Transcription Factors/genetics , Zebrafish Proteins/genetics , Animals , Body Patterning/genetics , Embryo, Nonmammalian , Endoderm/metabolism , Epithelium/growth & development , Epithelium/metabolism , Gene Expression Regulation, Developmental/genetics , Gills/growth & development , Mesoderm/growth & development , Neural Crest/growth & development , Pharynx/growth & development , Signal Transduction/genetics , Zebrafish/genetics , Zebrafish/growth & development
8.
Exp Hematol ; 89: 37-42, 2020 09.
Article in English | MEDLINE | ID: mdl-32735907

ABSTRACT

The extra-embryonic hypoblast/visceral endoderm of Placentalia carries out a variety of functions during gestation, including hematopoietic induction. Results of decades-old and recent experiments have provided compelling evidence that, in addition to its inducing properties, hypoblast/visceral endoderm itself is a source of placental blood cells. Those observations that highlight extra-embryonic endoderm's role as an overlooked source of placental blood cells across species are briefly discussed here, with suggestions for future exploration.


Subject(s)
Allantois/cytology , Blood Cells/cytology , Endoderm/cytology , Erythroblasts/cytology , Placenta/cytology , Yolk Sac/cytology , Allantois/growth & development , Allantois/metabolism , Animals , Blood Cells/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Endoderm/growth & development , Endoderm/metabolism , Erythroblasts/metabolism , Female , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Patched-1 Receptor/genetics , Patched-1 Receptor/metabolism , Placenta/metabolism , Pregnancy , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Yolk Sac/growth & development , Yolk Sac/metabolism
9.
Mech Dev ; 163: 103634, 2020 09.
Article in English | MEDLINE | ID: mdl-32711047

ABSTRACT

All pancreatic cell populations arise from the standard gut endoderm layer in developing embryos, requiring a regulatory gene network to originate and maintain endocrine lineages and endocrine function. The pancreatic organogenesis is regulated by the temporal expression of transcription factors and plays a diverse role in the specification, development, differentiation, maturation, and functional maintenance. Altered expression and activity of these transcription factors are often associated with diabetes mellitus. Recent advancements in the stem cells and invitro derived islets to treat diabetes mellitus has attracted a great deal of interest in the understanding of factors regulating the development, differentiation, and functions of islets including transcription factors. This review discusses the myriad of transcription factors regulating the development of the pancreas, differentiation of ß-islets, and how these factors regulated in normal and disease states. Exploring these factors in such critical context and exogenous or endogenous expression of development and differentiation-specific transcription factors with improved epigenetic plasticity/signaling axis in diabetic milieu would useful for the development of ß-cells from other cell sources.


Subject(s)
Diabetes Mellitus/therapy , Insulin-Secreting Cells/cytology , Insulin/genetics , Organogenesis/genetics , Regeneration/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Endoderm/growth & development , Endoderm/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Pancreas/growth & development , Pancreas/pathology , Stem Cells/cytology , Transcription Factors/genetics
10.
Mech Dev ; 163: 103628, 2020 09.
Article in English | MEDLINE | ID: mdl-32603823

ABSTRACT

Among the basally branching metazoans, cnidarians display well-defined gastrulation processes leading to a diploblastic body plan, consisting of an endodermal and an ectodermal cell layer. As the outgroup to all Bilateria, cnidarians are an interesting group to investigate ancestral developmental mechanisms. Interestingly, all known gastrulation mechanisms known in Bilateria are already found in different species of Cnidaria. Here I review the morphogenetic processes found in different Cnidaria and focus on the investigation of the cellular and molecular mechanisms in the sea anemone Nematostella vectensis, which has been a major model organism among cnidarians for evolutionary developmental biology. Many of the genes involved in germ layer specification and morphogenetic processes in Bilateria are also found active during gastrulation of Nematostella and other cnidarians, suggesting an ancestral role of this process. The molecular analyses indicate a tight link between gastrulation and axis patterning processes by Wnt and FGF signaling. Interestingly, the endodermal layer displays many features of the mesodermal layer in Bilateria, while the pharyngeal ectoderm has an endodermal expression profile. Comparative analyses as well as experimental studies using embryonic aggregates suggest that minor differences in the gene regulatory networks allow the embryo to transition relatively easily from one mode of gastrulation to another.


Subject(s)
Body Patterning/genetics , Cnidaria/genetics , Gastrulation/genetics , Germ Layers/growth & development , Animals , Cnidaria/growth & development , Ectoderm/growth & development , Embryo, Nonmammalian/physiology , Endoderm/growth & development , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Germ Layers/metabolism , Mesoderm/growth & development , Sea Anemones/genetics , Sea Anemones/growth & development , Signal Transduction/genetics
11.
Mech Dev ; 163: 103629, 2020 09.
Article in English | MEDLINE | ID: mdl-32615151

ABSTRACT

Gastrulation is generally understood as the morphogenetic processes that result in the spatial organization of the blastomere into the three germ layers, ectoderm, mesoderm and endoderm. This review summarizes our current knowledge of the morphogenetic mechanisms in Drosophila gastrulation. In addition to the events that drive mesoderm invagination and germband elongation, we pay particular attention to other, less well-known mechanisms including midgut invagination, cephalic furrow formation, dorsal fold formation, and mesoderm layer formation. This review covers topics ranging from the identification and functional characterization of developmental and morphogenetic control genes to the analysis of the physical properties of cells and tissues and the control of cell and tissue mechanics of the morphogenetic movements in the gastrula.


Subject(s)
Drosophila melanogaster/genetics , Gastrula/growth & development , Gastrulation/genetics , Morphogenesis/genetics , Animals , Biomechanical Phenomena/genetics , Drosophila melanogaster/growth & development , Ectoderm/growth & development , Embryo, Nonmammalian , Endoderm/growth & development , Gastrula/metabolism , Gene Expression Regulation, Developmental/genetics , Mesoderm/growth & development
12.
Mech Dev ; 163: 103625, 2020 09.
Article in English | MEDLINE | ID: mdl-32526279

ABSTRACT

The Brachyury gene encodes a transcription factor that is conserved across all animals. In non-chordate metazoans, brachyury is primarily expressed in ectoderm regions that are added to the endodermal gut during development, and often form a ring around the site of endoderm internalization in the gastrula, the blastopore. In chordates, this brachyury ring is conserved, but the gene has taken on a new role in the formation of the mesoderm. In this phylum, a novel type of mesoderm that develops into notochord and somites has been added to the ancestral lateral plate mesoderm. Brachyury contributes to a shift in cell fate from neural ectoderm to posterior notochord and somites during a major lineage segregation event that in Xenopus and in the zebrafish takes place in the early gastrula. In the absence of this brachyury function, impaired formation of posterior mesoderm indirectly affects the gastrulation movements of peak involution and convergent extension. These movements are confined to specific regions and stages, leaving open the question why brachyury expression in an extensive, coherent ring, before, during and after gastrulation, is conserved in the two species whose gastrulation modes differ considerably, and also in many other metazoan gastrulae of diverse structure.


Subject(s)
Ectoderm/growth & development , Fetal Proteins/genetics , Gastrula/growth & development , Morphogenesis/genetics , T-Box Domain Proteins/genetics , Animals , Endoderm/growth & development , Fetal Proteins/ultrastructure , Mesoderm/growth & development , Notochord/growth & development , T-Box Domain Proteins/ultrastructure , Xenopus laevis/genetics , Xenopus laevis/growth & development , Zebrafish/genetics , Zebrafish/growth & development
13.
Mech Dev ; 163: 103617, 2020 09.
Article in English | MEDLINE | ID: mdl-32473204

ABSTRACT

During mouse embryonic development a mass of pluripotent epiblast tissue is transformed during gastrulation to generate the three definitive germ layers: endoderm, mesoderm, and ectoderm. During gastrulation, a spatiotemporally controlled sequence of events results in the generation of organ progenitors and positions them in a stereotypical fashion throughout the embryo. Key to the correct specification and differentiation of these cell fates is the establishment of an axial coordinate system along with the integration of multiple signals by individual epiblast cells to produce distinct outcomes. These signaling domains evolve as the anterior-posterior axis is established and the embryo grows in size. Gastrulation is initiated at the posteriorly positioned primitive streak, from which nascent mesoderm and endoderm progenitors ingress and begin to diversify. Advances in technology have facilitated the elaboration of landmark findings that originally described the epiblast fate map and signaling pathways required to execute those fates. Here we will discuss the current state of the field and reflect on how our understanding has shifted in recent years.


Subject(s)
Body Patterning/genetics , Cell Differentiation/genetics , Embryonic Development/genetics , Gastrulation/genetics , Organ Specificity/genetics , Animals , Cell Lineage/genetics , Ectoderm/growth & development , Endoderm/growth & development , Female , Gastrula/growth & development , Gastrulation/physiology , Germ Layers/growth & development , Mesoderm/growth & development , Mice , Pregnancy
14.
Genetics ; 214(3): 543-560, 2020 03.
Article in English | MEDLINE | ID: mdl-32132154

ABSTRACT

A critical juncture in early development is the partitioning of cells that will adopt different fates into three germ layers: the ectoderm, the mesoderm, and the endoderm. This step is achieved through the internalization of specified cells from the outermost surface layer, through a process called gastrulation. In Drosophila, gastrulation is achieved through cell shape changes (i.e., apical constriction) that change tissue curvature and lead to the folding of a surface epithelium. Folding of embryonic tissue results in mesoderm and endoderm invagination, not as individual cells, but as collective tissue units. The tractability of Drosophila as a model system is best exemplified by how much we know about Drosophila gastrulation, from the signals that pattern the embryo to the molecular components that generate force, and how these components are organized to promote cell and tissue shape changes. For mesoderm invagination, graded signaling by the morphogen, Spätzle, sets up a gradient in transcriptional activity that leads to the expression of a secreted ligand (Folded gastrulation) and a transmembrane protein (T48). Together with the GPCR Mist, which is expressed in the mesoderm, and the GPCR Smog, which is expressed uniformly, these signals activate heterotrimeric G-protein and small Rho-family G-protein signaling to promote apical contractility and changes in cell and tissue shape. A notable feature of this signaling pathway is its intricate organization in both space and time. At the cellular level, signaling components and the cytoskeleton exhibit striking polarity, not only along the apical-basal cell axis, but also within the apical domain. Furthermore, gene expression controls a highly choreographed chain of events, the dynamics of which are critical for primordium invagination; it does not simply throw the cytoskeletal "on" switch. Finally, studies of Drosophila gastrulation have provided insight into how global tissue mechanics and movements are intertwined as multiple tissues simultaneously change shape. Overall, these studies have contributed to the view that cells respond to forces that propagate over great distances, demonstrating that cellular decisions, and, ultimately, tissue shape changes, proceed by integrating cues across an entire embryo.


Subject(s)
Endoderm/growth & development , Gastrulation/genetics , Mesoderm/growth & development , Physical Phenomena , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Embryo, Nonmammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/genetics , Morphogenesis/genetics , Signal Transduction/genetics , rho GTP-Binding Proteins/genetics
15.
RNA ; 26(5): 550-563, 2020 05.
Article in English | MEDLINE | ID: mdl-32075940

ABSTRACT

Transposable elements (TEs) can damage genomes, thus organisms use a variety of mechanisms to repress TE expression. The PIWI-piRNA pathway is a small RNA pathway that represses TE expression in the germline of animals. Here we explore the function of the pathway in the somatic stem cells of Hydra, a long-lived freshwater cnidarian. Hydra have three stem cell populations, all of which express PIWI proteins; endodermal and ectodermal epithelial stem cells (ESCs) are somatic, whereas the interstitial stem cells have germline competence. To study somatic function of the pathway, we isolated piRNAs from Hydra that lack the interstitial lineage and found that these somatic piRNAs map predominantly to TE transcripts and display the conserved sequence signatures typical of germline piRNAs. Three lines of evidence suggest that the PIWI-piRNA pathway represses TEs in Hydra ESCs. First, epithelial knockdown of the Hydra piwi gene hywi resulted in up-regulation of TE expression. Second, degradome sequencing revealed evidence of PIWI-mediated cleavage of TE RNAs in epithelial cells using the ping-pong mechanism. Finally, we demonstrated a direct association between Hywi protein and TE transcripts in epithelial cells using RNA immunoprecipitation. Altogether, our data reveal that the PIWI-piRNA pathway represses TE expression in the somatic cell lineages of Hydra, which we propose contributes to the extreme longevity of the organism. Furthermore, our results, in combination with others, suggest that somatic TE repression is an ancestral function of the PIWI-piRNA pathway.


Subject(s)
Cell Lineage/genetics , DNA Transposable Elements/genetics , Hydra/genetics , RNA, Small Interfering/genetics , Animals , Argonaute Proteins/genetics , Ectoderm/growth & development , Ectoderm/metabolism , Endoderm/growth & development , Endoderm/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Silencing , Hydra/growth & development , RNA Interference , Stem Cells/cytology
16.
Genes Genet Syst ; 95(1): 1-10, 2020 Apr 22.
Article in English | MEDLINE | ID: mdl-31839648

ABSTRACT

MicroRNAs are a class of short non-coding RNAs that contain approximately 22 nucleotides and play a regulatory role in RNA silencing and translational repression. miR-92 belongs to the miR-17-92 family and has a regulatory effect on cell proliferation, apoptosis, and expression of proto-oncogenes and tumor suppressor genes. However, its function in flatfish is unclear. In this study, we used farmed Japanese flounder, Paralichthys olivaceus, and showed that gata5 is a target gene of miR-92. Experiments on miR-92 overexpression indicated that gata5 and sox17 were downregulated, while the transcription level of ntl increased. By contrast, depletion of miR-92 resulted in increased gata5 and sox17 levels and reduced ntl level. Moreover, thiourea treatment indicated that miR-92 may inhibit the metamorphic development of Japanese flounder. Our study suggests that miR-92 regulates the fate of endoderm and mesoderm by controlling gata5.


Subject(s)
Flounder/genetics , Gene Expression Regulation, Developmental/genetics , MicroRNAs/genetics , Amino Acid Sequence , Animals , Endoderm/growth & development , Female , Flounder/growth & development , GATA5 Transcription Factor/genetics , GATA5 Transcription Factor/metabolism , Genes, Reporter , Japan , Male , Mesoderm/growth & development , Metamorphosis, Biological , Zebrafish
17.
Differentiation ; 112: 17-26, 2020.
Article in English | MEDLINE | ID: mdl-31869687

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are potentially an invaluable source of cells for regenerative medicine, disease modeling and drug discovery. However, the differentiation of hiPSCs into fully functional hepatocytes remains a major challenge. Despite the importance of the information carried by metabolomes, the exploitation of metabolomics for characterizing and understanding hiPSC differentiation remains largely unexplored. Here, to increase knowledge of hiPSC maturation into mature hepatocytes, we investigated their metabolomics profiles during sequential step-by-step differentiation: definitive endoderm (DE), specification into hepatocytes (HB-pro (hepatoblast progenitors)), progenitor hepatocytes (Pro-HEP) and mature hepatocyte-like cells (HLCs). Metabolomics analysis illustrated a switch from glycolysis-based respiration in DE step to oxidative phosphorylation in HLCs step. DE was characterized by fatty acid beta oxidation, sorbitol metabolism and pentose phosphate pathway, and glutamine and glucose metabolisms as various potential energy sources. The complex lipid metabolism switch was monitored via the reduction of lipid production from DE to HLCs step, whereas high glycerol production occurred mainly in HLCs. The nitrogen cycle, via urea production, was also a typical mechanism revealed in HLCs step. Our analysis may contribute to better understanding of differentiation and suggest new targets for improving iPSC maturation into functional hepatocytes.


Subject(s)
Cell Differentiation/genetics , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Metabolome/genetics , Endoderm/growth & development , Endoderm/metabolism , Gene Expression Regulation, Developmental/genetics , Glucose/genetics , Glucose/metabolism , Glutamine/genetics , Glutamine/metabolism , Glycolysis/genetics , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Lipid Metabolism/genetics , Metabolomics/methods , Oxidative Phosphorylation
18.
Nat Commun ; 10(1): 5658, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827102

ABSTRACT

Organoids have extensive therapeutic potential and are increasingly opening up new avenues within regenerative medicine. However, their clinical application is greatly limited by the lack of effective GMP-compliant systems for organoid expansion in culture. Here, we envisage that the use of extracellular matrix (ECM) hydrogels derived from decellularized tissues (DT) can provide an environment capable of directing cell growth. These gels possess the biochemical signature of tissue-specific ECM and have the potential for clinical translation. Gels from decellularized porcine small intestine (SI) mucosa/submucosa enable formation and growth of endoderm-derived human organoids, such as gastric, hepatic, pancreatic, and SI. ECM gels can be used as a tool for direct human organoid derivation, for cell growth with a stable transcriptomic signature, and for in vivo organoid delivery. The development of these ECM-derived hydrogels opens up the potential for human organoids to be used clinically.


Subject(s)
Endoderm/growth & development , Extracellular Matrix/metabolism , Organoids/growth & development , Animals , Cell Proliferation , Endoderm/metabolism , Extracellular Matrix/chemistry , Humans , Hydrogels/chemistry , Hydrogels/metabolism , Organoids/metabolism , Swine , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry
19.
Stem Cells Dev ; 28(19): 1334-1345, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31337269

ABSTRACT

Smooth muscle cells (SMCs) are important cell type for regenerative medicine. Previous studies showed that retinoic acid (RA) induces differentiation of SMCs from monolayer-cultured embryonic stem cells (ESCs) with high efficiency. However, the underlying mechanisms are still poorly defined. Here, we identified Wnt signaling as a primary regulator for RA-induced ESC differentiation. The activation of Wnt signaling inhibited the epithelial-mesenchymal transition during ESC differentiation, leading to inhibition of RA-induced SMC differentiation and promoting differentiation of ESCs toward primitive endoderm (PrE) lineage instead, while the inhibition of Wnt signaling promoted RA-induced SMC differentiation. Loss-of-function studies revealed that 7-like 2 (Tcf7l2) was the key transcription factor that Wnt operate through during RA-induced differentiation. Thus, this study revealed that the Tcf7l2-mediated Wnt signaling is a switch in determining the mesoderm/PrE fates in RA-induced ESC differentiation.


Subject(s)
Cell Differentiation/drug effects , Mouse Embryonic Stem Cells/drug effects , Myocytes, Smooth Muscle/drug effects , Tretinoin/pharmacology , Wnt Signaling Pathway/drug effects , Actins/genetics , Actins/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Endoderm/cytology , Endoderm/growth & development , Endoderm/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Feeder Cells , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Heterocyclic Compounds, 3-Ring/pharmacology , Mesoderm/cytology , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Wnt Signaling Pathway/genetics , Calponins
20.
In Vitro Cell Dev Biol Anim ; 55(5): 355-367, 2019 May.
Article in English | MEDLINE | ID: mdl-30993557

ABSTRACT

N-terminal acetylation (Nt-acetylation) refers to the acetylation of the free α-amino group at the N-terminus of a polypeptide. While the effects of Nt-acetylation are multifaceted, its most known function is in the acetylation-dependent N-end rule protein degradation pathway (Ac/N-end rule pathway), where Nt-acetylation is recognized as a degron by designated E3 ligases, eventually leading to target degradation by the ubiquitin-proteasome system. Naa10 is the catalytic subunit of the major Nt-acetylation enzyme NatA, which Nt-acetylates proteins whose second amino acid has a small side chain. In humans, NAA10 is the responsible mutated gene in Ogden syndrome and is thought to play important roles in development. However, it is unclear how the Ac/N-end rule pathway affects the differentiation ability of mouse embryonic stem cells (mESCs). We hypothesized that the balance of pluripotency factors may be maintained by the Ac/N-end rule pathway. Thus, we established Naa10 knockout mESCs to test this hypothesis. We found that Naa10 deficiency attenuated differentiation towards the epiblast lineage, deviating towards primitive endoderm. However, this was not caused by disturbing the balance of pluripotency factors, rather by augmenting FGF/MAPK signaling.


Subject(s)
Cell Lineage/genetics , Germ Layers/growth & development , Mouse Embryonic Stem Cells/metabolism , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase E/genetics , Acetylation , Animals , Cell Differentiation/genetics , Endoderm/growth & development , Endoderm/metabolism , Fibroblast Growth Factors/genetics , Gene Knockout Techniques , Germ Layers/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/metabolism , Protein Processing, Post-Translational/genetics , Proteolysis , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...