Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(49): eadh8152, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38055823

ABSTRACT

During vertebrate gastrulation, an embryo transforms from a layer of epithelial cells into a multilayered gastrula. This process requires the coordinated movements of hundreds to tens of thousands of cells, depending on the organism. In the chick embryo, patterns of actomyosin cables spanning several cells drive coordinated tissue flows. Here, we derive a minimal theoretical framework that couples actomyosin activity to global tissue flows. Our model predicts the onset and development of gastrulation flows in normal and experimentally perturbed chick embryos, mimicking different gastrulation modes as an active stress instability. Varying initial conditions and a parameter associated with active cell ingression, our model recapitulates distinct vertebrate gastrulation morphologies, consistent with recently published experiments in the chick embryo. Altogether, our results show how changes in the patterning of critical cell behaviors associated with different force-generating mechanisms contribute to distinct vertebrate gastrulation modes via a self-organizing mechanochemical process.


Subject(s)
Actomyosin , Gastrulation , Animals , Chick Embryo , Gastrula , Vertebrates
2.
Elife ; 122023 04 11.
Article in English | MEDLINE | ID: mdl-37039463

ABSTRACT

Convergence-extension in embryos is controlled by chemical and mechanical signalling. A key cellular process is the exchange of neighbours via T1 transitions. We propose and analyse a model with positive feedback between recruitment of myosin motors and mechanical tension in cell junctions. The model produces active T1 events, which act to elongate the tissue perpendicular to the main direction of tissue stress. Using an idealised tissue patch comprising several active cells embedded in a matrix of passive hexagonal cells, we identified an optimal range of mechanical stresses to trigger an active T1 event. We show that directed stresses also generate tension chains in a realistic patch made entirely of active cells of random shapes and leads to convergence-extension over a range of parameters. Our findings show that active intercalations can generate stress that activates T1 events in neighbouring cells, resulting in tension-dependent tissue reorganisation, in qualitative agreement with experiments on gastrulation in chick embryos.


Subject(s)
Gastrulation , Mechanotransduction, Cellular , Animals , Chick Embryo , Feedback , Gastrulation/physiology , Morphogenesis , Intercellular Junctions
3.
Sci Adv ; 9(1): eabn5429, 2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36598979

ABSTRACT

The morphology of gastrulation driving the internalization of the mesoderm and endoderm differs markedly among vertebrate species. It ranges from involution of epithelial sheets of cells through a circular blastopore in amphibians to ingression of mesenchymal cells through a primitive streak in amniotes. By targeting signaling pathways controlling critical cell behaviors in the chick embryo, we generated crescent- and ring-shaped mesendoderm territories in which cells can or cannot ingress. These alterations subvert the formation of the chick primitive streak into the gastrulation modes seen in amphibians, reptiles, and teleost fish. Our experimental manipulations are supported by a theoretical framework linking cellular behaviors to self-organized multicellular flows outlined in detail in the accompanying paper. Together, this suggests that the evolution of gastrulation movements is largely determined by changes in a few critical cell behaviors in the mesendoderm territory across different species and controlled by a relatively small number of signaling pathways.

4.
Proc Natl Acad Sci U S A ; 117(21): 11444-11449, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32381735

ABSTRACT

Morphogenetic flows in developmental biology are characterized by the coordinated motion of thousands of cells that organize into tissues, naturally raising the question of how this collective organization arises. Using only the kinematics of tissue deformation, which naturally integrates local and global mechanisms along cell paths, we identify the dynamic morphoskeletons behind morphogenesis, i.e., the evolving centerpieces of multicellular trajectory patterns. These features are model- and parameter-free, frame-invariant, and robust to measurement errors and can be computed from unfiltered cell-velocity data. We reveal the spatial attractors and repellers of the embryo by quantifying its Lagrangian deformation, information that is inaccessible to simple trajectory inspection or Eulerian methods that are local and typically frame-dependent. Computing these dynamic morphoskeletons in wild-type and mutant chick and fly embryos, we find that they capture the early footprint of known morphogenetic features, reveal new ones, and quantitatively distinguish between different phenotypes.


Subject(s)
Chick Embryo/cytology , Chick Embryo/growth & development , Drosophila melanogaster/embryology , Models, Biological , Animals , Animals, Genetically Modified , Biomechanical Phenomena , Chick Embryo/drug effects , Computer Simulation , Drosophila Proteins/genetics , Embryo, Nonmammalian/cytology , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/metabolism , Gastrula/growth & development , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Indazoles/pharmacology , Microscopy/methods , Morphogenesis , Mutation , Twist-Related Protein 1/genetics
5.
Ecotoxicol Environ Saf ; 194: 110415, 2020 May.
Article in English | MEDLINE | ID: mdl-32151871

ABSTRACT

Zinc oxide Nanoparticles (ZnO NPs) are widely used as emerging materials in agricultural and food-related fields, which exists potential safety hazards to public health and environment while bringing an added level of convenience to our original life. It has been proved that ZnO NPs could be taken up by pregnant women and passed through human placental barrier. However, the toxic potential for embryo development remains largely unanswered. In this study, we discovered that ZnO NPs caused the cytotoxicity in vitro. Inhibition of free Zn2+ ions in solution by EDTA or inhibition of Zn2+ ions absorption by CaCl2 could partially eliminate ZnO NPs-mediated cell toxicity, though not redeem completely. This indicated that both nanoparticles and the release of Zn2+ ions were involved in ZnO NPs-mediated cytotoxicity. In addition, we also found that both nanoparticles and Zn2+ ion release triggered reactive oxygen species (ROS) production, which further induced cell toxicity, inflammation and apoptosis, which are mediated by NF-κB signaling cascades and the mitochondria dysfunction, respectively. Eventually, these events lead to the suppressed production and migration of cranial neural crest cells (CNCCs), which subsequently prompts the craniofacial defects in chicken embryos. The application of the antioxidant N-Acetyl-L-cysteine (NAC) rescued the ZnO NPs-induced cell toxicity and malformation of the CNCCs, which further verified our hypothesis. Our results revealed the relevant mechanism of ZnO NPs exposure-inhibited the development of CNCCs, which absolutely contribute to assess the risk of nanoparticles application.


Subject(s)
Embryonic Development/drug effects , Nanoparticles/toxicity , Neural Crest/drug effects , Oxidative Stress/drug effects , Zinc Oxide/toxicity , Acetylcysteine/pharmacology , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cell Survival/drug effects , Chick Embryo , Female , HEK293 Cells , Humans , Mitochondria/drug effects , NF-kappa B/metabolism , Nanoparticles/chemistry , Neural Crest/embryology , Reactive Oxygen Species/metabolism , Zinc Oxide/chemistry
6.
Development ; 147(3)2020 02 04.
Article in English | MEDLINE | ID: mdl-31964776

ABSTRACT

Directional cell intercalations of epithelial cells during gastrulation has, in several organisms, been shown to be associated with a planar cell polarity in the organisation of the actin-myosin cytoskeleton and is postulated to reflect directional tension that drives oriented cell intercalations. We have characterised and applied a recently introduced non-destructive optical manipulation technique to measure the tension in individual epithelial cell junctions of cells in various locations and orientations in the epiblast of chick embryos in the early stages of primitive streak formation. Junctional tension of mesendoderm precursors in the epiblast is higher in junctions oriented in the direction of intercalation than in junctions oriented perpendicular to the direction of intercalation and higher than in junctions of other cells in the epiblast. The kinetic data fit best with a simple viscoelastic Maxwell model, and we find that junctional tension, and to a lesser extent viscoelastic relaxation time, are dependent on myosin activity.


Subject(s)
Epithelial Cells/metabolism , Gastrulation/physiology , Intercellular Junctions/metabolism , Optical Tweezers , Primitive Streak/growth & development , Animals , Animals, Genetically Modified , Cell Movement/physiology , Cell Polarity/physiology , Chick Embryo , Gastrula/metabolism , Germ Layers/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hydrocarbons, Chlorinated/pharmacology , Microscopy, Fluorescence/methods , Myosin Type I/antagonists & inhibitors , Myosin Type I/metabolism , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Pyrroles/pharmacology , Signal Transduction/physiology
7.
Exp Clin Endocrinol Diabetes ; 127(9): 590-597, 2019 Oct.
Article in English | MEDLINE | ID: mdl-28950393

ABSTRACT

The incidence of gestational diabetes mellitus (GDM) has increased dramatically amongst multiethnic population. However, how gestational diabetes mellitus damages the developing embryo is still unknown. In this study, we used yolk sac membrane (YSM) model to investigate angiogenesis in the developing chick embryo. We determined that in the presence of high glucose, it retarded the growth and extension of the embryonic vascular plexus and it also reduced the density of the vasculature in yolk sac membrane model. Using the same strategy, we used the chorioallantoic membrane (CAM) as a model to investigate the influence of high glucose on the vasculature. We established that high glucose inhibited development of the blood vessel plexus and the blood vessels formed had a narrower diameter than control vessels. Concurrent with the abnormal angiogenesis, we also examined how it impacted cardiogenesis. We determined the myocardium in the right ventricle and left atrium were significantly thicker than the control and also there was a reduction in glycogen content in cardiomyocytes. The high glucose also induced excess reactive oxygen species (ROS) production in the cardiomyocytes. We postulated that it was the excess reactive oxygen species that damaged the cardiomyocytes resulting in cardiac hyperplasia.


Subject(s)
Chorioallantoic Membrane , Embryonic Development/drug effects , Glucose/pharmacology , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Yolk Sac , Animals , Chick Embryo , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , Glucose/metabolism , Hyperplasia/chemically induced , Hyperplasia/embryology , Hyperplasia/pathology , Myocytes, Cardiac/pathology , Yolk Sac/metabolism , Yolk Sac/pathology
8.
Zygote ; 26(6): 457-464, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30520400

ABSTRACT

SummaryFibroblast growth factor (FGF) signalling acts as one of modulators that control neural crest cell (NCC) migration, but how this is achieved is still unclear. In this study, we investigated the effects of FGF signalling on NCC migration by blocking this process. Constructs that were capable of inducing Sprouty2 (Spry2) or dominant-negative FGFR1 (Dn-FGFR1) expression were transfected into the cells making up the neural tubes. Our results revealed that blocking FGF signalling at stage HH10 (neurulation stage) could enhance NCC migration at both the cranial and trunk levels in the developing embryos. It was established that FGF-mediated NCC migration was not due to altering the expression of N-cadherin in the neural tube. Instead, we determined that cyclin D1 was overexpressed in the cranial and trunk levels when Sprouty2 was upregulated in the dorsal neural tube. These results imply that the cell cycle was a target of FGF signalling through which it regulates NCC migration at the neurulation stage.


Subject(s)
Chick Embryo/cytology , Chick Embryo/metabolism , Fibroblast Growth Factors/metabolism , Neural Crest/cytology , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Movement , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/metabolism , Neural Tube/embryology , Neural Tube/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
9.
Cell Physiol Biochem ; 48(5): 2084-2090, 2018.
Article in English | MEDLINE | ID: mdl-30099448

ABSTRACT

BACKGROUND/AIMS: Angiogenesis plays a key role during embryonic development. The vascular endothelin (ET) system is involved in the regulation of angiogenesis. Lipopolysaccharides (LPS) could induce angiogenesis. The effects of ET blockers on baseline and LPS-stimulated angiogenesis during embryonic development remain unknown so far. METHODS: The blood vessel density (BVD) of chorioallantoic membranes (CAMs), which were treated with saline (control), LPS, and/or BQ123 and the ETB blocker BQ788, were quantified and analyzed using an IPP 6.0 image analysis program. Moreover, the expressions of ET-1, ET-2, ET3, ET receptor A (ETRA), ET receptor B (ETRB) and VEGFR2 mRNA during embryogenesis were analyzed by semi-quantitative RT-PCR. RESULTS: All components of the ET system are detectable during chicken embryogenesis. LPS increased angiogenesis substantially. This process was completely blocked by the treatment of a combination of the ETA receptor blockers-BQ123 and the ETB receptor blocker BQ788. This effect was accompanied by a decrease in ETRA, ETRB, and VEGFR2 gene expression. However, the baseline angiogenesis was not affected by combined ETA/ETB receptor blockade. CONCLUSION: During chicken embryogenesis, the LPS-stimulated angiogenesis, but not baseline angiogenesis, is sensitive to combined ETA/ETB receptor blockade.


Subject(s)
Endothelin B Receptor Antagonists/pharmacology , Lipopolysaccharides/pharmacology , Neovascularization, Physiologic/drug effects , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Animals , Chickens , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Embryonic Development/drug effects , Endothelin-1/genetics , Endothelin-1/metabolism , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Piperidines/pharmacology , Receptor, Endothelin A/chemistry , Receptor, Endothelin A/genetics , Receptor, Endothelin B/chemistry , Receptor, Endothelin B/genetics , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
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
11.
Cell Death Dis ; 9(2): 234, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445081

ABSTRACT

In this study, the effects of Baicalin on the hyperglycemia-induced cardiovascular malformation during embryo development were investigated. Using early chick embryos, an optimal concentration of Baicalin (6 µM) was identified which could prevent hyperglycemia-induced cardiovascular malformation of embryos. Hyperglycemia-enhanced cell apoptosis was reduced in embryos and HUVECs in the presence of Baicalin. Hyperglycemia-induced excessive ROS production was inhibited when Baicalin was administered. Analyses of SOD, GSH-Px, MQAE and GABAA suggested Baicalin plays an antioxidant role in chick embryos possibly through suppression of outwardly rectifying Cl(-) in the high-glucose microenvironment. In addition, hyperglycemia-enhanced autophagy fell in the presence of Baicalin, through affecting the ubiquitin of p62 and accelerating autophagy flux. Both Baicalin and Vitamin C could decrease apoptosis, but CQ did not, suggesting autophagy to be a protective function on the cell survival. In mice, Baicalin reduced the elevated blood glucose level caused by streptozotocin (STZ). Taken together, these data suggest that hyperglycemia-induced embryonic cardiovascular malformation can be attenuated by Baicalin administration through suppressing the excessive production of ROS and autophagy. Baicalin could be a potential candidate drug for women suffering from gestational diabetes mellitus.


Subject(s)
Autophagy/drug effects , Cardiovascular System/drug effects , Diabetes Mellitus, Experimental/drug therapy , Flavonoids/pharmacology , Hypoglycemic Agents/pharmacology , Organogenesis/drug effects , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Autophagy/genetics , Blood Glucose/metabolism , Cardiovascular System/growth & development , Cardiovascular System/metabolism , Cardiovascular System/pathology , Chick Embryo , Chloride Channels/genetics , Chloride Channels/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Embryo, Nonmammalian , Female , Gene Expression Regulation , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Organogenesis/genetics , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Streptozocin , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
12.
J Cell Physiol ; 233(12): 9271-9283, 2018 12.
Article in English | MEDLINE | ID: mdl-28885685

ABSTRACT

Cytotoxicity and inflammation-associated toxic responses could be induced by bacterial lipopolysaccharides (LPS) in vitro and in vivo, respectively. However, the mechanism involved in LPS-induced cardiac malformation in prenatal fetus is still unknown. In this study, we demonstrated that LPS was induced in gut microbiota imbalance mice, and next, LPS exposure during gastrulation in the chick embryo increased the incidence of cardia bifida. Gene transfection and tissue transplantation trajectory indicated that LPS exposure restricted the cell migration of cardiac progenitors to primary heart field in gastrula chick embryos. In vitro explant allograft of GFP-labeled anterior primitive streak demonstrated that LPS treatments could inhibit cell migration. A similar observation was also obtained from the cell migration assay of scratch wounds using primary culture of cardiomyocytes or H9c2 cells. In the embryos exposed to LPS, expressions of Nkx2.5 and GATA5 were disturbed. These genes are associated with cardiomyocyte differentiation when heart tube fusion occurs. Furthermore, pHIS3, C-caspase3 immunohistological staining indicated that cell proliferation decreased, cell apoptosis increased in the heart tube of chick embryo. Meanwhile, in vivo, pHIS3 immunohistological staining and Hochest/PI staining also draw the similar conclusions. The LPS exposure also caused the production of excess ROS, which might damage the cardiac precursor cells of developing embryos. At last, we showed that LPS-induced cardia bifida could be partially rescued through the addition of antioxidants. Together, these results reveal that excess ROS generation is involved in the LPS-induced defects in heart tube during chick embryo development.


Subject(s)
Endotoxins/toxicity , Gastrointestinal Microbiome/drug effects , Heart Defects, Congenital/embryology , Heart/embryology , Organogenesis/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Chick Embryo , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Heart Defects, Congenital/genetics , Humans , Lipopolysaccharides/toxicity , Male , Mice , Models, Biological , Organogenesis/genetics , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Reproduction/drug effects
13.
Mol Neurobiol ; 55(4): 3523-3536, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28509082

ABSTRACT

Autophagy plays a very important role in numerous physiological and pathological events. However, it still remains unclear whether Atg7-induced autophagy is involved in the regulation of neural crest cell production. In this study, we found the co-location of Atg7 and Pax7+ neural crest cells in early chick embryo development. Upregulation of Atg7 with unilateral transfection of full-length Atg7 increased Pax7+ and HNK-1+ cephalic and trunk neural crest cell numbers compared to either Control-GFP transfection or opposite neural tubes, suggesting that Atg7 over-expression in neural tubes could enhance the production of neural crest cells. BMP4 in situ hybridization and p-Smad1/5/8 immunofluorescent staining demonstrated that upregulation of Atg7 in neural tubes suppressed the BMP4/Smad signaling, which is considered to promote the delamination of neural crest cells. Interestingly, upregulation of Atg7 in neural tubes could significantly accelerate cell progression into the S phase, implying that Atg7 modulates cell cycle progression. However, ß-catenin expression was not significantly altered. Finally, we demonstrated that upregulation of the Atg7 gene could activate autophagy as did Atg8. We have also observed that similar phenotypes, such as more HNK-1+ neural crest cells in the unilateral Atg8 transfection side of neural tubes, and the transfection with full-length Atg8-GFP certainly promote the numbers of BrdU+ neural crest cells in comparison to the GFP control. Taken together, we reveal that Atg7-induced autophagy is involved in regulating the production of neural crest cells in early chick embryos through the modification of the cell cycle.


Subject(s)
Autophagy-Related Protein 7/metabolism , Autophagy , Neural Crest/cytology , Neurogenesis , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Protein 8 Family/metabolism , Bone Morphogenetic Protein 4/metabolism , Cell Cycle , Cell Line, Tumor , Chick Embryo , Gene Expression Regulation, Developmental , Models, Biological , Neural Crest/metabolism , Neural Crest/ultrastructure , Neural Tube/cytology , Neural Tube/embryology , Neural Tube/metabolism , Neural Tube/ultrastructure , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Signal Transduction , Smad Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
14.
Exp Cell Res ; 361(1): 73-84, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28987541

ABSTRACT

Slit/Robo signaling plays an important role in the guidance of developing neurons in developing embryos. However, it remains obscure whether and how Slit/Robo signaling is involved in the production of cranial neural crest cells. In this study, we examined Robo1 deficient mice to reveal developmental defects of mouse cranial frontal and parietal bones, which are derivatives of cranial neural crest cells. Therefore, we determined the production of HNK1+ cranial neural crest cells in early chick embryo development after knock-down (KD) of Robo1 expression. Detection of markers for pre-migratory and migratory neural crest cells, PAX7 and AP-2α, showed that production of both was affected by Robo1 KD. In addition, we found that the transcription factor slug is responsible for the aberrant delamination/EMT of cranial neural crest cells induced by Robo1 KD, which also led to elevated expression of E- and N-Cadherin. N-Cadherin expression was enhanced when blocking FGF signaling with dominant-negative FGFR1 in half of the neural tube. Taken together, we show that Slit/Robo signaling influences the delamination/EMT of cranial neural crest cells, which is required for cranial bone development.


Subject(s)
Craniofacial Abnormalities/pathology , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/physiology , Neural Crest/cytology , Receptors, Immunologic/physiology , Animals , Cell Differentiation , Cells, Cultured , Chick Embryo , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Female , Male , Mice , Mice, Knockout , Neural Crest/metabolism , Neurogenesis , Roundabout Proteins
15.
Toxicol Lett ; 281: 53-64, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-28919490

ABSTRACT

Excess alcohol consumption during pregnancy could lead to fetal alcohol syndrome (FAS). However, the molecular mechanism leading to craniofacial abnormality, a feature of FAS, is still poorly understood. The cranial neural crest cells (NCCs) contribute to the formation of the craniofacial bones. Therefore, NCCs exposed to ethanol was investigated - using chick embryos and in vitro explant culture as experimental models. We demonstrated that exposure to 2% ethanol induced craniofacial defects, which includes parietal defect, in the developing chick fetus. Immunofluorescent staining revealed that ethanol treatment downregulated Ap-2ɑ, Pax7 and HNK-1 expressions by cranial NCCs. Using double-immunofluorescent stainings for Ap-2ɑ/pHIS3 and Ap-2ɑ/c-Caspase3, we showed that ethanol treatment inhibited cranial NCC proliferation and increased NCC apoptosis, respectively. Moreover, ethanol treatment of the dorsal neuroepithelium increased Laminin, N-Cadherin and Cadherin 6B expressions while Cadherin 7 expression was repressed. In situ hybridization also revealed that ethanol treatment up-regulated Cadherin 6B expression but down-regulated slug, Msx1, FoxD3 and BMP4 expressions. In summary, our experimental results demonstrated that ethanol treatment interferes with the production of cranial NCCs by affecting the proliferation and apoptosis of these cells. In addition, ethanol affected the delamination, epithelial-mesenchymal transition (EMT) and cell migration of cranial NCCs, which may have contributed to the etiology of the craniofacial defects.


Subject(s)
Craniofacial Abnormalities/pathology , Ethanol/toxicity , Gene Expression Regulation, Developmental , Neural Crest/drug effects , Organogenesis/drug effects , Animals , Apoptosis/drug effects , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , CD57 Antigens/genetics , CD57 Antigens/metabolism , Cadherins/genetics , Cadherins/metabolism , Chick Embryo , Craniofacial Abnormalities/chemically induced , Disease Models, Animal , Down-Regulation , Fetal Alcohol Spectrum Disorders/physiopathology , Laminin/genetics , Laminin/metabolism , Neural Crest/pathology , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism
16.
Reprod Toxicol ; 73: 96-104, 2017 10.
Article in English | MEDLINE | ID: mdl-28789864

ABSTRACT

Ethanol's effect on embryonic vasculogenesis and its underlying mechanism is obscure. Using VE-cadherin in situ hybridization, we found blood islands formation was inhibited in area opaca, but abnormal VE-cadherin+ cells were seen in area pellucida. We hypothesise ethanol may affect blood island progenitor cell migration and differentiation. DiI and in vitro experiments revealed ethanol inhibited cell migration, Quantitative PCR analysis revealed that ethanol exposure enhanced cell differentiation in area pellucida of HH5 chick embryos and repressed cell differentiation in area pellucida of HH8 chick embryos. By exposing to 2,2'-azobis-amidinopropane dihydrochloride, a ROS inducer, which gave a similar anti-vasculogenesis effect as ethanol and this anti-vasculogenesis effect could be reversed by vitamin C. Overall, exposing early chick embryos to ethanol represses blood island progenitor cell migration but disturbed differentiation at a different stage, so that the disorder of blood island formation occurs through excess ROS production and altered vascular-associated gene expression.


Subject(s)
Chick Embryo/drug effects , Ethanol/toxicity , Hemangioblasts/drug effects , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Chick Embryo/embryology , Embryonic Development/drug effects , Hemangioblasts/physiology , Reactive Oxygen Species/metabolism
17.
Toxicol Sci ; 158(1): 36-47, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28453788

ABSTRACT

Dexamethasone (Dex) is commonly used in the treatment of a variety of benign and malignant conditions. Unfortunately, although it has a variety of teratogenic effects, it remains used in clinical practice for pregnant women mainly due to limited alternatives. However, there is limited knowledge of the mechanisms that lead to the observed teratogenic effects. In this study, the effects of Dex during embryogenesis on neural crest development were evaluated in the early chick embryos. First, we demonstrated that 100 µl 10-6 M Dex treatment leads to craniofacial developmental defects, and also retards embryo growth and plausibly can cause embryo demise. Second, we demonstrated that Dex represses the production of HNK-1, PAX7, and AP-2α labeled cranial neural crest cells, the progenitor cells of the craniofacial skeleton. Third, double immunofluorescent staining of pHIS3/PAX7 and AP-2α/c-Caspase3 revealed that Dex promotes cell apoptosis but does not change cell proliferation rates. Last, fibroblast growth factor signaling molecules were inhibited by Dex treatment. Dex also inhibited neural crest cells production by repressing Msx1 expression in the developing neural tube and by altering expression of epithelial-mesenchymal transition-related adhesion molecules and cell migration genes. Overall, we obtained experimental evidence that Dex treatment during embryogenesis disrupts cranial neural crest development which in turn causes defective cranial bone development.


Subject(s)
Dexamethasone/adverse effects , Neural Crest/pathology , Animals , Chick Embryo , In Situ Hybridization , Risk Factors
18.
Cell Death Dis ; 8(3): e2697, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28333135

ABSTRACT

The BRE (brain and reproductive expression) gene, highly expressed in nervous and reproductive system organs, plays an important role in modulating DNA damage repair under stress response and pathological conditions. Folliculogenesis, a process that ovarian follicle develops into maturation, is closely associated with the interaction between somatic granulosa cell and oocyte. However, the regulatory role of BRE in follicular development remains undetermined. In this context, we found that BRE is normally expressed in the oocytes and granulosa cells from the primordial follicle stage. There was a reduction in follicles number of BRE mutant (BRE-/-) mice. It was attributed to increase the follicular atresia in ovaries, as a result of retarded follicular development. We established that cell proliferation was inhibited, while apoptosis was markedly increased in the granulosa cells in the absence of BRE. In addition, expressions of γ-H2AX (marker for showing DNA double-strand breaks) and DNA damage-relevant genes are both upregulated in BRE-/- mice. In sum, these results suggest that the absence of BRE, deficiency in DNA damage repair, causes increased apoptosis in granulosa cells, which in turn induces follicular atresia in BRE-/- mice.


Subject(s)
Cell Death/physiology , Follicular Atresia/metabolism , Granulosa Cells/metabolism , Granulosa Cells/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Ovarian Follicle/metabolism , Ovary/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , DNA Damage/physiology , DNA Repair/physiology , Female , Follicular Atresia/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/metabolism , Ovarian Follicle/physiology
19.
J Agric Food Chem ; 64(47): 9078-9088, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27792329

ABSTRACT

As a neonicotinoid pesticide, imidacloprid is widely used to control sucking insects on agricultural planting and fleas on domestic animals. However, the extent to which imidacloprid exposure has an influence on cardiogensis in early embryogenesis is still poorly understood. In vertebrates, the heart is the first organ to be formed. In this study, to address whether imidacloprid exposure affects early heart development, the early chick embryo has been used as an experimental model because of its accessibility at its early developmental stage. The results demonstrate that exposure of the early chick embryo to imidacloprid caused malformation of heart tube. Furthermore, the data reveal that down-regulation of GATA4, NKX2.5, and BMP4 and up-regulation of Wnt3a led to aberrant cardiomyocyte differentiation. In addition, imidacloprid exposure interfered with basement membrane breakdown, E-cadherin/laminin expression, and mesoderm formation during the epithelial-mesenchymal transition (EMT) in gastrula chick embryos. Finally, the DiI-labeled cell migration trajectory indicated that imidacloprid restricted the cell migration of cardiac progenitors to primary heart field in gastrula chick embryos. A similar observation was also obtained from the cell migration assay of scratch wounds in vitro. Additionally, imidacloprid exposure negatively affected the cytoskeleton structure and expression of corresponding adhesion molecules. Taken together, these results reveal that the improper EMT, cardiac progenitor migration, and differentiation are responsible for imidacloprid exposure-induced malformation of heart tube during chick embryo development.


Subject(s)
Gene Expression Regulation, Developmental , Heart Valve Diseases/pathology , Heart/drug effects , Heart/embryology , Imidazoles/toxicity , Insecticides/toxicity , Nitro Compounds/toxicity , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Chick Embryo , Down-Regulation , Epithelial-Mesenchymal Transition/drug effects , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Heart Valve Diseases/chemically induced , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Laminin/genetics , Laminin/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neonicotinoids , Rats , Up-Regulation , Wnt3A Protein/genetics , Wnt3A Protein/metabolism
20.
Front Pharmacol ; 7: 349, 2016.
Article in English | MEDLINE | ID: mdl-27746734

ABSTRACT

Phenobarbital is an antiepileptic drug that is widely used to treat epilepsy in a clinical setting. However, a long term of phenobarbital administration in pregnant women may produce side effects on embryonic skeletogenesis. In this study, we aim to investigate the mechanism by which phenobarbital treatment induces developmental defects in long bones. We first determined that phenobarbital treatment decreased chondrogenesis and inhibited the proliferation of chondrocytes in chick embryos. Phenobarbital treatment also suppressed mineralization in both in vivo and in vitro long bone models. Next, we established that phenobarbital treatment delayed blood vessel invasion in a cartilage template, and this finding was supported by the down-regulation of vascular endothelial growth factor in the hypertrophic zone following phenobarbital treatment. Phenobarbital treatment inhibited tube formation and the migration of human umbilical vein endothelial cells. In addition, it impaired angiogenesis in chick yolk sac membrane model and chorioallantoic membrane model. In summary, phenobarbital exposure led to shortened lengths of long bones during embryogenesis, which might result from inhibiting mesenchyme differentiation, chondrocyte proliferation, and delaying mineralization by impairing vascular invasion.

SELECTION OF CITATIONS
SEARCH DETAIL
...