Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Sci Adv ; 9(49): eadh8152, 2023 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-38055823

RESUMEN

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.


Asunto(s)
Actomiosina , Gastrulación , Animales , Embrión de Pollo , Gástrula , Vertebrados
2.
Elife ; 122023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37039463

RESUMEN

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.


Asunto(s)
Gastrulación , Mecanotransducción Celular , Animales , Embrión de Pollo , Retroalimentación , Gastrulación/fisiología , Morfogénesis , Uniones Intercelulares
3.
Sci Adv ; 9(1): eabn5429, 2023 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-36598979

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-32381735

RESUMEN

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.


Asunto(s)
Embrión de Pollo/citología , Embrión de Pollo/crecimiento & desarrollo , Drosophila melanogaster/embriología , Modelos Biológicos , Animales , Animales Modificados Genéticamente , Fenómenos Biomecánicos , Embrión de Pollo/efectos de los fármacos , Simulación por Computador , Proteínas de Drosophila/genética , Embrión no Mamífero/citología , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/metabolismo , Gástrula/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Indazoles/farmacología , Microscopía/métodos , Morfogénesis , Mutación , Proteína 1 Relacionada con Twist/genética
5.
Ecotoxicol Environ Saf ; 194: 110415, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32151871

RESUMEN

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.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Nanopartículas/toxicidad , Cresta Neural/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Óxido de Zinc/toxicidad , Acetilcisteína/farmacología , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Femenino , Células HEK293 , Humanos , Mitocondrias/efectos de los fármacos , FN-kappa B/metabolismo , Nanopartículas/química , Cresta Neural/embriología , Especies Reactivas de Oxígeno/metabolismo , Óxido de Zinc/química
6.
Development ; 147(3)2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31964776

RESUMEN

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.


Asunto(s)
Células Epiteliales/metabolismo , Gastrulación/fisiología , Uniones Intercelulares/metabolismo , Pinzas Ópticas , Línea Primitiva/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Embrión de Pollo , Gástrula/metabolismo , Estratos Germinativos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hidrocarburos Clorados/farmacología , Microscopía Fluorescente/métodos , Miosina Tipo I/antagonistas & inhibidores , Miosina Tipo I/metabolismo , Miosina Tipo II/antagonistas & inhibidores , Miosina Tipo II/metabolismo , Pirroles/farmacología , Transducción de Señal/fisiología
7.
Exp Clin Endocrinol Diabetes ; 127(9): 590-597, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28950393

RESUMEN

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.


Asunto(s)
Membrana Corioalantoides , Desarrollo Embrionario/efectos de los fármacos , Glucosa/farmacología , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Saco Vitelino , Animales , Embrión de Pollo , Membrana Corioalantoides/metabolismo , Membrana Corioalantoides/patología , Glucosa/metabolismo , Hiperplasia/inducido químicamente , Hiperplasia/embriología , Hiperplasia/patología , Miocitos Cardíacos/patología , Saco Vitelino/metabolismo , Saco Vitelino/patología
8.
Zygote ; 26(6): 457-464, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30520400

RESUMEN

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.


Asunto(s)
Embrión de Pollo/citología , Embrión de Pollo/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Cresta Neural/citología , Animales , Cadherinas/genética , Cadherinas/metabolismo , Movimiento Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/metabolismo , Tubo Neural/embriología , Tubo Neural/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
9.
Cell Physiol Biochem ; 48(5): 2084-2090, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30099448

RESUMEN

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.


Asunto(s)
Antagonistas de los Receptores de la Endotelina B/farmacología , Lipopolisacáridos/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Animales , Pollos , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Endotelina-1/genética , Endotelina-1/metabolismo , Oligopéptidos/farmacología , Péptidos Cíclicos/farmacología , Piperidinas/farmacología , Receptor de Endotelina A/química , Receptor de Endotelina A/genética , Receptor de Endotelina B/química , Receptor de Endotelina B/genética , Transducción de Señal/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
J Cell Physiol ; 233(9): 7120-7133, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29574800

RESUMEN

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.


Asunto(s)
Gastrulación/efectos de los fármacos , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/metabolismo , Corazón/embriología , Cloruro de Sodio Dietético/toxicidad , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Pollos , Desarrollo Embrionario/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Cardiopatías Congénitas/patología , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas
11.
Cell Death Dis ; 9(2): 234, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29445081

RESUMEN

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.


Asunto(s)
Autofagia/efectos de los fármacos , Sistema Cardiovascular/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Flavonoides/farmacología , Hipoglucemiantes/farmacología , Organogénesis/efectos de los fármacos , Animales , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Autofagia/genética , Glucemia/metabolismo , Sistema Cardiovascular/crecimiento & desarrollo , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patología , Embrión de Pollo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Embrión no Mamífero , Femenino , Regulación de la Expresión Génica , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Organogénesis/genética , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Estreptozocina , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
12.
Mol Neurobiol ; 55(4): 3523-3536, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28509082

RESUMEN

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.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/metabolismo , Autofagia , Cresta Neural/citología , Neurogénesis , Animales , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Proteína 7 Relacionada con la Autofagia/genética , Familia de las Proteínas 8 Relacionadas con la Autofagia/genética , Familia de las Proteínas 8 Relacionadas con la Autofagia/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Ciclo Celular , Línea Celular Tumoral , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Modelos Biológicos , Cresta Neural/metabolismo , Cresta Neural/ultraestructura , Tubo Neural/citología , Tubo Neural/embriología , Tubo Neural/metabolismo , Tubo Neural/ultraestructura , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
13.
J Cell Physiol ; 233(12): 9271-9283, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-28885685

RESUMEN

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.


Asunto(s)
Endotoxinas/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Cardiopatías Congénitas/embriología , Corazón/embriología , Organogénesis/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Cardiopatías Congénitas/genética , Humanos , Lipopolisacáridos/toxicidad , Masculino , Ratones , Modelos Biológicos , Organogénesis/genética , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Reproducción/efectos de los fármacos
14.
Exp Cell Res ; 361(1): 73-84, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28987541

RESUMEN

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.


Asunto(s)
Anomalías Craneofaciales/patología , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/fisiología , Cresta Neural/citología , Receptores Inmunológicos/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Embrión de Pollo , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados , Cresta Neural/metabolismo , Neurogénesis , Proteínas Roundabout
15.
Toxicol Lett ; 281: 53-64, 2017 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-28919490

RESUMEN

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.


Asunto(s)
Anomalías Craneofaciales/patología , Etanol/toxicidad , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/efectos de los fármacos , Organogénesis/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Antígenos CD57/genética , Antígenos CD57/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Embrión de Pollo , Anomalías Craneofaciales/inducido químicamente , Modelos Animales de Enfermedad , Regulación hacia Abajo , Trastornos del Espectro Alcohólico Fetal/fisiopatología , Laminina/genética , Laminina/metabolismo , Cresta Neural/patología , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo
16.
Reprod Toxicol ; 73: 96-104, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28789864

RESUMEN

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.


Asunto(s)
Embrión de Pollo/efectos de los fármacos , Etanol/toxicidad , Hemangioblastos/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Embrión de Pollo/embriología , Desarrollo Embrionario/efectos de los fármacos , Hemangioblastos/fisiología , Especies Reactivas de Oxígeno/metabolismo
17.
Toxicol Sci ; 158(1): 36-47, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28453788

RESUMEN

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.


Asunto(s)
Dexametasona/efectos adversos , Cresta Neural/patología , Animales , Embrión de Pollo , Hibridación in Situ , Factores de Riesgo
18.
Cell Death Dis ; 8(3): e2697, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28333135

RESUMEN

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.


Asunto(s)
Muerte Celular/fisiología , Atresia Folicular/metabolismo , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Folículo Ovárico/metabolismo , Ovario/metabolismo , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Daño del ADN/fisiología , Reparación del ADN/fisiología , Femenino , Atresia Folicular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Folículo Ovárico/fisiología
19.
J Agric Food Chem ; 64(47): 9078-9088, 2016 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-27792329

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Enfermedades de las Válvulas Cardíacas/patología , Corazón/efectos de los fármacos , Corazón/embriología , Imidazoles/toxicidad , Insecticidas/toxicidad , Nitrocompuestos/toxicidad , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Cadherinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Embrión de Pollo , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Enfermedades de las Válvulas Cardíacas/inducido químicamente , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Laminina/genética , Laminina/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Neonicotinoides , Ratas , Regulación hacia Arriba , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
20.
Front Pharmacol ; 7: 349, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27746734

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA