Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 815
1.
Dev Biol ; 478: 183-204, 2021 10.
Article En | MEDLINE | ID: mdl-34216573

The mechanisms regulating nervous system development are still unknown for a wide variety of taxa. In insects and vertebrates, bone morphogenetic protein (BMP) signaling plays a key role in establishing the dorsal-ventral (D-V) axis and limiting the neuroectoderm to one side of that axis, leading to speculation about the conserved evolution of centralized nervous systems. Studies outside of insects and vertebrates show a more diverse picture of what, if any role, BMP signaling plays in neural development across Bilateria. This is especially true in the morphologically diverse Spiralia (≈Lophotrochozoa). Despite several studies of D-V axis formation and neural induction in spiralians, there is no consensus for how these two processes are related, or whether BMP signaling may have played an ancestral role in either process. To determine the function of BMP signaling during early development of the spiralian annelid Capitella teleta, we incubated embryos and larvae in BMP4 protein for different amounts of time. Adding exogenous BMP protein to early-cleaving C. teleta embryos had a striking effect on formation of the brain, eyes, foregut, and ventral midline in a time-dependent manner. However, adding BMP did not block brain or VNC formation or majorly disrupt the D-V axis. We identified three key time windows of BMP activity. 1) BMP treatment around birth of the 3rd-quartet micromeres caused the loss of the eyes, radialization of the brain, and a reduction of the foregut, which we interpret as a loss of A- and C-quadrant identities with a possible trans-fate switch to a D-quadrant identity. 2) Treatment after the birth of micromere 4d induced formation of a third ectopic brain lobe, eye, and foregut lobe, which we interpret as a trans-fate switch of B-quadrant micromeres to a C-quadrant identity. 3) Continuous BMP treatment from late cleavage (4d â€‹+ â€‹12 â€‹h) through mid-larval stages resulted in a modest expansion of Ct-chrdl expression in the dorsal ectoderm and a concomitant loss of the ventral midline (neurotroch ciliary band). Loss of the ventral midline was accompanied by a collapse of the bilaterally-symmetric ventral nerve cord, although the total amount of neural tissue was not greatly affected. Our results compared with those from other annelids and molluscs suggest that BMP signaling was not ancestrally involved in delimiting neural tissue to one region of the D-V axis. However, the effects of ectopic BMP on quadrant-identity during cleavage stages may represent a non-axial organizing signal that was present in the last common ancestor of annelids and mollusks. Furthermore, in the last common ancestor of annelids, BMP signaling may have functioned in patterning ectodermal fates along the D-V axis in the trunk. Ultimately, studies on a wider range of spiralian taxa are needed to determine the role of BMP signaling during neural induction and neural patterning in the last common ancestor of this group. Ultimately, these comparisons will give us insight into the evolutionary origins of centralized nervous systems and body plans.


Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Proteins/metabolism , Polychaeta/embryology , Polychaeta/metabolism , Zebrafish Proteins/pharmacology , Animals , Body Patterning/drug effects , Bone Morphogenetic Proteins/genetics , Brain/embryology , Digestive System/embryology , Embryo, Nonmammalian/metabolism , Embryonic Development , Eye/embryology , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Polychaeta/drug effects , Polychaeta/growth & development , Recombinant Proteins/pharmacology , Signal Transduction , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Smad8 Protein/genetics , Smad8 Protein/metabolism
2.
J Cell Biol ; 220(8)2021 08 02.
Article En | MEDLINE | ID: mdl-34047771

Mesenchymal-to-epithelial transition (MET) converts cells from migratory mesenchymal to polarized epithelial states. Despite its importance for both normal and pathological processes, very little is known about the regulation of MET in vivo. Here we exploit midgut morphogenesis in Drosophila melanogaster to investigate the mechanisms underlying MET. We show that down-regulation of the EMT transcription factor Serpent is required for MET, but not sufficient, as interactions with the surrounding mesoderm are also essential. We find that midgut MET relies on the secretion of specific laminins via the CopII secretory pathway from both mesoderm and midgut cells. We show that secretion of the laminin trimer containing the Wingblister α-subunit from the mesoderm is an upstream cue for midgut MET, leading to basal polarization of αPS1 integrin in midgut cells. Polarized αPS1 is required for the formation of a monolayered columnar epithelium and for the apical polarization of αPS3, Baz, and E-Cad. Secretion of a distinct LamininA-containing trimer from midgut cells is required to reinforce the localization of αPS1 basally, and αPS3 apically, for robust repolarization. Our data suggest that targeting these MET pathways, in conjunction with therapies preventing EMT, may present a two-pronged strategy toward blocking metastasis in cancer.


Digestive System/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Laminin/metabolism , Animals , Animals, Genetically Modified , COP-Coated Vesicles/genetics , COP-Coated Vesicles/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Movement , Cell Polarity , Digestive System/embryology , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Laminin/genetics , Microscopy, Confocal , Microscopy, Fluorescence , Signal Transduction , Time Factors , Time-Lapse Imaging
3.
Cells ; 10(3)2021 03 12.
Article En | MEDLINE | ID: mdl-33809074

Human GBA1 encodes lysosomal acid ß-glucocerebrosidase (GCase), which hydrolyzes cleavage of the beta-glucosidic linkage of glucosylceramide (GlcCer). Mutations in this gene lead to reduced GCase activity, accumulation of glucosylceramide and glucosylsphingosine, and development of Gaucher disease (GD). Drosophila melanogaster has two GBA1 orthologs. Thus far, GBA1b was documented as a bone fide GCase-encoding gene, while the role of GBA1a encoded protein remained unclear. In the present study, we characterized a mutant variant of the fly GBA1a, which underwent ERAD and mildly activated the UPR machinery. RNA-seq analyses of homozygous mutant flies revealed upregulation of inflammation-associated as well as of cell-cycle related genes and reduction in programmed cell death (PCD)-associated genes, which was confirmed by qRT-PCR. We also observed compromised cell death in the midgut of homozygous larvae and a reduction in pupation. Our results strongly indicated that GBA1a-encoded protein plays a role in midgut maturation during larvae development.


Digestive System/enzymology , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Glucosylceramidase/metabolism , Animals , Animals, Genetically Modified , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Digestive System/embryology , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Endoplasmic Reticulum-Associated Degradation , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Glucosylceramidase/genetics , Homozygote , Inflammation Mediators/metabolism , Locomotion , Longevity , Morphogenesis , Mutation , Signal Transduction , Transcriptome
4.
Dev Biol ; 474: 37-47, 2021 06.
Article En | MEDLINE | ID: mdl-33571486

Canonical Wnt signaling plays a key role during organ development, homeostasis and regeneration and these processes are conserved between invertebrates and vertebrates. Mutations in Wnt pathway components are commonly found in various types of cancer. Upon activation of canonical Wnt signaling, ß-catenin binds in the nucleus to members of the TCF-LEF family and activates the transcription of target genes. Multiple Wnt target genes, including Lgr5/LGR5 and Axin2/AXIN2, have been identified in mouse models and human cancer cell lines. Here we set out to identify the transcriptional targets of Wnt signaling in five human tissues using organoid technology. Organoids are derived from adult stem cells and recapitulate the functionality as well as the structure of the original tissue. Since the Wnt pathway is critical to maintain the organoids from the human intestine, colon, liver, pancreas and stomach, organoid technology allows us to assess Wnt target gene expression in a human wildtype situation. We performed bulk mRNA sequencing of organoids immediately after inhibition of Wnt pathway and identified 41 genes as commonly regulated genes in these tissues. We also identified large numbers of target genes specific to each tissue. One of the shared target genes is TEAD4, a transcription factor driving expression of YAP/TAZ signaling target genes. In addition to TEAD4, we identified a variety of genes which encode for proteins that are involved in Wnt-independent pathways, implicating the possibility of direct crosstalk between Wnt signaling and other pathways. Collectively, this study identified tissue-specific and common Wnt target gene signatures and provides evidence for a conserved role for these Wnt targets in different tissues.


Digestive System/cytology , Gene Expression Regulation, Developmental , Organoids/metabolism , Wnt Signaling Pathway , Adult , Digestive System/embryology , Digestive System/metabolism , Endoderm , Gene Expression Profiling , Humans , Organ Specificity
5.
Thorac Cardiovasc Surg ; 69(1): 2-7, 2021 01.
Article En | MEDLINE | ID: mdl-31756748

Organ systems do not exist in a vacuum. However, in an era of increasingly specialized medicine, the focus is often on the organ system alone. Many symptoms are associated with differential diagnoses from upper gastrointestinal (GI) and cardiovascular medical and surgical specialties. Furthermore, a large number of rare but deadly conditions cross paths between the upper GI tract and cardiovascular system; a significant proportion of these are iatrogenic injuries from a parallel specialty. These include unusual fistulae, herniae, and embolisms that transcend specialties. This review highlights these conditions and the shared anatomy and embryology of the two organ systems.


Cardiovascular Diseases/etiology , Cardiovascular System/physiopathology , Digestive System Diseases/etiology , Digestive System/physiopathology , Iatrogenic Disease , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/therapy , Cardiovascular System/embryology , Digestive System/embryology , Digestive System Diseases/diagnosis , Digestive System Diseases/physiopathology , Digestive System Diseases/therapy , Humans , Morphogenesis , Prognosis , Risk Assessment , Risk Factors
6.
Dev Biol ; 470: 1-9, 2021 02.
Article En | MEDLINE | ID: mdl-33191200

Caudal homeobox genes are found across animals, typically linked to two other homeobox genes in what has been called the ParaHox cluster. These genes have been proposed to pattern the anterior-posterior axis of the endoderm ancestrally, but the expression of Caudal in extant groups is varied and often occurs in other germ layers. Here we examine the role of Caudal in the embryo of the mollusc Tritia (Ilyanassa) obsoleta. ToCaudal expression is initially broad, then becomes progressively restricted and is finally only in the developing hindgut (a.k.a. intestine). Knockdown of ToCaudal using morpholino oligonucleotides specifically blocks hindgut development, indicating that despite its initially broad expression, the functional role of ToCaudal is in hindgut patterning. This is the first functional characterization of Caudal in an animal with spiralian development, which is an ancient mode of embryogenesis that arose early in bilaterian animal evolution. These results are consistent with the hypothesis that the ancestral role of the ParaHox genes was anterior-posterior patterning of the endoderm.


Body Patterning/genetics , Gene Expression Regulation, Developmental , Genes, Homeobox , Snails/embryology , Snails/genetics , Animals , Digestive System/embryology , Embryo, Nonmammalian/metabolism , Gene Knockdown Techniques , Germ Layers/embryology , Germ Layers/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Multigene Family , Phylogeny , Snails/metabolism
7.
Nat Commun ; 11(1): 4158, 2020 08 27.
Article En | MEDLINE | ID: mdl-32855417

Visceral organs, such as the lungs, stomach and liver, are derived from the fetal foregut through a series of inductive interactions between the definitive endoderm (DE) and the surrounding splanchnic mesoderm (SM). While DE patterning is fairly well studied, the paracrine signaling controlling SM regionalization and how this is coordinated with epithelial identity is obscure. Here, we use single cell transcriptomics to generate a high-resolution cell state map of the embryonic mouse foregut. This identifies a diversity of SM cell types that develop in close register with the organ-specific epithelium. We infer a spatiotemporal signaling network of endoderm-mesoderm interactions that orchestrate foregut organogenesis. We validate key predictions with mouse genetics, showing the importance of endoderm-derived signals in mesoderm patterning. Finally, leveraging these signaling interactions, we generate different SM subtypes from human pluripotent stem cells (hPSCs), which previously have been elusive. The single cell data can be explored at: https://research.cchmc.org/ZornLab-singlecell .


Digestive System/metabolism , Endoderm/metabolism , Gene Regulatory Networks , Mesoderm/metabolism , Organogenesis/genetics , Signal Transduction/genetics , Animals , Cell Lineage/genetics , Digestive System/cytology , Digestive System/embryology , Endoderm/cytology , Endoderm/embryology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Humans , Internet , Mesoderm/cytology , Mesoderm/embryology , Mice, Inbred C57BL , Single-Cell Analysis/methods , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Toxins (Basel) ; 12(6)2020 06 24.
Article En | MEDLINE | ID: mdl-32599715

Evolution of insect resistance to Bt toxins challenges the use of Cry toxins to control agricultural pests. In lepidopterans, Cry toxin affinity towards multiple midgut epithelial receptors has become a matter of dispute. Cry1Ah toxin-binding proteins were identified in the larval midgut of susceptible (ACB-BtS) and resistant (ACB-AhR) strains of the Asian corn borer (ACB). A pull-down assay was performed using biotinylated Cry1Ah toxin, and the binding proteins were identified by employing liquid chromatography-tandem mass spectrometry (LC-MS/MS). This study aimed to find the binding consistency of the midgut epithelial protein to the Cry1Ah toxin. The binding proteins from different fractions of SDS-PAGE showed a different pattern. We observed an isoform of prophenoloxidase PPO1b (UniProt Acc No. A0A1Q1MKI0), which was found only in the ACB-AhR fractions. Prophenoloxidase (proPO) is an extraordinary defense molecule activated in insect species during pathogen invasion and the wound healing process. Importantly, this prophenoloxidase might have direct/indirect interaction with the Cry1Ah toxin. Our data also suggest that factors like techniques, enrichment of binding proteins in the sample and the reversible and irreversible nature of the brush border membrane vesicles (BBMVs) to Cry toxins could cause the inconsistency in the protein-protein interactions. Moreover, inside the larva midgut, the influence of the Cry toxins under physiological conditions might be different from the laboratory procedures.


Bacillus thuringiensis Toxins/metabolism , Digestive System/metabolism , Endotoxins/metabolism , Hemolysin Proteins/metabolism , Insect Proteins/metabolism , Lepidoptera/metabolism , Membrane Proteins/metabolism , Zea mays/parasitology , Animals , Bacillus thuringiensis Toxins/pharmacology , Digestive System/embryology , Endotoxins/pharmacology , Hemolysin Proteins/pharmacology , Insecticide Resistance , Larva/metabolism , Lepidoptera/drug effects , Lepidoptera/embryology , Protein Binding
9.
Bull Math Biol ; 81(7): 2220-2238, 2019 07.
Article En | MEDLINE | ID: mdl-30945102

Growth in biological systems occurs as a consequence of cell proliferation fueled by a nutrient supply. In general, the nutrient gradient of the system will be nonconstant, resulting in biased cell proliferation. We develop a uniaxial discrete cellular automaton with biased cell proliferation using a probability distribution which reflects the nutrient gradient of the system. An explicit probability mass function for the displacement of any tracked cell under the cellular automaton model is derived and verified against averaged simulation results; this displacement distribution has applications in predicting cell trajectories and evolution of expected site occupancies.


Cell Proliferation/physiology , Models, Biological , Algorithms , Animals , Body Patterning/physiology , Cell Movement/physiology , Computer Simulation , Digestive System/embryology , Linear Models , Markov Chains , Mathematical Concepts , Probability , Quail/embryology , Spatio-Temporal Analysis , Systems Analysis
10.
Dev Biol ; 449(1): 1-13, 2019 05 01.
Article En | MEDLINE | ID: mdl-30797757

Wnt proteins can activate different intracellular signaling pathways. These pathways need to be tightly regulated for proper cardiogenesis. The canonical Wnt/ß-catenin inhibitor Dkk1 has been shown to be sufficient to trigger cardiogenesis in gain-of-function experiments performed in multiple model systems. Loss-of-function studies however did not reveal any fundamental function for Dkk1 during cardiogenesis. Using Xenopus laevis as a model we here show for the first time that Dkk1 is required for proper differentiation of cardiomyocytes, whereas specification of cardiomyocytes remains unaffected in absence of Dkk1. This effect is at least in part mediated through regulation of non-canonical Wnt signaling via Wnt11. In line with these observations we also found that Isl1, a critical regulator for specification of the common cardiac progenitor cell (CPC) population, acts upstream of Dkk1.


Cell Differentiation , Intercellular Signaling Peptides and Proteins/metabolism , Myocardium/cytology , Wnt Signaling Pathway , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Animals , Biomarkers/metabolism , Body Patterning , Digestive System/embryology , Digestive System/metabolism , Down-Regulation/genetics , Embryo, Nonmammalian/metabolism , Endoderm/metabolism , Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/metabolism , Mesoderm/metabolism , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Transcription Factors/metabolism , Wnt Proteins/metabolism
11.
Dev Biol ; 446(1): 22-33, 2019 02 01.
Article En | MEDLINE | ID: mdl-30448439

The enteric nervous system is mostly derived from vagal neural crest (NC) cells adjacent to somites (s)1-7. We used in ovo focal fluorescent vital dyes and focal electroporation of fluorophore-encoding plasmids in quail embryos to investigate NC cell migration to the foregut initially and later throughout the entire gut. NC cells of different somite-level origins were largely separate until reaching the foregut at about QE2.5, when all routes converged. By QE3.5, NC cells of different somite-levels became mixed, although s1-s2 NC cells were mainly confined to rostral foregut. Mid-vagal NC-derived cells (s3 and s4 level) arrived earliest at the foregut, and occurred in greatest number. By QE6.5 ENS was present from foregut to hindgut. Mid-vagal NC-derived cells occurred in greatest numbers from foregut to distal hindgut. NC-derived cells of s2, s5, and s6 levels were fewer and were widely distributed but were never observed in the distal hindgut. Rostro-vagal (s1) and caudo-vagal (s7) levels were few and restricted to the foregut. Single somite levels of quail neural tube/NC from s1 to s8 were combined with chick aneural ChE4.5 midgut and hindgut and the ensemble was grown on the chorio-allantoic membrane for 6 days. This tests ENS-forming competence in the absence of intra-segmental competition between NC cells, of differential influences of segmental paraxial tissues, and of positional advantage. All vagal NC-levels, but not s8 level, furnished enteric plexuses in the recipient gut, but the density of both ENS cells in total and neurons was highest from mid-vagal level donors, as was the length colonised. We conclude that the fate and competence for ENS formation of vagal NC sub-levels is not uniform over the vagal level but is biased to favour mid-vagal levels. Overviewing this and prior studies suggests the vagal region is, as in its traditional sense, a natural unit but with complex sub-divisions.


Enteric Nervous System/embryology , Neural Crest/embryology , Somites/embryology , Vagus Nerve/embryology , Animals , Body Patterning , Cell Differentiation , Cell Movement , Chick Embryo , Chickens , Coturnix , Digestive System/cytology , Digestive System/embryology , Digestive System/metabolism , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Intestines/cytology , Intestines/embryology , Intestines/innervation , Neural Crest/cytology , Neural Crest/metabolism , Neurons/cytology , Neurons/metabolism , Somites/cytology , Somites/metabolism , Vagus Nerve/cytology , Vagus Nerve/metabolism
12.
Dev Biol ; 446(1): 34-42, 2019 02 01.
Article En | MEDLINE | ID: mdl-30529057

Cells of the vagal neural crest (NC) form most of the enteric nervous system (ENS) by a colonising wave in the embryonic gut, with high cell proliferation and differentiation. Enteric neuropathies have an ENS deficit and cell replacement has been suggested as therapy. This would be performed post-natally, which raises the question of whether the ENS cell population retains its initial ENS-forming potential with age. We tested this on the avian model in organ culture in vitro (3 days) using recipient aneural chick midgut/hindgut combined with ENS-donor quail midgut or hindgut of ages QE5 to QE10. ENS cells from young donor tissues (≤ QE6) avidly colonised the aneural recipient, but this capacity dropped rapidly 2-3 days after the transit of the ENS cell wavefront. This loss in capability was autonomous to the ENS population since a similar decline was observed in ENS cells isolated by HNK1 FACS. Using QE5, 6, 8 and 10 midgut donors and extending the time of assay to 8 days in chorio-allantoic membrane grafts did not produce 'catch up' colonisation. NC-derived cells were counted in dissociated quail embryo gut and in transverse sections of chick embryo gut using NC, neuron and glial marker antibodies. This showed that the decline in ENS-forming ability correlated with a decrease in proportion of ENS cells lacking both neuronal and glial differentiation markers, but there were still large numbers of such cells even at stages with low colonisation ability. Moreover, ENS cells in small numbers from young donors were far superior in colonisation ability to larger numbers of apparently undifferentiated cells from older donors. This suggests that the decline of ENS-forming ability has both quantitative and qualitative aspects. In this case, ENS cells for cell therapies should aim to replicate the embryonic ENS stage rather than using post-natal ENS stem/progenitor cells.


Digestive System/embryology , Enteric Nervous System/embryology , Intestine, Small/embryology , Neural Crest/embryology , Animals , Cell Differentiation , Cell Movement , Cells, Cultured , Chick Embryo , Chickens , Chorioallantoic Membrane/transplantation , Coturnix , Digestive System/cytology , Digestive System/metabolism , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Intestine, Small/cytology , Intestine, Small/innervation , Neural Crest/cytology , Neural Crest/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Organ Culture Techniques
13.
PLoS Genet ; 14(11): e1007772, 2018 11.
Article En | MEDLINE | ID: mdl-30419011

Cell type-specific modifications of conventional endosomal trafficking pathways lead to the formation of lysosome-related organelles (LROs). C. elegans gut granules are intestinally restricted LROs that coexist with conventional degradative lysosomes. The formation of gut granules requires the Rab32 family member GLO-1. We show that the loss of glo-1 leads to the mistrafficking of gut granule proteins but does not significantly alter conventional endolysosome biogenesis. GLO-3 directly binds to CCZ-1 and they both function to promote the gut granule association of GLO-1, strongly suggesting that together, GLO-3 and CCZ-1 activate GLO-1. We found that a point mutation in GLO-1 predicted to spontaneously activate, and function independently of it guanine nucleotide exchange factor (GEF), localizes to gut granules and partially restores gut granule protein localization in ccz-1(-) and glo-3(-) mutants. CCZ-1 forms a heterodimeric complex with SAND-1(MON1), which does not function in gut granule formation, to activate RAB-7 in trafficking pathways to conventional lysosomes. Therefore, our data suggest a model whereby the function of a Rab GEF can be altered by subunit exchange. glo-3(-) mutants, which retain low levels of GLO-3 activity, generate gut granules that lack GLO-1 and improperly accumulate RAB-7 in a SAND-1 dependent process. We show that GLO-1 and GLO-3 restrict the distribution of RAB-7 to conventional endolysosomes, providing insights into the segregation of pathways leading to conventional lysosomes and LROs.


Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cytoplasmic Granules/metabolism , Digestive System/embryology , Digestive System/metabolism , Genes, Helminth , Lysosomes/metabolism , Mutation , Organelle Biogenesis , Protein Interaction Domains and Motifs , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/genetics
14.
Development ; 145(9)2018 05 08.
Article En | MEDLINE | ID: mdl-29678817

The enteric nervous system (ENS) arises from neural crest cells that migrate, proliferate, and differentiate into enteric neurons and glia within the intestinal wall. Many extracellular matrix (ECM) components are present in the embryonic gut, but their role in regulating ENS development is largely unknown. Here, we identify heparan sulfate proteoglycan proteins, including collagen XVIII (Col18) and agrin, as important regulators of enteric neural crest-derived cell (ENCDC) development. In developing avian hindgut, Col18 is expressed at the ENCDC wavefront, while agrin expression occurs later. Both proteins are normally present around enteric ganglia, but are absent in aganglionic gut. Using chick-mouse intestinal chimeras and enteric neurospheres, we show that vagal- and sacral-derived ENCDCs from both species secrete Col18 and agrin. Whereas glia express Col18 and agrin, enteric neurons only express the latter. Functional studies demonstrate that Col18 is permissive whereas agrin is strongly inhibitory to ENCDC migration, consistent with the timing of their expression during ENS development. We conclude that ENCDCs govern their own migration by actively remodeling their microenvironment through secretion of ECM proteins.


Agrin/metabolism , Avian Proteins/metabolism , Chickens/metabolism , Collagen/metabolism , Digestive System , Neural Crest/embryology , Stem Cell Niche/physiology , Agrin/genetics , Animals , Avian Proteins/genetics , Cell Movement/physiology , Chick Embryo , Chickens/genetics , Collagen/genetics , Digestive System/cytology , Digestive System/embryology , Digestive System/innervation , Gene Expression Regulation, Developmental/physiology , Mice , Neural Crest/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
15.
PLoS One ; 13(3): e0193612, 2018.
Article En | MEDLINE | ID: mdl-29518114

The Drosophila GATA factor gene serpent (srp) is required for the early differentiation of the anterior and posterior midgut primordia. In particular, srp is sufficient and necessary for the primordial gut cells to undertake an epithelial-to-mesenchimal transition (EMT). Two other GATA factor genes, dGATAe and grain (grn), are also specifically expressed in the midgut. On the one hand, dGATAe expression is activated by srp. Embryos homozygous for a deficiency uncovering dGATAe were shown to lack the expression of some differentiated midgut genes. Moreover, ectopic expression of dGATAe was sufficient to drive the expression of some of these differentiation marker genes, thus establishing the role of dGATAe in the regulation of their expression. However, due to the gross abnormalities associated with this deficiency, it was not possible to assess whether, similarly to srp, dGATAe might play a role in setting the midgut morphology. To further investigate this role we decided to generate a dGATAe mutant. On the other hand, grn is expressed in the midgut primordia around stage 11 and remains expressed until the end of embryogenesis. Yet, no midgut function has been described for grn. First, here we report that, as for dGATAe, midgut grn expression is dependent on srp; conversely, dGATAe and grn expression are independent of each other. Our results also indicate that, unlike srp, dGATAe and grn are not responsible for setting the general embryonic midgut morphology. We also show that the analysed midgut genes whose expression is lacking in embryos homozygous for a deficiency uncovering dGATAe are indeed dGATAe-dependent genes. Conversely, we do not find any midgut gene to be grn-dependent, with the exception of midgut repression of the proventriculus iroquois (iro) gene. In conclusion, our results clarify the expression patterns and function of the GATA factor genes expressed in the embryonic midgut.


Drosophila Proteins/metabolism , Drosophila/embryology , GATA Transcription Factors/metabolism , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Digestive System/embryology , Digestive System/metabolism , Digestive System/pathology , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Embryo, Nonmammalian/metabolism , GATA Transcription Factors/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , In Situ Hybridization , Mutation
16.
Pediatr Res ; 83(1-1): 183-189, 2018 01.
Article En | MEDLINE | ID: mdl-28910276

BackgroundInsulin-like growth factor 2 (IGF2) is a key determinant of fetal growth, and the altered expression of IGF2 is implicated in fetal growth disorders and maternal metabolic derangements including gestational diabetes. Here we studied how increased levels of IGF2 in late pregnancy affect fetal growth.MethodsWe employed a rat model of repeated intrafetal IGF2 administration in late pregnancy, i.e., during GD19-GD21, and measured the consequences on fetal organ weight and expression of insulin/IGF-axis components.ResultsIGF2 treatment tended to increase fetal weight, but only weight increase of the fetal stomach reached significance (+33±9%; P<0.01). Sex-dependent data analysis revealed a sexual dimorphism of IGF2 action. In male fetuses, IGF2 administration significantly increased fetal weight (+13±3%; P<0.05) and weight of fetal stomach (+42±10%; P<0.01), intestine (+26±5%; P<0.05), liver (+13±4%; P<0.05), and pancreas (+25±8%; P<0.05). Weights of heart, lungs, and kidneys were unchanged. In female fetuses, IGF2 increased only stomach weight (+26±9%; P<0.05). Furthermore, gene expression of insulin/IGF axis in the heart, lungs, liver, and stomach was more sensitive toward IGF2 treatment in male than in female fetuses.ConclusionData suggest that elevated circulating IGF2 in late pregnancy predominantly stimulates organ growth of the digestive system, and male fetuses are more susceptible toward the IGF2 effects than female fetuses.


Digestive System/embryology , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor II/physiology , Animals , Female , Humans , Insulin/metabolism , Male , Organ Size , Pregnancy , Pregnancy, Animal , Rats , Rats, Wistar , Sex Factors , Tissue Distribution
17.
Early Hum Dev ; 114: 1-6, 2017 11.
Article En | MEDLINE | ID: mdl-28927754

Gastrointestinal motility disorders are common in the pediatric population and may affect the entire gastrointestinal tract and can vary from mild to severe conditions. They may clinically manifest as gastro-esophageal reflux symptoms, feeding difficulties and failure to thrive, constipation and diarrhea amongst others. This review first highlights the embryologic development of the gastrointestinal tract, after which the prenatal and neonatal development of gastrointestinal motility is discussed. Normal motility patterns as seen in (preterm) infants are described as a background for the discussion of the most common congenital and acquired motility disorders in infancy. This review specifically focuses on the role of preterm birth on the development of these disorders.


Gastrointestinal Diseases/etiology , Gastrointestinal Motility , Infant, Newborn, Diseases/etiology , Digestive System/embryology , Gastrointestinal Diseases/physiopathology , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases/physiopathology
18.
Dev Biol ; 431(2): 194-204, 2017 11 15.
Article En | MEDLINE | ID: mdl-28939335

In insects, the hindgut is a homeostatic region of the digestive tract, divided into pylorus, ileum, and rectum, that reabsorbs water, ions, and small molecules produced during hemolymph filtration. The hindgut anatomy in bee larvae is different from that of adult workers. This study reports the morphological changes and cellular events that occur in the hindgut during the metamorphosis of the honeybee Apis mellifera. We describe the occurrence of autophagosomes and the ultrastructure of the epithelial cells and cuticle, suggesting that cuticular degradation begins in prepupae, with the cuticle being reabsorbed and recycled by autophagosomes in white- and pink-eyed pupae, followed by the deposition of new cuticle in light-brown-eyed pupae. In L5S larvae and prepupae, the hindgut undergoes cell proliferation in the anterior and posterior ends. In the pupae, the pylorus, ileum, and rectum regions are differentiated, and cell proliferation ceases in dark-brown-eyed pupae. Apoptosis occurs in the hindgut from the L5S larval to the pink-eyed pupal stage. In light-brown- and dark-brown-eyed pupae, the ileum epithelium changes from pseudostratified to simple only after the production of the basal lamina, whereas the rectal epithelium is always flattened. In black-eyed pupae, ileum epithelial cells have large vacuoles and subcuticular spaces, while in adult forager workers these cells have long invaginations in the cell apex and many mitochondria, indicating a role in the transport of compounds. Our findings show that hindgut morphogenesis is a dynamic process, with tissue remodeling and cellular events taking place for the formation of different regions of the organ, the reconstruction of a new cuticle, and the remodeling of visceral muscles.


Apoptosis , Bees/anatomy & histology , Bees/embryology , Digestive System/cytology , Digestive System/embryology , Hierarchy, Social , Integumentary System/anatomy & histology , Animals , Autophagy , Bees/ultrastructure , Caspase 3/metabolism , Cell Proliferation , Digestive System/ultrastructure , Histones/metabolism , Larva/cytology , Larva/ultrastructure , Pupa/cytology , Pupa/ultrastructure
19.
Nature ; 547(7662): 209-212, 2017 07 13.
Article En | MEDLINE | ID: mdl-28678781

Despite the wide variety of adaptive modifications in the oral and facial regions of vertebrates, their early oropharyngeal development is considered strictly uniform. It involves sequential formation of the mouth and pharyngeal pouches, with ectoderm outlining the outer surface and endoderm the inner surface, as a rule. At the extreme anterior domain of vertebrate embryos, the ectoderm and endoderm directly juxtapose and initial development of this earliest ecto-endoderm interface, the primary mouth, typically involves ectodermal stomodeal invagination that limits the anterior expansion of the foregut endoderm. Here we present evidence that in embryos of extant non-teleost fishes, oral (stomodeal) formation is preceded by the development of prominent pre-oral gut diverticula (POGD) between the forebrain and roof of the forming mouth. Micro-computed tomography (micro-CT) imaging of bichir, sturgeon and gar embryos revealed that foregut outpocketing at the pre-oral domain begins even before the sequential formation of pharyngeal pouches. The presence of foregut-derived cells in the front of the mouth was further confirmed by in vivo experiments that allowed specific tracing of the early endodermal lining. We show that POGD in sturgeons contribute to the orofacial surface of their larvae, comprising oral teeth, lips, and sensory barbels. To our knowledge, this is the first thorough evidence for endodermal origin of external craniofacial structures in any vertebrate. In bichir and gar embryos, POGD form prominent cranial adhesive organs that are characteristic of the ancient bauplan of free-living chordate larvae. POGD hence seem arguably to be ancestral for all ray-finned fishes, and their topology, pharyngeal-like morphogenesis and gene expression suggest that they are evolutionarily related to the foregut-derived diverticula of early chordate and hemichordate embryos. The formation of POGD might thus represent an ancestral developmental module with deep deuterostome origins.


Digestive System/embryology , Endoderm/embryology , Fishes/anatomy & histology , Fishes/embryology , Maxillofacial Development , Mouth/embryology , Animals , Fishes/classification , Fishes/genetics , Gene Expression Regulation, Developmental , Larva/genetics , Larva/growth & development , Maxillofacial Development/genetics , Phylogeny , Skull/embryology , Tooth/embryology , X-Ray Microtomography
20.
Dev Biol ; 427(1): 155-164, 2017 07 01.
Article En | MEDLINE | ID: mdl-28465040

The ancestral state of animal gastrulation and its bearing for our understanding of bilaterian evolution still is one of the most controversially discussed topics in the field of evolutionary and developmental biology. One hypothesis, the so-called amphistomy scenario, suggests the presence of a slit-like blastopore in the last common ancestor of Bilateria. Onychophoran ontogeny at least superficially appears to support this scenario since a ventral groove clearly forms during gastrulation. The origin and nature of this groove, however, is another matter of ongoing controversy; i.e. the question of whether this structure actually represents the blastopore, or at least part of it. Recent research using genetic markers argued against the furrow representing a blastoporal structure. Here we investigate the origin of endoderm, which usually originates from the blastopore. We find conserved expression patterns of the endoderm- and gut-marker genes GATA456, GATA123, Hnf4 and fkh during gut development, and discuss the formation of the onychophoran gut in comparison with that in a range of arthropods. Despite expression of endodermal markers in and around the furrow we do not find convincing evidence that the furrow may be part of the blastopore, and thus we suggest that onychophoran development does not yield support for the amphistomy scenario.


Biomarkers/metabolism , Digestive System/metabolism , Endoderm/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , Invertebrates/metabolism , Animals , Digestive System/embryology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endoderm/embryology , Forkhead Transcription Factors/genetics , GATA Transcription Factors/classification , GATA Transcription Factors/genetics , Gastrula/embryology , Gastrula/metabolism , Hepatocyte Nuclear Factor 4/genetics , In Situ Hybridization , Invertebrates/embryology , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction
...