Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 301
Filter
1.
PLoS Genet ; 19(11): e1011030, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37948459

ABSTRACT

Hirschsprung disease (HSCR) is associated with deficiency of the receptor tyrosine kinase RET, resulting in loss of cells of the enteric nervous system (ENS) during fetal gut development. The major contribution to HSCR risk is from common sequence variants in RET enhancers with additional risk from rare coding variants in many genes. Here, we demonstrate that these RET enhancer variants specifically alter the human fetal gut development program through significant decreases in gene expression of RET, members of the RET-EDNRB gene regulatory network (GRN), other HSCR genes, with an altered transcriptome of 2,382 differentially expressed genes across diverse neuronal and mesenchymal functions. A parsimonious hypothesis for these results is that beyond RET's direct effect on its GRN, it also has a major role in enteric neural crest-derived cell (ENCDC) precursor proliferation, its deficiency reducing ENCDCs with relative expansion of non-ENCDC cells. Thus, genes reducing RET proliferative activity can potentially cause HSCR. One such class is the 23 RET-dependent transcription factors enriched in early gut development. We show that their knockdown in human neuroblastoma SK-N-SH cells reduces RET and/or EDNRB gene expression, expanding the RET-EDNRB GRN. The human embryos we studied had major remodeling of the gut transcriptome but were unlikely to have had HSCR: thus, genetic or epigenetic changes in addition to those in RET are required for aganglionosis.


Subject(s)
Enhancer Elements, Genetic , Gastrointestinal Tract , Proto-Oncogene Proteins c-ret , Haplotypes , Humans , Proto-Oncogene Proteins c-ret/genetics , Neuroblastoma , Cell Line, Tumor , Hirschsprung Disease/genetics , Fetus , Gastrointestinal Tract/embryology , Neural Crest/cytology , Enteric Nervous System/embryology , Single-Cell Gene Expression Analysis , Gene Expression Regulation, Developmental
2.
Lancet Gastroenterol Hepatol ; 7(1): 96-106, 2022 01.
Article in English | MEDLINE | ID: mdl-34822760

ABSTRACT

Over the past 5 years, systematic investigation of the mesenteric organ has expanded and shown that the mesentery is the organ in and on which all abdominal digestive organs develop and remain connected to. In turn, this observation has clarified the anatomical foundation of the abdomen and the fundamental order at that level. Findings related to the shape and development of the mesentery have illuminated its function, advancing our understanding of the pathobiology, diagnosis, and treatment of several abdominal and systemic diseases. Inclusion of the mesentery in surgical resections alters the course of benign and malignant diseases. Mesenteric-based scoring systems can enhance the radiological interpretation of abdominal disease. Emerging findings reconcile observations across scientific and clinical fields and have been assimilated into reference curricula and practice guidelines. This Review summarises the developmental, anatomical, and clinical advances made since the mesentery was redesignated as an organ in 2016.


Subject(s)
Gastrointestinal Diseases/therapy , Gastrointestinal Tract/embryology , Mesentery/anatomy & histology , Mesentery/physiology , Gastrointestinal Diseases/diagnostic imaging , Gastrointestinal Diseases/etiology , Humans , Lymphatic Metastasis , Mesentery/pathology
3.
Elife ; 102021 09 07.
Article in English | MEDLINE | ID: mdl-34490846

ABSTRACT

The spatial organization of gut microbiota influences both microbial abundances and host-microbe interactions, but the underlying rules relating bacterial dynamics to large-scale structure remain unclear. To this end, we studied experimentally and theoretically the formation of three-dimensional bacterial clusters, a key parameter controlling susceptibility to intestinal transport and access to the epithelium. Inspired by models of structure formation in soft materials, we sought to understand how the distribution of gut bacterial cluster sizes emerges from bacterial-scale kinetics. Analyzing imaging-derived data on cluster sizes for eight different bacterial strains in the larval zebrafish gut, we find a common family of size distributions that decay approximately as power laws with exponents close to -2, becoming shallower for large clusters in a strain-dependent manner. We show that this type of distribution arises naturally from a Yule-Simons-type process in which bacteria grow within clusters and can escape from them, coupled to an aggregation process that tends to condense the system toward a single massive cluster, reminiscent of gel formation. Together, these results point to the existence of general, biophysical principles governing the spatial organization of the gut microbiome that may be useful for inferring fast-timescale dynamics that are experimentally inaccessible.


The human gut is home to vast numbers of bacteria that grow, compete and cooperate in a dynamic, densely packed space. The spatial arrangement of organisms ­ for example, if they are clumped together or broadly dispersed ­ plays a major role in all ecosystems; but how bacteria are organized in the human gut remains mysterious and difficult to investigate. Zebrafish larvae provide a powerful tool for studying microbes in the gut, as they are optically transparent and anatomically similar to other vertebrates, including humans. Furthermore, zebrafish can be easily manipulated so that one species of bacteria can be studied at a time. To investigate whether individual bacterial species are arranged in similar ways, Scholmann and Parthasarathy exposed zebrafish with no gut bacteria to one of eight different strains. Each species was then monitored using three-dimensional microscopy to see how the population shaped itself into clusters (or colonies). Schlomann and Parthasarathy used this data to build a mathematical model that can predict the size of the clusters formed by different gut bacteria. This revealed that the spatial arrangement of each species depended on the same biological processes: bacterial growth, aggregation and fragmentation of clusters, and expulsion from the gut. These new details about how bacteria are organized in zebrafish may help scientists learn more about gut health in humans. Although it is not possible to peer into the human gut and watch how bacteria behave, scientists could use the same analysis method to study the size of bacterial colonies in fecal samples. This may provide further clues about how microbes are spatially arranged in the human gut and the biological processes underlying this formation.


Subject(s)
Bacteria/growth & development , Bacterial Physiological Phenomena , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Animals , Gastrointestinal Tract/embryology , Gels , Kinetics , Models, Theoretical , Population Density , Zebrafish/embryology , Zebrafish/microbiology
4.
Biomed Pharmacother ; 138: 111521, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34311525

ABSTRACT

Euphorbiae pekinensis Radix (EP) is effective in treating various diseases, but it's toxicity is a major obstacle in use in clinical. Although EP was processed with vinegar to reduce it's toxicity, the detailed mechanism of toxicity in EP have not been clearly delineated. This study investigate the toxicity attenuation-mechanism of Euphorbiae pekinensis after being processed with vinegar (VEP) and the toxic mechanism of four compounds from EP on zebrafish embryos. The contents of four compounds decreased obviously in VEP. Correspondingly, slower development on embryos can be seen as some symptoms like reduction of heart rate, liver area and gastrointestinal peristalsis after exposed to the compounds. Some obvious pathological signals such as pericardial edema and yolk sac edema were observed. Furthermore, the compounds could increase the contents of MDA and GSH-PX and induce oxidative damage by inhibiting the activity of SOD. Also, four compounds could provoke apoptosis by up-regulating the expression level of p53, MDM2, Bax, Bcl-2 and activating the activity of caspase-3, caspase-9. In conclusion, the four compounds play an important role in the toxicity attenuation effects of VEP, which may be related to the apoptosis induction and oxidative damage. This would contribute to the clinical application and further toxicity-reduction mechanism research.


Subject(s)
Euphorbia/toxicity , Gastrointestinal Tract/drug effects , Heart/drug effects , Liver/drug effects , Phytochemicals/toxicity , Plant Extracts/toxicity , Zebrafish/embryology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cardiotoxicity , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Euphorbia/chemistry , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Heart/embryology , Liver/embryology , Liver/metabolism , Oxidative Stress/drug effects , Phytochemicals/isolation & purification , Plant Extracts/isolation & purification , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Commun Biol ; 4(1): 770, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162999

ABSTRACT

While the colonization of the embryonic gut by neural crest cells has been the subject of intense scrutiny over the past decades, we are only starting to grasp the morphogenetic transformations of the enteric nervous system happening in the fetal stage. Here, we show that enteric neural crest cell transit during fetal development from an isotropic cell network to a square grid comprised of circumferentially-oriented cell bodies and longitudinally-extending interganglionic fibers. We present ex-vivo dynamic time-lapse imaging of this isotropic-to-nematic phase transition and show that it occurs concomitantly with circular smooth muscle differentiation in all regions of the gastrointestinal tract. Using conditional mutant embryos with enteric neural crest cells depleted of ß1-integrins, we show that cell-extracellular matrix anchorage is necessary for ganglia to properly reorient. We demonstrate by whole mount second harmonic generation imaging that fibrous, circularly-spun collagen I fibers are in direct contact with neural crest cells during the orientation transition, providing an ideal orientation template. We conclude that smooth-muscle associated extracellular matrix drives a critical reorientation transition of the enteric nervous system in the mammalian fetus.


Subject(s)
Gastrointestinal Tract/embryology , Neural Crest/cytology , Animals , Cell Adhesion , Cell Differentiation , Extracellular Matrix/physiology , Gastrointestinal Tract/innervation , Integrin beta1/physiology , Mice , Muscle, Smooth/embryology
6.
Cell ; 184(13): 3394-3409.e20, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34077752

ABSTRACT

The human fetal immune system begins to develop early during gestation; however, factors responsible for fetal immune-priming remain elusive. We explored potential exposure to microbial agents in utero and their contribution toward activation of memory T cells in fetal tissues. We profiled microbes across fetal organs using 16S rRNA gene sequencing and detected low but consistent microbial signal in fetal gut, skin, placenta, and lungs in the 2nd trimester of gestation. We identified several live bacterial strains including Staphylococcus and Lactobacillus in fetal tissues, which induced in vitro activation of memory T cells in fetal mesenteric lymph node, supporting the role of microbial exposure in fetal immune-priming. Finally, using SEM and RNA-ISH, we visualized discrete localization of bacteria-like structures and eubacterial-RNA within 14th weeks fetal gut lumen. These findings indicate selective presence of live microbes in fetal organs during the 2nd trimester of gestation and have broader implications toward the establishment of immune competency and priming before birth.


Subject(s)
Bacteria/metabolism , Embryonic Development , Fetus/cytology , Fetus/microbiology , Leukocytes/cytology , Adult , Bacteria/genetics , Bacteria/ultrastructure , Cell Proliferation , Dendritic Cells/metabolism , Female , Fetus/ultrastructure , Gastrointestinal Tract/embryology , Gastrointestinal Tract/ultrastructure , Humans , Immunologic Memory , Lymphocyte Activation/immunology , Microbial Viability , Pregnancy , Pregnancy Trimester, Second , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Reproducibility of Results , T-Lymphocytes/cytology
7.
Hereditas ; 157(1): 46, 2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33213520

ABSTRACT

BACKGROUND: The Drosophila hindgut is commonly used model for studying various aspects of organogenesis like primordium establishment, further specification, patterning, and morphogenesis. During embryonic development of Drosophila, many transcriptional activators are involved in the formation of the hindgut. The transcription factor Orthopedia (Otp), a member of the 57B homeobox gene cluster, is expressed in the hindgut and nervous system of developing Drosophila embryos, but due to the lack of mutants no functional analysis has been conducted yet. RESULTS: We show that two different otp transcripts, a hindgut-specific and a nervous system-specific form, are present in the Drosophila embryo. Using an Otp antibody, a detailed expression analysis during hindgut development was carried out. Otp was not only expressed in the embryonic hindgut, but also in the larval and adult hindgut. To analyse the function of otp, we generated the mutant otp allele otpGT by ends-out gene targeting. In addition, we isolated two EMS-induced otp alleles in a genetic screen for mutants of the 57B region. All three otp alleles showed embryonic lethality with a severe hindgut phenotype. Anal pads were reduced and the large intestine was completely missing. This phenotype is due to apoptosis in the hindgut primordium and the developing hindgut. CONCLUSION: Our data suggest that Otp is another important factor for hindgut development of Drosophila. As a downstream factor of byn Otp is most likely present only in differentiated hindgut cells during all stages of development rather than in stem cells.


Subject(s)
Drosophila Proteins/genetics , Drosophila/embryology , Drosophila/genetics , Gastrointestinal Tract/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Organogenesis/genetics , Alleles , Animals , Apoptosis , Embryonic Development , Fluorescent Antibody Technique , Genetic Loci , Homeodomain Proteins/metabolism , In Situ Hybridization , Larva , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
8.
Am J Physiol Gastrointest Liver Physiol ; 319(4): G519-G528, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32877218

ABSTRACT

The first contractile waves in the developing embryonic gut are purely myogenic; they only involve smooth muscle. Here, we provide evidence for a transition from smooth muscle to interstitial cell of Cajal (ICC)-driven contractile waves in the developing chicken gut. In situ hybridization staining for anoctamin-1 (ANO1), a known ICC marker, shows that ICCs are already present throughout the gut, as from embryonic day (E)7. We devised a protocol to reveal ICC oscillatory and propagative calcium activity in embryonic gut whole mount and found that the first steady calcium oscillations in ICCs occur on (E14). We show that the activation of ICCs leads to an increase in contractile wave frequency, regularity, directionality, and velocity between E12 and E14. We finally demonstrate that application of the c-KIT antagonist imatinib mesylate in organ culture specifically depletes the ICC network and inhibits the transition to a regular rhythmic wave pattern. We compare our findings to existing results in the mouse and predict that a similar transition should take place in the human fetus between 12 and 14 wk of development. Together, our results point to an abrupt physiological transition from smooth muscle mesenchyme self-initiating waves to ICC-driven motility in the fetus and clarify the contribution of ICCs to the contractile wave pattern.NEW & NOTEWORTHY We reveal a sharp transition from smooth muscle to interstitial cell of Cajal (ICC)-driven motility in the chicken embryo, leading to higher-frequency, more rhythmic contractile waves. We predict the transition to happen between 12 and 14 embryonic wk in humans. We image for the first time the onset of ICC activity in an embryonic gut by calcium imaging. We show the first KIT and anoctamin-1 (ANO1) in situ hybridization micrographs in the embryonic chicken gut.


Subject(s)
Gastrointestinal Motility/physiology , Interstitial Cells of Cajal/physiology , Intestines/embryology , Animals , Anoctamin-1/analysis , Calcium/metabolism , Chick Embryo , Gastrointestinal Tract/embryology , Gastrointestinal Tract/physiology , Humans , Interstitial Cells of Cajal/chemistry , Intestines/physiology , Mice , Muscle Contraction/physiology , Muscle, Smooth/embryology , Muscle, Smooth/physiology , Time Factors
9.
J Neurosci ; 40(35): 6691-6708, 2020 08 26.
Article in English | MEDLINE | ID: mdl-32690615

ABSTRACT

Precise extrinsic afferent (visceral sensory) and efferent (sympathetic and parasympathetic) innervation of the gut is fundamental for gut-brain cross talk. Owing to the limitation of intrinsic markers to distinctively visualize the three classes of extrinsic axons, which intimately associate within the gut mesentery, detailed information on the development of extrinsic gut-innervating axons remains relatively sparse. Here, we mapped extrinsic innervation of the gut and explored the relationships among various types of extrinsic axons during embryonic development in mice. Visualization with characterized intrinsic markers revealed that visceral sensory, sympathetic, and parasympathetic axons arise from different anatomic locations, project in close association via the gut mesentery, and form distinctive innervation patterns within the gut from embryonic day (E)10.5 to E16.5. Genetic ablation of visceral sensory trajectories results in the erratic extension of both sympathetic and parasympathetic axons, implicating that afferent axons provide an axonal scaffold to route efferent axons. Coculture assay further confirmed the attractive effect of sensory axons on sympathetic axons. Taken together, our study provides key information regarding the development of extrinsic gut-innervating axons occurring through heterotypic axonal interactions and provides an anatomic basis to uncover neural circuit assembly in the gut-brain axis (GBA).SIGNIFICANCE STATEMENT Understanding the development of extrinsic innervation of the gut is essential to unravel the bidirectional neural communication between the brain and the gut. Here, with characterized intrinsic markers targeting vagal sensory, spinal sensory, sympathetic, and parasympathetic axons, respectively, we comprehensively traced the spatiotemporal development of extrinsic axons to the gut during embryonic development in mice. Moreover, in line with the somatic nervous system, pretarget sorting via heterotypic axonal interactions is revealed to play critical roles in patterning extrinsic efferent trajectories to the gut. These findings provide basic anatomic information to explore the mechanisms underlying the process of assembling neural circuitry in the gut-brain axis (GBA).


Subject(s)
Autonomic Nervous System/embryology , Gastrointestinal Tract/innervation , Animals , Autonomic Nervous System/physiology , Axons/physiology , Brain/embryology , Brain/physiology , Gastrointestinal Tract/embryology , Mesentery/embryology , Mesentery/innervation , Mice , Morphogenesis , Neuroanatomical Tract-Tracing Techniques
10.
Toxins (Basel) ; 12(6)2020 06 08.
Article in English | MEDLINE | ID: mdl-32521650

ABSTRACT

Harmful algal blooms (HAB) have become a major health concern worldwide, not just to humans that consume and recreate on contaminated waters, but also to the fauna that inhabit the environments surrounding affected areas. HABs contain heterotrophic bacteria, cyanobacterial lipopolysaccharide, and cyanobacterial toxins such as microcystins, that can cause severe toxicity in many aquatic species as well as bioaccumulation within various organs. Thus, the possibility of trophic transference of this toxin through the food chain has potentially important health implications for other organisms in the related food web. While some species have developed adaptions to attenuate the toxic effects of HAB toxins, there are still numerous species that remain vulnerable, including Lithobates catesbeiana (American bullfrog) tadpoles. In the current study we demonstrate that acute, short-term exposure of tadpoles to HAB toxins containing 1 µg/L (1 nmol/L) of total microcystins for only 7 days results in significant liver and intestinal toxicity within tadpoles. Exposed tadpoles had increased intestinal diameter, decreased intestinal fold heights, and a constant number of intestinal folds, indicating pathological intestinal distension, similar to what is seen in various disease processes, such as toxic megacolon. HAB-toxin-exposed tadpoles also demonstrated hepatocyte hypertrophy with increased hepatocyte binucleation consistent with carcinogenic and oxidative processes within the liver. Both livers and intestines of HAB-toxin-exposed tadpoles demonstrated significant increases in protein carbonylation consistent with oxidative stress and damage. These findings demonstrate that short-term exposure to HAB toxins, including microcystins, can have significant adverse effects in amphibian populations. This acute, short-term toxicity highlights the need to evaluate the influence HAB toxins may have on other vulnerable species within the food web and how those may ultimately also impact human health.


Subject(s)
Gastrointestinal Tract/drug effects , Harmful Algal Bloom , Liver/drug effects , Microcystins/toxicity , Rana catesbeiana , Water Microbiology , Animals , Food Chain , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Larva/drug effects , Liver/embryology , Liver/metabolism , Protein Carbonylation/drug effects , Rana catesbeiana/embryology , Time Factors , Toxicity Tests, Acute
11.
BMC Biol ; 18(1): 68, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546156

ABSTRACT

BACKGROUND: The homeobox genes Pdx and Cdx are widespread across the animal kingdom and part of the small ParaHox gene cluster. Gene expression patterns suggest ancient roles for Pdx and Cdx in patterning the through-gut of bilaterian animals although functional data are available for few lineages. To examine evolutionary conservation of Pdx and Cdx gene functions, we focus on amphioxus, small marine animals that occupy a pivotal position in chordate evolution and in which ParaHox gene clustering was first reported. RESULTS: Using transcription activator-like effector nucleases (TALENs), we engineer frameshift mutations in the Pdx and Cdx genes of the amphioxus Branchiostoma floridae and establish mutant lines. Homozygous Pdx mutants have a defect in amphioxus endoderm, manifest as loss of a midgut region expressing endogenous GFP. The anus fails to open in homozygous Cdx mutants, which also have defects in posterior body extension and epidermal tail fin development. Treatment with an inverse agonist of retinoic acid (RA) signalling partially rescues the axial and tail fin phenotypes indicating they are caused by increased RA signalling. Gene expression analyses and luciferase assays suggest that posterior RA levels are kept low in wild type animals by a likely direct transcriptional regulation of a Cyp26 gene by Cdx. Transcriptome analysis reveals extensive gene expression changes in mutants, with a disproportionate effect of Pdx and Cdx on gut-enriched genes and a colinear-like effect of Cdx on Hox genes. CONCLUSIONS: These data reveal that amphioxus Pdx and Cdx have roles in specifying middle and posterior cell fates in the endoderm of the gut, roles that likely date to the origin of Bilateria. This conclusion is consistent with these two ParaHox genes playing a role in the origin of the bilaterian through-gut with a distinct anus, morphological innovations that contributed to ecological change in the Cambrian. In addition, we find that amphioxus Cdx promotes body axis extension through a molecular mechanism conserved with vertebrates. The axial extension role for Cdx dates back at least to the origin of Chordata and may have facilitated the evolution of the post-anal tail and active locomotion in chordates.


Subject(s)
Anal Canal/embryology , Gastrointestinal Tract/embryology , Homeodomain Proteins/genetics , Lancelets/embryology , Mutation , Tail/embryology , Transcription Factors/genetics , Animals , Embryo, Nonmammalian , Embryonic Development/genetics , Genes, Homeobox , Homeodomain Proteins/metabolism , Lancelets/genetics , Transcription Factors/metabolism
13.
Zygote ; 28(3): 208-216, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32077403

ABSTRACT

In the present study, the morphological development of the Brycon amazonicus digestive tract is described to provide basic knowledge for nutritional studies and, therefore, increase the survival of this species during larviculture. Samples were collected from hatching up to 25 days of age, measured, processed and observed under a stereomicroscope and light microscopy. Newly hatched larvae presented their digestive tract as a straight tube, dorsal to the yolk sac, lined with a single layer of undifferentiated cells. At 24 h post-hatching (hPH), the buccopharyngeal cavity was open, but the posterior region of the digestive tube remained closed. At 25 hPH, the digestive tube was completely open and could be divided into buccopharyngeal cavity, oesophagus and intestine. At 35 hPH, the intestine presented a dilatation in the proximal region, which had the function of storing food. Differentiation of the stomach started at 83 hPH, and mucous cells were observed in the epithelium. These cells are important in the production of mucus, whose function is to protect the organ against acidity, although the gastric glands began developing only from 171 hPH, when three stomach regions were observed: cardiac, fundic and pyloric. The gastric glands were observed in the cardiac region, indicating that this organ already had digestive functionality. From 243 hPH, the absorption and assimilation of nutrients were already possible but, only from 412 hPH, the digestive tract was completely developed and functional.


Subject(s)
Characiformes/growth & development , Gastrointestinal Tract/growth & development , Animals , Branchial Region/cytology , Branchial Region/embryology , Branchial Region/growth & development , Characiformes/anatomy & histology , Characiformes/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryonic Development/physiology , Gastric Mucosa/cytology , Gastric Mucosa/embryology , Gastric Mucosa/growth & development , Gastrointestinal Tract/cytology , Gastrointestinal Tract/embryology , Larva/cytology , Larva/growth & development , Mouth Mucosa/cytology , Mouth Mucosa/embryology , Mouth Mucosa/growth & development , Time Factors
14.
Gene Expr Patterns ; 35: 119097, 2020 01.
Article in English | MEDLINE | ID: mdl-32007595

ABSTRACT

The poly(C)-binding protein (PCBP) family members belong to a subtype of RNA-binding proteins that are ubiquitously expressed with diverse functions. In mammals, PCBP family, also known as hnRNP E family, is composed of four proteins, namely PCBP1, PCBP2, PCBP3 and PCBP4. So far, no study has been documented on the physiological roles of each member in vertebrate development. Here we analysed the spatiotemporal expression patterns of zebrafish (Danio rerio) pcbp2 (identical to pcbp1 and pcbp2 in mammals), pcbp3 and pcbp4 at various stages of zebrafish embryonic development by whole-mount in situ hybridization. Our results revealed that all pcbp genes are maternally expressed, especially pcbp2, which is strongly expressed from the embryogenetic stage to larva. The expression patterns of PCBP members are similar to each other at the very early developmental stage sharing with common strong expression in the intestine, otic vesicle, retina and brain of zebrafish. Subsequently, the messenger RNAs of PCBP members are gradually constrained and highly expressed in intestines of the larvae. Collectively, our study figured out the expression pattern of each PCBP member in diverse organogenesis during embryo development, indicating that PCBP members may play predominant roles in the development of neural and digestive systems to maintain their normal physiological functions. Moreover, the similar expression patterns at the developmental stages and organ types among this family suggest that the aberrant expression of these genes would lead to the neural or intestinal diseases.


Subject(s)
Gene Expression Regulation, Developmental , Organogenesis , RNA-Binding Proteins/genetics , Zebrafish Proteins/genetics , Animals , Brain/embryology , Brain/metabolism , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , RNA-Binding Proteins/metabolism , Retina/embryology , Retina/metabolism , Zebrafish , Zebrafish Proteins/metabolism
15.
Biol Open ; 9(2)2020 02 07.
Article in English | MEDLINE | ID: mdl-31988094

ABSTRACT

In the anterior foregut (AFG) of mouse embryos, the transcription factor SOX2 is expressed in the epithelia of the esophagus and proximal branches of respiratory organs comprising the trachea and bronchi, whereas NKX2.1 is expressed only in the epithelia of respiratory organs. Previous studies using hypomorphic Sox2 alleles have indicated that reduced SOX2 expression causes the esophageal epithelium to display some respiratory organ characteristics. In the present study, we produced mouse embryos with AFG-specific SOX2 deficiency. In the absence of SOX2 expression, a single NKX2.1-expressing epithelial tube connected the pharynx and the stomach, and a pair of bronchi developed in the middle of the tube. Expression patterns of NKX2.1 and SOX9 revealed that the anterior and posterior halves of SOX2-deficient AFG epithelial tubes assumed the characteristics of the trachea and bronchus, respectively. In addition, we found that mesenchymal tissues surrounding the SOX2-deficient NKX2.1-expressing epithelial tube changed to those surrounding the trachea and bronchi in the anterior and posterior halves, as indicated by the arrangement of smooth muscle cells and SOX9-expressing cells and by the expression of Wnt4 (esophagus specific), Tbx4 (respiratory organ specific), and Hoxb6 (distal bronchus specific). The impact of mesenchyme-derived signaling on the early stage of AFG epithelial specification has been indicated. Our study demonstrated an opposite trend where epithelial tissue specification causes concordant changes in mesenchymal tissues, indicating a reciprocity of epithelial-mesenchymal interactions.


Subject(s)
Esophagus/abnormalities , Gastrointestinal Tract/abnormalities , Organogenesis/genetics , SOXB1 Transcription Factors/deficiency , Trachea/abnormalities , Animals , Cell Differentiation/genetics , Endoderm/abnormalities , Endoderm/embryology , Epithelium/embryology , Esophagus/embryology , Fluorescent Antibody Technique , Gastrointestinal Tract/embryology , Gene Expression Regulation, Developmental , Mesoderm/embryology , Mice , Mice, Transgenic , Trachea/embryology
16.
Pediatr Res ; 87(5): 847-852, 2020 04.
Article in English | MEDLINE | ID: mdl-31756731

ABSTRACT

BACKGROUND: Fetal swallowing of human amniotic fluid (hAF) containing trophic factors (TFs) promotes gastrointestinal tract (GIT) development. Preterm birth interrupts hAF swallowing, which may increase the risk of necrotizing enterocolitis (NEC). Postnatally, it is difficult to replicate fetal swallowing of hAF due to volume. We aimed to evaluate whether hAF lyophilization is feasible and its effect on hAF-borne TFs. METHODS: We collected hAF (n = 16) from uncomplicated pregnancies. hAF was divided into three groups: unprocessed control (C), concentration by microfiltration (F), and by dialysis and lyophilization (L). EGF, HGF, GM-CSF, and TGF-α were measured in each group by multiplex assay. Bioavailability of TFs was measured by proliferation and LPS-induced IL-8 production by intestinal epithelial cells FHs74. RESULTS: After dialysis/lyophilization, GM-CSF and TGF-α were preserved with partial loss of EGF and HGF. hAF increased cell proliferation and reduced LPS-induced IL-8 production compared to medium alone. Compared to control, dialysis/lyophilization and filtration of hAF increased FHs74 cell proliferation (p < 0.001) and decreased LPS-induced IL-8 production (p < 0.01). CONCLUSIONS: Lyophilization and filtration of hAF is feasible with partial loss of TFs but maintains and even improves bioavailability of TFs measured by proliferation and LPS-induced IL-8 production by FHs74.


Subject(s)
Amniotic Fluid/metabolism , Enterocolitis, Necrotizing/metabolism , Freeze Drying , Gastrointestinal Tract/embryology , Amniotic Fluid/chemistry , Cell Proliferation , Cryopreservation , Deglutition , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation , Interleukin-8/metabolism , Pregnancy , Transforming Growth Factor alpha/metabolism
17.
BMC Dev Biol ; 19(1): 25, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31884948

ABSTRACT

BACKGROUND: Rab proteins are GTPases responsible for intracellular vesicular trafficking regulation. Rab11 proteins, members of the Rab GTPase family, are known to regulate vesicular recycling during embryonic development. In zebrafish, there are 3 rab11 paralogues, known as rab11a, rab11ba and rab11bb, sharing high identity with each other. However, the expression analysis of rab11 is so far lacking. RESULTS: Here, by phylogeny analysis, we found the three rab11 genes are highly conserved especially for their GTPase domains. We examined the expression patterns of rab11a, rab11ba and rab11bb using RT-PCR and in situ hybridization. We found that all the three genes were highly enriched in the central nervous system, but in different areas of the brain. Apart from brain, rab11a was also expressed in caudal vein, pronephric duct, proctodeum, pharyngeal arches and digestive duct, rab11ba was detected to express in muscle, and rab11bb was expressed in kidney, fin and spinal cord. Different from rab11a and rab11ba, which both have maternal expressions in embryos, rab11bb only expresses during 24hpf to 96hpf. CONCLUSIONS: Our results suggest that rab11 genes play important but distinct roles in the development of the nervous system in zebrafish. The findings could provide new evidences for better understanding the functions of rab11 in the development of zebrafish embryos.


Subject(s)
Zebrafish/embryology , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , Animals , Central Nervous System/embryology , Central Nervous System/metabolism , Conserved Sequence , Female , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Developmental , Maternal Inheritance , Multigene Family , Protein Domains , Tissue Distribution , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rab GTP-Binding Proteins/chemistry
18.
Autophagy ; 15(11): 2019-2021, 2019 11.
Article in English | MEDLINE | ID: mdl-31470757

ABSTRACT

Macroautophagy/autophagy has been demonstrated to play an essential role in embryonic development. However, the role of autophagy during human fetal digestive tract development has not been investigated. Here, by using over 5,000 human embryonic digestive tract cells ranging from 6 weeks to 25 weeks, we explored the dynamic expression of autophagy-related genes at single-cell resolution, and found that the transcriptional activity of autophagy-related genes boosted remarkably and specifically in the early (between 6 and 9 weeks) stages. Interestingly, the small intestine cells at 9 weeks showed the most significant enrichment of autophagy-related genes than any other stages. In summary, our results for the first time revealed that autophagy may play an essential role in the development of the digestive tract, especially for the small intestine, in early human embryos. Abbreviations: GI: gastrointestinal; S-Intes: small intestine; t-SNE: t-distributed stochastic neighbor embedding.


Subject(s)
Autophagy-Related Proteins/genetics , Autophagy/genetics , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Autophagy-Related Proteins/metabolism , Databases, Genetic , Fetus , Genomics , Humans , Sequence Analysis, RNA , Single-Cell Analysis
19.
Anat Sci Int ; 94(2): 192-198, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30600445

ABSTRACT

In this study we examined the ontogenic development of the digestive tract of Vimba bream (Vimba vimba, Family: Cyprinidae) during the first 60 days of life (hatching to 60 days after hatching [DAH]). Samples of developing Vimba bream were randomly selected at various stages of development: 1-8, 10, 15, 20, 25, 30, 40, 50, and 60 DAH. For the histological and histochemical studies on the development of the alimentary canal, tissue sections prepared from the sampled hatchlings were stained with hematoxylin-eosin and periodic acid-Schiff and observed under a light microscope. The histological structures of both the mouth and esophagus were fully developed at 5 and 7 DAH, respectively. Intestinal differentiation was observed at 2 DAH, while mucosal folds appeared on the intestinal bulb at 7 DAH. At 5 DAH, with the appearance of goblet cells in the epithelium of the mouth, pharynx, and esophagus, the larvae showed secretion activity in these organs. At 6 DAH, secretion was observed in the intestine; at this stage of development, the surface of the gastrointestinal tract was covered in a neutral mucous-like layer of polysaccharide. The histological observations indicate that the early development of the digestive tract in Vimba vimba enables larvae to efficiently ingest and digest exogenous feed very quickly after hatching.


Subject(s)
Fishes/anatomy & histology , Fishes/embryology , Gastrointestinal Tract/anatomy & histology , Gastrointestinal Tract/embryology , Larva/anatomy & histology , Larva/growth & development , Animals , Digestion , Eating , Fishes/growth & development , Fishes/physiology , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/physiology , Larva/physiology , Polysaccharides/metabolism
20.
Nature ; 565(7740): 480-484, 2019 01.
Article in English | MEDLINE | ID: mdl-30651642

ABSTRACT

The embryonic gut tube is a cylindrical structure from which the respiratory and gastrointestinal tracts develop1. Although the early emergence of the endoderm as an epithelial sheet2,3 and later morphogenesis of the definitive digestive and respiratory organs4-6 have been investigated, the intervening process of gut tube formation remains relatively understudied7,8. Here we investigate the molecular control of macroscopic forces underlying early morphogenesis of the gut tube in the chick embryo. The gut tube has been described as forming from two endodermal invaginations-the anterior intestinal portal (AIP) towards the rostral end of the embryo and the caudal intestinal portal (CIP) at the caudal end-that migrate towards one another, internalizing the endoderm until they meet at the yolk stalk (umbilicus in mammals)1,6. Migration of the AIP to form foregut has been descriptively characterized8,9, but the hindgut is likely to form by a distinct mechanism that has not been fully explained10. We find that the hindgut is formed by collective cell movements through a stationary CIP, rather than by movement of the CIP itself. Further, combining in vivo imaging, biophysics and mathematical modelling with molecular and embryological approaches, we identify a contractile force gradient that drives cell movements in the hindgut-forming endoderm, enabling tissue-scale posterior extension of the forming hindgut tube. The force gradient, in turn, is established in response to a morphogenic gradient of fibroblast growth factor signalling. As a result, we propose that an important positive feedback arises, whereby contracting cells draw passive cells from low to high fibroblast growth factor levels, recruiting them to contract and pull more cells into the elongating hindgut. In addition to providing insight into the early gut development, these findings illustrate how large-scale tissue level forces can be traced to developmental signals during vertebrate morphogenesis.


Subject(s)
Gastrointestinal Tract/embryology , Morphogenesis , Animals , Body Patterning , Cell Movement , Chick Embryo , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Fibroblast Growth Factor 8/metabolism , Gastrointestinal Tract/cytology , Gastrointestinal Tract/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...