Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
Elife ; 102021 12 17.
Article in English | MEDLINE | ID: mdl-34919051

ABSTRACT

Metastasis is responsible for approximately 90% of cancer-associated mortality but few models exist that allow for rapid and effective screening of anti-metastasis drugs. Current mouse models of metastasis are too expensive and time consuming to use for rapid and high-throughput screening. Therefore, we created a unique screening concept utilizing conserved mechanisms between zebrafish gastrulation and cancer metastasis for identification of potential anti-metastatic drugs. We hypothesized that small chemicals that interrupt zebrafish gastrulation might also suppress metastatic progression of cancer cells and developed a phenotype-based chemical screen to test the hypothesis. The screen used epiboly, the first morphogenetic movement in gastrulation, as a marker and enabled 100 chemicals to be tested in 5 hr. The screen tested 1280 FDA-approved drugs and identified pizotifen, an antagonist for serotonin receptor 2C (HTR2C) as an epiboly-interrupting drug. Pharmacological and genetic inhibition of HTR2C suppressed metastatic progression in a mouse model. Blocking HTR2C with pizotifen restored epithelial properties to metastatic cells through inhibition of Wnt signaling. In contrast, HTR2C induced epithelial-to-mesenchymal transition through activation of Wnt signaling and promoted metastatic dissemination of human cancer cells in a zebrafish xenotransplantation model. Taken together, our concept offers a novel platform for discovery of anti-metastasis drugs.


Subject(s)
Cell Movement/drug effects , Embryo, Nonmammalian/drug effects , Epithelial-Mesenchymal Transition , Gastrulation/drug effects , High-Throughput Screening Assays/methods , Pizotyline/pharmacology , Receptor, Serotonin, 5-HT2C/genetics , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Drug Discovery , Female , Humans , Mice, Inbred BALB C , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/prevention & control , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Transplantation, Heterologous , Zebrafish , Zebrafish Proteins/genetics
2.
Sci Rep ; 11(1): 19357, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34588480

ABSTRACT

During gastrulation of the zebrafish embryo, the cap of blastoderm cells organizes into the axial body plan of the embryo with left-right symmetry and head-tail, dorsal-ventral polarities. Our labs have been interested in the mechanics of early development and have investigated whether these large-scale cell movements can be described as tissue-level mechanical strain by a tectonics-based approach. The first step is to image the positions of all nuclei from mid-epiboly to early segmentation by digital sheet light microscopy, organize the surface of the embryo into multi-cell spherical domains, construct velocity fields from the movements of these domains and extract strain rate maps from the change in density of the domains. During gastrulation, tensile/expansive and compressive strains in the axial and equatorial directions are detected as anterior and posterior expansion along the anterior-posterior axis and medial-lateral compression across the dorsal-ventral axis and corresponds to the well characterized morphological movements of convergence and extension. Following gastrulation strain is represented by localized medial expansion at the onset of segmentation and anterior expansion at the onset of neurulation. In addition to linear strain, symmetric patterns of rotation/curl are first detected in the animal hemispheres at mid-epiboly and then the vegetal hemispheres by the end of gastrulation. In embryos treated with C59, a Wnt inhibitor that inhibits head and tail extension, the axial extension and vegetal curl are absent. By analysing the temporal sequence of large-scale movements, deformations across the embryo can be attributed to a combination of epiboly and dorsal convergence-extension.


Subject(s)
Body Patterning/physiology , Gastrulation/physiology , Animals , Benzeneacetamides/pharmacology , Body Patterning/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Embryo, Nonmammalian/embryology , Gastrulation/drug effects , Intravital Microscopy , Pyridines/pharmacology , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
3.
Toxicol Appl Pharmacol ; 409: 115277, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33049266

ABSTRACT

The pyridine derivative Y-27632 inhibits Rho-associated coiled-coil-containing protein kinase (ROCK) signaling, which is involved in numerous developmental processes during embryogenesis, primarily by controlling actin-cytoskeleton assembly and cell contractility. Somite formation requires rearrangement of the cytoskeleton and assists in major morphological mechanisms, including ventral body wall formation. Administration of Y-27632 impairs cytoskeletal arrangements in post-gastrulation chick embryos leading to ventral body wall defects (VBWD) at later stages of development. The aim of this study was to investigate the effect of Y-27632 on somite development in post-gastrulation chick embryos during early embryogenesis. After 60 h incubation, embryos in shell-less culture were treated with Y-27632 or vehicle for controls. Following administration, abnormality rates were assessed. In treatment groups, embryos showed a kinked longitudinal body axis. Western blot confirmed impaired ROCK downstream signaling by decreased expression of phosphorylated cofilin-2. Histology, Lysotracker studies and RT-PCR demonstrated increased cell death in somites, the neural tube and the ectoderm. RT-PCR and Western blot of factors known to be involved during somitogenesis revealed reduced expression in the treatment group compared to controls. We hypothesize that administration of Y-27632 disrupts somite development causing axial kinking and embryo malformation, which may lead to VBWD.


Subject(s)
Amides/pharmacology , Embryonic Development/drug effects , Gastrulation/drug effects , Pyridines/pharmacology , Teratogenesis/drug effects , Actin Depolymerizing Factors/metabolism , Animals , Cell Death/drug effects , Chick Embryo , Gene Expression Regulation, Developmental/drug effects , Protein Kinase Inhibitors/pharmacology , Somites/drug effects , Somites/metabolism , rho-Associated Kinases/antagonists & inhibitors
4.
Sci Rep ; 10(1): 15028, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32958954

ABSTRACT

Vitamin E (VitE) deficiency results in embryonic lethality. Knockdown of the gene ttpa encoding for the VitE regulatory protein [α-tocopherol transfer protein (α-TTP)] in zebrafish embryos causes death within 24 h post-fertilization (hpf). To test the hypothesis that VitE, not just α-TTP, is necessary for nervous system development, adult 5D strain zebrafish, fed either VitE sufficient (E+) or deficient (E-) diets, were spawned to obtain E+ and E- embryos, which were subjected to RNA in situ hybridization and RT-qPCR. Ttpa was expressed ubiquitously in embryos up to 12 hpf. Early gastrulation (6 hpf) assessed by goosecoid expression was unaffected by VitE status. By 24 hpf, embryos expressed ttpa in brain ventricle borders, which showed abnormal closure in E- embryos. They also displayed disrupted patterns of paired box 2a (pax2a) and SRY-box transcription factor 10 (sox10) expression in the midbrain-hindbrain boundary, spinal cord and dorsal root ganglia. In E- embryos, the collagen sheath notochord markers (col2a1a and col9a2) appeared bent. Severe developmental errors in E- embryos were characterized by improper nervous system patterning of the usually carefully programmed transcriptional signals. Histological analysis also showed developmental defects in the formation of the fore-, mid- and hindbrain and somites of E- embryos at 24 hpf. Ttpa expression profile was not altered by the VitE status demonstrating that VitE itself, and not ttpa, is required for development of the brain and peripheral nervous system in this vertebrate embryo model.


Subject(s)
Embryo, Nonmammalian/abnormalities , Nervous System/embryology , Vitamin E/physiology , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Brain/embryology , Carrier Proteins/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/innervation , Gastrulation/drug effects , Gastrulation/genetics , Gene Expression Regulation, Developmental/drug effects , PAX2 Transcription Factor/genetics , SOXE Transcription Factors/genetics , Vitamin E/pharmacology , Vitamin E Deficiency/embryology
5.
Sci Rep ; 10(1): 3951, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32127575

ABSTRACT

Ethanol exposure during prenatal development causes fetal alcohol spectrum disorder (FASD), the most frequent preventable birth defect and neurodevelopmental disability syndrome. The molecular targets of ethanol toxicity during development are poorly understood. Developmental stages surrounding gastrulation are very sensitive to ethanol exposure. To understand the effects of ethanol on early transcripts during embryogenesis, we treated zebrafish embryos with ethanol during pre-gastrulation period and examined the transcripts by Affymetrix GeneChip microarray before gastrulation. We identified 521 significantly dysregulated genes, including 61 transcription factors in ethanol-exposed embryos. Sox2, the key regulator of pluripotency and early development was significantly reduced. Functional annotation analysis showed enrichment in transcription regulation, embryonic axes patterning, and signaling pathways, including Wnt, Notch and retinoic acid. We identified all potential genomic targets of 25 dysregulated transcription factors and compared their interactions with the ethanol-dysregulated genes. This analysis predicted that Sox2 targeted a large number of ethanol-dysregulated genes. A gene regulatory network analysis showed that many of the dysregulated genes are targeted by multiple transcription factors. Injection of sox2 mRNA partially rescued ethanol-induced gene expression, epiboly and gastrulation defects. Additional studies of this ethanol dysregulated network may identify therapeutic targets that coordinately regulate early development.


Subject(s)
Ethanol/pharmacology , Gastrulation/genetics , Zebrafish/embryology , Animals , Blastula/cytology , Blastula/drug effects , Blastula/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Female , Gastrulation/drug effects , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Gene Ontology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
6.
PLoS Biol ; 17(10): e3000498, 2019 10.
Article in English | MEDLINE | ID: mdl-31613879

ABSTRACT

During gastrulation, the pluripotent epiblast self-organizes into the 3 germ layers-endoderm, mesoderm and ectoderm, which eventually form the entire embryo. Decades of research in the mouse embryo have revealed that a signaling cascade involving the Bone Morphogenic Protein (BMP), WNT, and NODAL pathways is necessary for gastrulation. In vivo, WNT and NODAL ligands are expressed near the site of gastrulation in the posterior of the embryo, and knockout of these ligands leads to a failure to gastrulate. These data have led to the prevailing view that a signaling gradient in WNT and NODAL underlies patterning during gastrulation; however, the activities of these pathways in space and time have never been directly observed. In this study, we quantify BMP, WNT, and NODAL signaling dynamics in an in vitro model of human gastrulation. Our data suggest that BMP signaling initiates waves of WNT and NODAL signaling activity that move toward the colony center at a constant rate. Using a simple mathematical model, we show that this wave-like behavior is inconsistent with a reaction-diffusion-based Turing system, indicating that there is no stable signaling gradient of WNT/NODAL. Instead, the final signaling state is homogeneous, and spatial differences arise only from boundary effects. We further show that the durations of WNT and NODAL signaling control mesoderm differentiation, while the duration of BMP signaling controls differentiation of CDX2-positive extra-embryonic cells. The identity of these extra-embryonic cells has been controversial, and we use RNA sequencing (RNA-seq) to obtain their transcriptomes and show that they closely resemble human trophoblast cells in vivo. The domain of BMP signaling is identical to the domain of differentiation of these trophoblast-like cells; however, neither WNT nor NODAL forms a spatial pattern that maps directly to the mesodermal region, suggesting that mesoderm differentiation is controlled dynamically by the combinatorial effect of multiple signals. We synthesize our data into a mathematical model that accurately recapitulates signaling dynamics and predicts cell fate patterning upon chemical and physical perturbations. Taken together, our study shows that the dynamics of signaling events in the BMP, WNT, and NODAL cascade in the absence of a stable signaling gradient control fate patterning of human gastruloids.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Gastrulation/genetics , Mesoderm/metabolism , Nodal Protein/genetics , Signal Transduction , Wnt Proteins/genetics , Benzothiazoles/pharmacology , Body Patterning/drug effects , Body Patterning/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Line , Gastrula/cytology , Gastrula/drug effects , Gastrula/metabolism , Gastrulation/drug effects , Gene Expression Regulation , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Mesoderm/cytology , Mesoderm/drug effects , Models, Biological , Models, Statistical , Nodal Protein/deficiency , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Wnt Proteins/metabolism
7.
PLoS Biol ; 17(10): e3000081, 2019 10.
Article in English | MEDLINE | ID: mdl-31634368

ABSTRACT

In vitro models of postimplantation human development are valuable to the fields of regenerative medicine and developmental biology. Here, we report characterization of a robust in vitro platform that enabled high-content screening of multiple human pluripotent stem cell (hPSC) lines for their ability to undergo peri-gastrulation-like fate patterning upon bone morphogenetic protein 4 (BMP4) treatment of geometrically confined colonies and observed significant heterogeneity in their differentiation propensities along a gastrulation associable and neuralization associable axis. This cell line-associated heterogeneity was found to be attributable to endogenous Nodal expression, with up-regulation of Nodal correlated with expression of a gastrulation-associated gene profile, and Nodal down-regulation correlated with a preneurulation-associated gene profile expression. We harness this knowledge to establish a platform of preneurulation-like fate patterning in geometrically confined hPSC colonies in which fates arise because of a BMPs signalling gradient conveying positional information. Our work identifies a Nodal signalling-dependent switch in peri-gastrulation versus preneurulation-associated fate patterning in hPSC cells, provides a technology to robustly assay hPSC differentiation outcomes, and suggests conserved mechanisms of organized fate specification in differentiating epiblast and ectodermal tissues.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Lineage/drug effects , Gene Expression Regulation, Developmental , Nodal Protein/genetics , Pluripotent Stem Cells/drug effects , Biomechanical Phenomena , Body Patterning/genetics , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line , Cell Lineage/genetics , Gastrulation/drug effects , Gastrulation/genetics , Gene Expression Profiling , Genetic Heterogeneity , High-Throughput Screening Assays , Humans , Models, Biological , Neurogenesis/drug effects , Neurogenesis/genetics , Nodal Protein/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Surface Properties
8.
Alcohol Clin Exp Res ; 42(11): 2136-2143, 2018 11.
Article in English | MEDLINE | ID: mdl-30129265

ABSTRACT

BACKGROUND: MNS1 (meiosis-specific nuclear structural protein 1) is necessary for motile cilia function, such as sperm flagella or those found in the embryonic primitive node. While little is known regarding the function or expression pattern of MNS1 in the embryo, co-immunoprecipitation experiments in sperm have determined that MNS1 interacts with ciliary proteins, which are also important during development. Establishment of morphogenic gradients is dependent on normal ciliary motion in the primitive node beginning during gastrulation (gestational day [GD] 7 in the mouse, second-third week of pregnancy in humans), a critical window for face, eye, and brain development and particularly susceptible to perturbations of developmental signals. The current study investigates the role of Mns1 in craniofacial defects associated with gastrulation-stage alcohol exposure. METHODS: On GD7, pregnant Mns1+/- dams were administered 2 doses of ethanol (5.8 g/kg total) or vehicle 4 hours apart to target gastrulation. On GD17, fetuses were examined for ocular defects by scoring each eye on a scale from 1 to 7 (1 = normal, 2 to 7 = defects escalating in severity). Craniofacial and brain abnormalities were also assessed. RESULTS: Prenatal alcohol exposure (PAE) significantly increased the rate of defects in wild-type fetuses, as PAE fetuses had an incidence rate of 41.18% compared to a 10% incidence rate in controls. Furthermore, PAE interacted with genotype to significantly increase the defect rate and severity in Mns1+/- (64.29%) and Mns1-/- mice (92.31%). PAE Mns1-/- fetuses with severe eye defects also presented with craniofacial dysmorphologies characteristic of fetal alcohol syndrome and midline tissue loss in the brain, palate, and nasal septum. CONCLUSIONS: These data demonstrate that a partial or complete knockdown of Mns1 interacts with PAE to increase the susceptibility to ocular defects and correlating craniofacial and brain anomalies, likely though interaction of alcohol with motile cilia function. These results further our understanding of genetic risk factors that may underlie susceptibility to teratogenic exposures.


Subject(s)
Central Nervous System Depressants/toxicity , Craniofacial Abnormalities/chemically induced , Craniofacial Abnormalities/genetics , Ethanol/toxicity , Fetal Alcohol Spectrum Disorders/genetics , Gastrulation/drug effects , Nuclear Proteins/genetics , Animals , Cell Cycle Proteins , Central Nervous System/abnormalities , Central Nervous System/pathology , Craniofacial Abnormalities/epidemiology , Eye Abnormalities/chemically induced , Eye Abnormalities/epidemiology , Eye Abnormalities/pathology , Female , Fetal Alcohol Spectrum Disorders/epidemiology , Fetal Alcohol Spectrum Disorders/pathology , Fetus/pathology , Gene Knockdown Techniques , Incidence , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy
9.
Sci Rep ; 8(1): 10518, 2018 Jul 12.
Article in English | MEDLINE | ID: mdl-30002406

ABSTRACT

Marijuana is one of the most commonly used illicit recreational drugs and is widely used for medicinal purposes. The psychoactive ingredient in marijuana is ∆9-tetrahydrocannabinol (∆9-THC), whereas the major non-psychoactive ingredient is cannabidiol (CBD). Here, we exposed zebrafish embryos to ∆9-THC or CBD for 5 hours during the critical stage of development known as gastrulation. Embryos were allowed to develop normally and were examined at 2 and 5 days post fertilization. THC and CBD treated embryos exhibited reduced heart rates, axial malformations and shorter trunks. Cannabinoid treatment altered synaptic activity at neuromuscular junctions (NMJs), and fluorescent labelling of primary and secondary motor neurons indicated a change in branching patterns and a reduction in the number of axonal branches in the trunk musculature. Furthermore, there were alterations in the α-bungarotoxin labelling of nicotinic acetylcholine receptors at NMJs. Locomotion studies show that larvae exposed to THC or CBD during gastrulation exhibited drastic reductions in the number of C-start escape responses to sound stimuli, but not to touch stimuli. Together these findings indicate that zebrafish embryos exposed to ∆9-THC or CBD during the brief but critical period of gastrulation exhibited alterations in heart rate, motor neuronal morphology, synaptic activity at the NMJ and locomotor responses to sound.


Subject(s)
Cannabis/toxicity , Gastrulation/drug effects , Motor Neurons/drug effects , Neurogenesis/drug effects , Animals , Cannabidiol/toxicity , Cannabis/chemistry , Dronabinol/toxicity , Embryo, Nonmammalian , Female , Heart Rate/drug effects , Locomotion/drug effects , Male , Models, Animal , Time Factors , Toxicity Tests, Acute/methods , Zebrafish
10.
BMC Dev Biol ; 18(1): 6, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29544468

ABSTRACT

BACKGROUND: Signaling cascades, such as the extracellular signal-regulated kinase (ERK) pathway, play vital roles in early vertebrate development. Signals through these pathways are initiated by a growth factor or hormone, are transduced through a kinase cascade, and result in the expression of specific downstream genes that promote cellular proliferation, growth, or differentiation. Tight regulation of these signals is provided by positive or negative modulators at varying levels in the pathway, and is required for proper development and function. Two members of the dual-specificity phosphatase (Dusp) family, dusp6 and dusp2, are believed to be negative regulators of the ERK pathway and are expressed in both embryonic and adult zebrafish, but their specific roles in embryogenesis remain to be fully understood. RESULTS: Using CRISPR/Cas9 genome editing technology, we generated zebrafish lines harboring germ line deletions in dusp6 and dusp2. We do not detect any overt defects in dusp2 mutants, but we find that approximately 50% of offspring from homozygous dusp6 mutants do not proceed through embryonic development. These embryos are fertilized, but are unable to proceed past the first zygotic mitosis and stall at the 1-cell stage for several hours before dying by 10 h post fertilization. We demonstrate that dusp6 is expressed in gonads of both male and female zebrafish, suggesting that loss of dusp6 causes defects in germ cell production. Notably, the 50% of homozygous dusp6 mutants that complete the first cell division appear to progress through embryogenesis normally and give rise to fertile adults. CONCLUSIONS: The fact that offspring of homozygous dusp6 mutants stall prior to activation of the zygotic genome, suggests that loss of dusp6 affects gametogenesis and/or parentally-directed early development. Further, since only approximately 50% of homozygous dusp6 mutants are affected, we postulate that ERK signaling is tightly regulated and that dusp6 is required to keep ERK signaling within a range that is permissive for proper embryogenesis. Lastly, since dusp6 is expressed throughout zebrafish embryogenesis, but dusp6 mutants do not exhibit defects after the first cell division, it is possible that other regulators of the ERK pathway compensate for loss of dusp6 at later stages.


Subject(s)
Dual Specificity Phosphatase 6/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Alleles , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cell Division/drug effects , Dual Specificity Phosphatase 6/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryonic Development/drug effects , Female , Gastrulation/drug effects , Gene Editing , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Germ Cells/drug effects , Germ Cells/metabolism , Homozygote , Male , Morpholinos/pharmacology , Mutation/genetics , Ovary/metabolism , Phenotype , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Testis/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics
11.
J Cell Physiol ; 233(9): 7120-7133, 2018 09.
Article in English | MEDLINE | ID: mdl-29574800

ABSTRACT

An association has been proved between high salt consumption and cardiovascular mortality. In vertebrates, the heart is the first functional organ to be formed. However, it is not clear whether high-salt exposure has an adverse impact on cardiogenesis. Here we report high-salt exposure inhibited basement membrane breakdown by affecting RhoA, thus disturbing the expression of Slug/E-cadherin/N-cadherin/Laminin and interfering with mesoderm formation during the epithelial-mesenchymal transition(EMT). Furthermore, the DiI+ cell migration trajectory in vivo and scratch wound assays in vitro indicated that high-salt exposure restricted cell migration of cardiac progenitors, which was caused by the weaker cytoskeleton structure and unaltered corresponding adhesion junctions at HH7. Besides, down-regulation of GATA4/5/6, Nkx2.5, TBX5, and Mef2c and up-regulation of Wnt3a/ß-catenin caused aberrant cardiomyocyte differentiation at HH7 and HH10. High-salt exposure also inhibited cell proliferation and promoted apoptosis. Most importantly, our study revealed that excessive reactive oxygen species(ROS)generated by high salt disturbed the expression of cardiac-related genes, detrimentally affecting the above process including EMT, cell migration, differentiation, cell proliferation and apoptosis, which is the major cause of malformation of heart tubes.


Subject(s)
Gastrulation/drug effects , Heart Defects, Congenital/embryology , Heart Defects, Congenital/metabolism , Heart/embryology , Sodium Chloride, Dietary/toxicity , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chick Embryo , Chickens , Embryonic Development/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Heart Defects, Congenital/pathology , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats
12.
Sci Rep ; 8(1): 2433, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29402947

ABSTRACT

During the gastrulation stage in animal embryogenesis, the cells leading the axial mesoderm migrate toward the anterior side of the embryo, vigorously extending cell protrusions such as lamellipodia. It is thought that the leading cells sense gradients of chemoattractants emanating from the ectodermal cells and translate them to initiate and maintain the cell movements necessary for gastrulation. However, it is unclear how the extracellular information is converted to the intracellular chemical reactions that lead to motion. Here we demonstrated that intracellular Ca2+ levels in the protrusion-forming leading cells are markedly higher than those of the following cells and the axial mesoderm cells. We also showed that inhibiting the intracellular Ca2+ significantly retarded the gastrulation cell movements, while increasing the intracellular Ca2+ with an ionophore enhanced the migration. We further found that the ionophore treatment increased the active form of the small GTPase Rac1 in these cells. Our results suggest that transient intracellular Ca2+ signals play an essential role in the active cell migration during gastrulation.


Subject(s)
Calcium Signaling , Calcium/metabolism , Gastrulation/physiology , Mesoderm/metabolism , Xenopus laevis/metabolism , Animals , Calcium Ionophores/pharmacology , Cell Movement/drug effects , Chelating Agents/pharmacology , Ectoderm/cytology , Ectoderm/drug effects , Ectoderm/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Embryo, Nonmammalian , Gastrula/cytology , Gastrula/drug effects , Gastrula/metabolism , Gastrulation/drug effects , Gene Expression , Ionomycin/pharmacology , Mesoderm/cytology , Mesoderm/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Xenopus laevis/growth & development , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
13.
Environ Toxicol ; 33(3): 370-380, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29265596

ABSTRACT

Despite the previous reports on melamine contamination in high concentrations some years ago, there were not many studies on low-level exposure in daily life, particularly in pregnancy. We investigated the effect of low-dose melamine on the kidneys of the pregnant rats and their developing embryos/fetuses during various gestational stages namely implantation, gastrulation, organogenesis, maturation and whole pregnancy. Our results showed that the repeated low level of melamine (12.5, 25, and 50 mg/kg bw/d) during pregnancy did not cause obstruction of renal tubules although more precipitating crystals were found in the early gestational periods. Simple hyperplasia in the maternal tubules and pelvic epithelium were more prominent after exposed to melamine during the whole gestational period. Neonatal kidneys significantly suffered more from congestion in glomeruli and interstitium, dilated tubules and interstitial edema after melamine administration to the mother in the late and the whole gestational periods. A trend of advance of glomerular development in fetuses was also observed. We conclude that in utero exposure of low-level melamine could post a risk on the kidneys of the pregnant mother as well as the developing fetuses, which may further increase the possibility of other health problems later in life.


Subject(s)
Kidney/drug effects , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/pathology , Triazines/toxicity , Animals , Embryonic Development , Female , Fetus , Gastrulation/drug effects , Kidney/embryology , Kidney/growth & development , Kidney/pathology , Maternal-Fetal Exchange , Organogenesis/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley
14.
Dev Biol ; 434(2): 249-266, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29287832

ABSTRACT

Control of microtubule dynamics is crucial for cell migration. We analyzed regulation of microtubule network dynamics in the zebrafish yolk cell during epiboly, the earliest coordinated gastrulation movement. We labeled microtubules with EMTB-3GFP and EB3-mCherry to visualize and measure microtubule dynamics by TIRF microscopy live imaging. Yolk cell microtubules dynamics is temporally modulated during epiboly progression. We used maternal zygotic Pou5f3 mutant (MZspg) embryos, which develop strong distortions of microtubule network organization and epiboly retardation, to investigate genetic control of microtubule dynamics. In MZspg embryos, microtubule plus-end growth tracks move slower and are less straight compared to wild-type. MZspg embryos have altered steroidogenic enzyme expression, resulting in increased pregnenolone and reduced progesterone levels. We show that progesterone positively affects microtubule plus-end growth and track straightness. Progesterone may thus act as a non-cell-autonomous regulator of microtubule dynamics across the large yolk cell, and may adjust differing demands on microtubule dynamics and stability during initiation and progression phases of epiboly.


Subject(s)
Gastrula/embryology , Gastrulation/drug effects , Microtubules/metabolism , Progesterone/pharmacology , Zebrafish/embryology , Animals , Gastrulation/physiology , Microtubules/genetics , Zebrafish/genetics
15.
Dev Biol ; 431(2): 215-225, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28923486

ABSTRACT

The lateral line system is a mechanosensory systems present in aquatic animals. The anterior and posterior lateral lines develop from anterior and posterior lateral line placodes (aLLp and pLLp), respectively. Although signaling molecules required for the induction of other cranial placodes have been well studied, the molecular mechanisms underlying formation of the lateral line placodes are unknown. In this study we tested the requirement of multiple signaling pathways, such as Wnt, Bmp Fgf, and Retinoic Acid for aLLp and pLLp induction. We determined that aLLp specification requires Fgf signaling, whilst pLLp specification requires retinoic acid which inhibits Fgf signaling. pLLp induction is also independent of Wnt and Bmp activities, even though these pathways limit the boundaries of the pLLp. This is the first report that the aLLp and pLLp depend on different inductive mechanisms and that pLLp induction requires the inhibition of Fgf, Wnt and Bmp signaling.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Fibroblast Growth Factors/metabolism , Lateral Line System/embryology , Signal Transduction , Tretinoin/pharmacology , Wnt Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Benzaldehydes/pharmacology , Body Patterning/drug effects , Body Patterning/genetics , Gastrulation/drug effects , Lateral Line System/drug effects , Lateral Line System/metabolism , Signal Transduction/drug effects , Zebrafish Proteins/metabolism
16.
Neurotoxicology ; 62: 100-110, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28587808

ABSTRACT

In this study, we examined the effects of alcohol exposure during gastrulation on zebrafish embryos, specifically focusing on excitatory synaptic activity associated with neurons (Mauthner cells) that are born during gastrulation. Furthermore, we determined whether co-treatment of alcohol and retinoic acid (RA) could prevent the effects of alcohol exposure during gastrulation. We exposed zebrafish embryos to ethanol (150mM), RA (1nM), or a combination of RA (1nM) plus ethanol (150mM) for 5.5h from 5.25h post fertilization (hpf) to 10.75 hpf (gastrulation). Ethanol treatment resulted in altered hatching rates, survivability and body lengths. Immunohistochemical analysis of Mauthner cells (M-cells) suggested that ethanol treatment resulted in smaller M-cell bodies and thinner axons, while electrophysiological recordings of AMPA miniature excitatory postsynaptic currents (mEPSCs) associated with M-cells showed that ethanol treated animals had a significantly reduced mEPSC frequency. Other mEPSC parameters such as amplitude, rise times and decay kinetics were not altered by exposure to alcohol. Locomotor studies showed that ethanol treatment resulted in altered C-bend escape responses. For instance, the C-bends of alcohol-treated fish were larger than control embryos. Thus, ethanol treatment during gastrulation altered a range of features in embryonic zebrafish. Importantly, co-treatment with RA prevented all of the effects of ethanol including survivability, body length, M-cell morphology, AMPA mEPSC frequency and escape response movements. Together these findings show that ethanol exposure during the brief period of gastrulation has a significant effect on neuronal morphology and activity, and that this can be prevented with RA co-treatment.


Subject(s)
Brain/cytology , Ethanol/toxicity , Gastrulation/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Tretinoin/pharmacology , Age Factors , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/embryology , Central Nervous System Depressants/toxicity , Edema/chemically induced , Embryo, Nonmammalian , Escape Reaction/drug effects , Excitatory Postsynaptic Potentials/drug effects , Female , Male , Movement/drug effects , Neurotransmitter Agents/pharmacology , Zebrafish , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
17.
Toxicol Sci ; 157(1): 235-245, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28184906

ABSTRACT

Establishment of effective non-animal alternatives for developmental toxicity screening assays is desirable to ensure maternal and fetal health outcomes. Validation of such assays requires a comparison between the in vitro responses to chemical exposures and the in vivo impacts of the corresponding compounds at equivalent concentrations. Here, we investigated how the P19C5 gastrulation model responds to 24 compounds at specific concentrations, some of which are categorized as positive exposures based on previously observed detrimental effects on development in vivo, whereas others are categorized as negative exposures due to lack of effects in vivo. The P19C5 gastrulation model consists of in vitro morphogenesis of mouse stem cells aggregated into embryoid bodies (EBs), which recapitulates growth and axial elongation of early embryos during four days of three-dimensional culture. Adverse impacts of chemical exposures were defined as: death, impaired growth, and altered axial elongation of EBs. Ten out of 17 positive exposures caused adverse impacts on EBs. In contrast, only three out of 17 negative exposures adversely affected EBs, although two of the three diminished viability of somatic cell lines (NIH/3T3, HEK293, and JEG3), suggesting general cytotoxicity. Overall, the study showed that 24 out of 34 exposures impacted EB development in a manner concordant with the in vivo developmental effects. Validation of other alternative assays using the same set of chemical exposures will provide information on the strengths and weaknesses of each assay, and should help determine the most effective ensemble of assays to detect a wide range of developmentally toxic exposures.


Subject(s)
Gastrulation/drug effects , Models, Animal , Teratogens/toxicity , Toxicity Tests , Animals , Embryoid Bodies/drug effects , In Vitro Techniques , Mice
18.
Environ Toxicol ; 32(1): 131-138, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26589250

ABSTRACT

Melamine is a heterocyclic, aromatic amine and nitrogen-enriched environmental toxicant, found in not only adulterated foodstuffs but also industrial household tableware and paints. Previous studies demonstrated adverse effects of high-dose melamine on human infants and pregnant animals, but effects of low-dose melamine on pregnancy have not been reported. In this study, reproductive effects of low-dose melamine were investigated in pregnant rats. Melamine in the range of 12.5-50 mg/kg was administered to pregnant rats at different gestational stages. Maternal weight gain was not significantly affected, and other maternal morbidity was not observed. Low-dose melamine exposure during pregnancy increased fetal size but reduced somite number in gastrulation (GD8.5-GD10.5) and organogenesis (GD10.5-GD16.5) periods, and increased incidence of stillbirth in whole gestational period (GD0.5 to delivery). Embryotoxicity of melamine was further confirmed by whole embryo culture in vitro that melamine retarded embryonic growth, impaired development of brain and heart, and induced open neural tube and atrioventricular defects with increased apoptosis. In conclusion, adverse reproductive effects of low-dose melamine during pregnancy were identified in the developing rat embryos and the perinatal effects of melamine were gestational and developmental stage dependent. Detailed hazard and risk assessment of melamine in reproduction system are warrant. © 2015 Wiley Periodicals, Inc. Environ Toxicol 32: 131-138, 2017.


Subject(s)
Reproduction/drug effects , Triazines/toxicity , Animals , Brain/drug effects , Brain/embryology , Embryo Culture Techniques , Embryonic Development/drug effects , Female , Fetal Development/drug effects , Gastrulation/drug effects , Heart/drug effects , Heart/embryology , Heart Septal Defects/chemically induced , Heart Septal Defects/pathology , Maternal Exposure , Neural Tube Defects/chemically induced , Neural Tube Defects/pathology , Organogenesis/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Stillbirth , Weight Gain/drug effects
19.
Reprod Toxicol ; 66: 68-83, 2016 12.
Article in English | MEDLINE | ID: mdl-27693483

ABSTRACT

Valproic acid (VPA), an antiepileptic drug, is a teratogen that causes neural tube and axial skeletal defects, although the mechanisms are not fully understood. We previously established a gastrulation model using mouse P19C5 stem cell embryoid bodies (EBs), which exhibits axial patterning and elongation morphogenesis in vitro. Here, we investigated the effects of VPA on the EB axial morphogenesis to gain insights into its teratogenic mechanisms. Axial elongation and patterning of EBs were inhibited by VPA at therapeutic concentrations. VPA elevated expression levels of various developmental regulators, including Cdx1 and Hoxa1, known transcriptional targets of retinoic acid (RA) signaling. Co-treatment of EBs with VPA and BMS493, an RA receptor antagonist, partially rescued axial elongation as well as gene expression profiles. These results suggest that VPA requires active RA signaling to interfere with EB morphogenesis.


Subject(s)
Anticonvulsants/toxicity , Embryoid Bodies/drug effects , Gastrulation/drug effects , Teratogens/toxicity , Tretinoin/metabolism , Valproic Acid/toxicity , Animals , Benzoates/pharmacology , Cell Line, Tumor , Embryoid Bodies/metabolism , Gene Expression Regulation, Developmental/drug effects , Histone Deacetylases/metabolism , Mice , Receptors, Retinoic Acid/antagonists & inhibitors , Signal Transduction/drug effects , Stilbenes/pharmacology , Transcriptome/drug effects
20.
BMC Biol ; 14: 61, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27480076

ABSTRACT

BACKGROUND: The nerve net of Nematostella is generated using a conserved cascade of neurogenic transcription factors. For example, NvashA, a homolog of the achaete-scute family of basic helix-loop-helix transcription factors, is necessary and sufficient to specify a subset of embryonic neurons. However, positive regulators required for the expression of neurogenic transcription factors remain poorly understood. RESULTS: We show that treatment with the MEK/MAPK inhibitor U0126 severely reduces the expression of known neurogenic genes, Nvath-like, NvsoxB(2), and NvashA, and known markers of differentiated neurons, suggesting that MAPK signaling is necessary for neural development. Interestingly, ectopic NvashA fails to rescue the expression of neural markers in U0126-treated animals. Double fluorescence in situ hybridization and transgenic analysis confirmed that NvashA targets represent both unique and overlapping populations of neurons. Finally, we used a genome-wide microarray to identify additional patterning genes downstream of MAPK that might contribute to neurogenesis. We identified 18 likely neural transcription factors, and surprisingly identified ~40 signaling genes and transcription factors that are expressed in either the aboral domain or animal pole that gives rise to the endomesoderm at late blastula stages. CONCLUSIONS: Together, our data suggest that MAPK is a key early regulator of neurogenesis, and that it is likely required at multiple steps. Initially, MAPK promotes neurogenesis by positively regulating expression of NvsoxB(2), Nvath-like, and NvashA. However, we also found that MAPK is necessary for the activity of the neurogenic transcription factor NvashA. Our forward molecular approach provided insight about the mechanisms of embryonic neurogenesis. For instance, NvashA suppression of Nvath-like suggests that inhibition of progenitor identity is an active process in newly born neurons, and we show that downstream targets of NvashA reflect multiple neural subtypes rather than a uniform neural fate. Lastly, analysis of the MAPK targets in the early embryo suggests that MAPK signaling is critical not only to neurogenesis, but also endomesoderm formation and aboral patterning.


Subject(s)
Cnidaria/enzymology , MAP Kinase Signaling System , Neurogenesis , Animals , Butadienes/pharmacology , Cnidaria/drug effects , Cnidaria/embryology , Down-Regulation/drug effects , Down-Regulation/genetics , Ectoderm/drug effects , Ectoderm/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gastrulation/drug effects , Gene Expression Regulation, Developmental/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Time Factors , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...