Antisense Morpholino Oligonucleotide Analog Induces Missplicing of C-mycmRNA

1999 ◽  
Vol 9 (2) ◽  
pp. 213-220 ◽  
Author(s):  
RICHARD V. GILES ◽  
DAVID G. SPILLER ◽  
RICHARD E. CLARK ◽  
DAVID M. TIDD
2015 ◽  
Vol 84 (7) ◽  
pp. 1246-1255.e1 ◽  
Author(s):  
Zuzana Linhartová ◽  
Taiju Saito ◽  
Vojtěch Kašpar ◽  
Marek Rodina ◽  
Eva Prášková ◽  
...  

2007 ◽  
Vol 292 (1) ◽  
pp. R470-R480 ◽  
Author(s):  
Masahiro Esaki ◽  
Kazuyuki Hoshijima ◽  
Sayako Kobayashi ◽  
Hidekazu Fukuda ◽  
Koichi Kawakami ◽  
...  

Uptake of Na+ from the environment is an indispensable strategy for the survival of freshwater fish, as they easily lose Na+ from the plasma to a diluted environment. Nevertheless, the location of and molecules involved in Na+ uptake remain poorly understood. In this study, we utilized Sodium Green, a Na+-dependent fluorescent reagent, to provide direct evidence that Na+ absorption takes place in a subset of the mitochondria-rich (MR) cells on the yolk sac surface of zebrafish larvae. Combined with immunohistochemistry, we revealed that the Na+-absorbing MR cells were exceptionally rich in vacuolar-type H+-ATPase (H+-ATPase) but moderately rich in Na+-K+-ATPase. We also addressed the function of foxi3a, a transcription factor that is specifically expressed in the H+-ATPase-rich MR cells. When foxi3a was depleted from zebrafish embryos by antisense morpholino oligonucleotide injection, differentiation of the MR cells was completely blocked and Na+ influx was severely reduced, indicating that MR cells are the primary sites for Na+ absorption. Additionally, foxi3a expression is initiated at the gastrula stage in the presumptive ectoderm; thus, we propose that foxi3a is a key gene in the control of MR cell differentiation. We also utilized a set of ion transport inhibitors to assess the molecules involved in the process and discuss the observations.


2006 ◽  
Vol 174 (6) ◽  
pp. 791-801 ◽  
Author(s):  
Suzanne Madgwick ◽  
David V. Hansen ◽  
Mark Levasseur ◽  
Peter K. Jackson ◽  
Keith T. Jones

During interkinesis, a metaphase II (MetII) spindle is built immediately after the completion of meiosis I. Oocytes then remain MetII arrested until fertilization. In mouse, we find that early mitotic inhibitor 2 (Emi2), which is an anaphase-promoting complex inhibitor, is involved in both the establishment and the maintenance of MetII arrest. In MetII oocytes, Emi2 needs to be degraded for oocytes to exit meiosis, and such degradation, as visualized by fluorescent protein tagging, occurred tens of minutes ahead of cyclin B1. Emi2 antisense morpholino knockdown during oocyte maturation did not affect polar body (PB) extrusion. However, in interkinesis the central spindle microtubules from meiosis I persisted for a short time, and a MetII spindle failed to assemble. The chromatin in the oocyte quickly decondensed and a nucleus formed. All of these effects were caused by the essential role of Emi2 in stabilizing cyclin B1 after the first PB extrusion because in Emi2 knockdown oocytes a MetII spindle was recovered by Emi2 rescue or by expression of nondegradable cyclin B1 after meiosis I.


Open Biology ◽  
2020 ◽  
Vol 10 (2) ◽  
pp. 190273
Author(s):  
I-Chen Hung ◽  
Tsung-Ming Chen ◽  
Jing-Ping Lin ◽  
Yu-Ling Tai ◽  
Tang-Long Shen ◽  
...  

Focal adhesion kinase (FAK) mediates vital cellular pathways during development. Despite its necessity, how FAK regulates and integrates with other signals during early embryogenesis remains poorly understood. We found that the loss of Fak1a impaired epiboly, convergent extension and hypoblast cell migration in zebrafish embryos. We also observed a clear disturbance in cortical actin at the blastoderm margin and distribution of yolk syncytial nuclei. In addition, we investigated a possible link between Fak1a and a well-known gastrulation regulator, Wnt5b, and revealed that the overexpression of fak1a or wnt5b could cross-rescue convergence defects induced by a wnt5b or fak1a antisense morpholino (MO), respectively. Wnt5b and Fak1a were shown to converge in regulating Rac1 and Cdc42, which could synergistically rescue wnt5b and fak1a morphant phenotypes. Furthermore, we generated several alleles of fak1a mutants using CRISPR/Cas9, but those mutants only revealed mild gastrulation defects. However, injection of a subthreshold level of the wnt5b MO induced severe gastrulation defects in fak1a mutants, which suggested that the upregulated expression of wnt5b might complement the loss of Fak1a. Collectively, we demonstrated that a functional interaction between Wnt and FAK signalling mediates gastrulation cell movements via the possible regulation of Rac1 and Cdc42 and subsequent actin dynamics.


Sign in / Sign up

Export Citation Format

Share Document