scholarly journals Confocal Imaging of Early Heart Development in Xenopus laevis

2000 ◽  
Vol 218 (1) ◽  
pp. 64-73 ◽  
Author(s):  
Sandra J Kolker ◽  
Urszula Tajchman ◽  
Daniel L Weeks
2007 ◽  
Vol 31 (2) ◽  
pp. 81-89
Author(s):  
Won Seok Koh ◽  
Li Guang Li ◽  
In-Shik Kim ◽  
Young-Hoon Lee ◽  
Byung Seok Ko ◽  
...  

2011 ◽  
Vol 114 (2) ◽  
pp. 391-400 ◽  
Author(s):  
Dao-jie Xu ◽  
Ji-wen Bu ◽  
Shan-ye Gu ◽  
Yi-meng Xia ◽  
Jiu-lin Du ◽  
...  

Background Celecoxib, a cyclooxygenase-2 inhibitor, is a commonly ingested drug that is used by some women during pregnancy. Although use of celecoxib is associated with increased cardiovascular risk in adults, its effect on fetal heart development remains unknown. Methods Zebrafish embryos were exposed to celecoxib or other relevant drugs from tailbud stage (10.3-72 h postfertilization). Heart looping and valve formation were examined at different developmental stages by in vivo confocal imaging. In addition, whole mount in situ hybridization was performed to examine drug-induced changes in the expression of heart valve marker genes. Results In celecoxib-treated zebrafish embryos, the heart failed to undergo normal looping and the heart valve was absent, causing serious blood regurgitation. Furthermore, celecoxib treatment disturbed the restricted expression of the heart valve markers bone morphogenetic protein 4 and versican-but not the cardiac chamber markers cardiac myosin light chain 2, ventricular myosin heavy chain, and atrial myosin heavy chain. These defects in heart development were markedly relieved by treatment with the cyclooxygenase-2 downstream product prostaglandin E2, and mimicked by the cyclooxygenase-2 inhibitor NS398, implying that celecoxib-induced heart defects were caused by the inhibition of cyclooxygenase-2 activity. Conclusions These findings provide the first in vivo evidence that celecoxib exposure impairs heart development in zebrafish embryos by inhibiting cyclooxygenase-2 activity.


2000 ◽  
Vol 218 (1) ◽  
pp. 74-88 ◽  
Author(s):  
Timothy J. Mohun ◽  
Li Ming Leong ◽  
Wolfgang J. Weninger ◽  
Duncan B. Sparrow

2018 ◽  
Author(s):  
Madison C. Sestak ◽  
Julia A. Pinette ◽  
Caithlin M. Lamoureux ◽  
Susan L. Whittemore

AbstractPolycyclic aromatic hydrocarbons (PAHs) are ubiquitous, persistent environmental contaminants, of which 16 are EPA-designated priority pollutants. Cardiotoxicity is observed in fish with developmental exposures to certain PAHs; however, the mechanism of toxicity can differ. Phenanthrene (PHE) and benzo(a)pyrene (BaP) are both cardiotoxic to fish, but PHE acts independently of aryl hydrocarbon receptor (AHR) activation while BaP-associated cardiotoxicity is AHR-dependent. To further understanding of mechanisms of toxicity, we compared the effects of early exposure to the priority PAHs pyrene (PYR), fluoranthene (FLA), PHE and BaP on cardiac function and cytochrome P450 type 1A (cyp1a) mRNA expression, an indicator of AHR activation, in a model system with lower AHR sensitivity than that of fish, the embryos and larvae of Xenopus laevis. Exposure to PYR, PHE, and FLA (0.25 – 25 μM) caused ventricular tachycardia early in heart development, but bradycardia and atrioventricular (AV) block in later stages. Elevated cyp1a mRNA levels indicate that FLA and BaP, but not PHE or PYR, are AHR agonists. The finding of FLA-induced cardiotoxicity and cyp1a expression (35-fold) is particularly surprising as FLA inhibits CYP1A activity in fish and, as a single compound, is not cardiotoxic. Our results suggest that early exposure to PHE, PYR, and FLA, but not to BaP, compromises cardiac function by altering normal pacemaker activity and conduction in Xenopus, effects associated with increased mortality. Our findings also reveal a considerable degree of species specificity between fish and frog regarding cardiac sensitivity to developmental PAH exposures and have implications for the cardiovascular health of PAH-exposed humans and wild amphibians.


2005 ◽  
Vol 114 (11) ◽  
pp. 853-858 ◽  
Author(s):  
Brian C. Bane ◽  
Jana M. Van Rybroek ◽  
Sandra J. Kolker ◽  
Daniel L. Weeks ◽  
Jose M. Manaligod

Objectives: We sought to determine the developmental anatomy and EYA1 protein distribution in the inner ear of Xenopus laevis. Methods: Xenopus laevis embryos were stained with monoclonal antibodies and imaged with confocal microscopy. Results: At stage 27, the otocyst fully forms, with strong tubulin staining of early sensory cells at its ventromedial aspect. Neuronal ingrowth follows at stage 33/34. At stage 50, the semicircular canals are complete. EYA1 localizes to the anterior aspect of the otocyst from stages 37 to 44. By stage 50, EYA1 distribution is localized primarily to the sensory maculae and the endolymphatic duct of the developing inner ear. Conclusions: Whole mount confocal imaging of the developing Xenopus inner ear delineates the exact timing of otic development, sensory cell differentiation, and innervation. EYA1 protein expression has a distinct distribution pattern at the anterior aspect of the developing otocyst in stages 41 and 44. Later stages have a more localized pattern, in which EYA1 is detected only in the sensory epithelium and endolymphatic duct. This specific pattern of expression indicates a possible role in the determination of the anterior-posterior orientation of the inner ear, as well as a later role in sensory cell differentiation.


2007 ◽  
Vol 27 (18) ◽  
pp. 6420-6432 ◽  
Author(s):  
Yong Chen ◽  
Wai Hong Yuen ◽  
Jianlin Fu ◽  
Guochang Huang ◽  
Alirio J. Melendez ◽  
...  

ABSTRACT The mitochondrial respiratory chain (MRC) plays crucial roles in cellular energy production. However, its function in early embryonic development remains largely unknown. To address this issue, GRIM-19, a newly identified MRC complex I subunit, was knocked down in Xenopus laevis embryos. A severe deficiency in heart formation was observed, and the deficiency could be rescued by reintroducing human GRIM-19 mRNA. The mechanism involved was further investigated. We found that the activity of NFAT, a transcription factor family that contributes to early organ development, was downregulated in GRIM-19 knockdown embryos. Furthermore, the expression of a constitutively active form of mouse NFATc4 in these embryos rescued the heart developmental defects. NFAT activity is controlled by a calcium-dependent protein phosphatase, calcineurin, which suggests that calcium signaling may be disrupted by GRIM-19 knockdown. Indeed, both the calcium response and calcium-induced NFAT activity were impaired in the GRIM-19 or NDUFS3 (another complex I subunit) knockdown cell lines. We also showed that NFAT can rescue expression of Nkx2.5, which is one of the key genes for early heart development. Our data demonstrated the essential role of MRC in heart formation and revealed the signal transduction and gene expression cascade involved in this process.


1993 ◽  
Vol 187 (3) ◽  
Author(s):  
YvetteN. Frunchak ◽  
GaleN. Martha ◽  
KennethD. McFadden ◽  
NadineC. Milos

Sign in / Sign up

Export Citation Format

Share Document