scholarly journals Prenatal Treatment of Mosaic Mice (Atp7a mo-ms) Mouse Model for Menkes Disease, with Copper Combined by Dimethyldithiocarbamate (DMDTC)

PLoS ONE ◽  
2012 ◽  
Vol 7 (7) ◽  
pp. e40400 ◽  
Author(s):  
Małgorzata Lenartowicz ◽  
Wojciech Krzeptowski ◽  
Paweł Koteja ◽  
Katarzyna Chrząścik ◽  
Lisbeth Birk Møller
1990 ◽  
Vol 267 (2) ◽  
pp. 417-422 ◽  
Author(s):  
G L Waldrop ◽  
M J Ettinger

Fibroblasts from the brindled mouse model of Menkes disease are known to accumulate excess copper. Most of the copper in the cytosol of these fibroblasts is bound to metallothionein (MT), which is elevated in Menkes or brindled mouse fibroblasts. Copper accumulation by normal fibroblasts containing excess MT was examined to determine if the excess copper accumulation phenotype was secondary to excess MT or associated with the primary defect in fibroblasts from the brindled mice. MT was induced in normal fibroblasts by copper, zinc or dexamethasone to levels comparable with those in brindled mice fibroblasts, as determined by radioimmunoassays. Normal fibroblasts containing excess MT accumulate copper normally, i.e. they do not exhibit the excess copper accumulation phenotype. Consistent with this result, copper efflux from normal fibroblasts containing excess MT was also normal. The data suggest that one function of the protein associated with the primary defect is to help determine how much copper is taken up and retained by fibroblasts and other cell types exhibiting the excess copper phenotype in Menkes disease. The capacity of this protein is apparently exceeded in normal fibroblasts if serum or albumin is not present extracellularly to limit total copper uptake. Consistent with a defect in an intracellular protein, the kinetics of copper transport by brindled mice fibroblasts were found to be normal.


2020 ◽  
Vol 107 (5) ◽  
pp. 911-931
Author(s):  
Faycal Guedj ◽  
Ashley E. Siegel ◽  
Jeroen L.A. Pennings ◽  
Fatimah Alsebaa ◽  
Lauren J. Massingham ◽  
...  

2018 ◽  
Vol 41 (6) ◽  
pp. 1285-1291 ◽  
Author(s):  
Takao Hoshina ◽  
Satoshi Nozaki ◽  
Takashi Hamazaki ◽  
Satoshi Kudo ◽  
Yuka Nakatani ◽  
...  

2018 ◽  
Vol 10 ◽  
pp. 165-178 ◽  
Author(s):  
Marie Reine Haddad ◽  
Eun-Young Choi ◽  
Patricia M. Zerfas ◽  
Ling Yi ◽  
Diego Martinelli ◽  
...  

2009 ◽  
Vol 201 (6) ◽  
pp. S255
Author(s):  
Maddalena Incerti ◽  
Robin Roberson ◽  
Christopher Nold ◽  
Laura Toso ◽  
Joy Vink ◽  
...  

1997 ◽  
Vol 42 (4) ◽  
pp. 436-442 ◽  
Author(s):  
Yoshiko Murata ◽  
Hiroko Kodama ◽  
Toshiaki Abe ◽  
Norio Ishida ◽  
Masahiko Nishimura ◽  
...  

2018 ◽  
Author(s):  
Faycal Guedj ◽  
Jeroen LA Pennings ◽  
Ashley E Siegel ◽  
Fatimah Alsebaa ◽  
Lauren J Massingham ◽  
...  

ABSTRACTHuman fetuses with trisomy 21 (T21) have atypical brain development that is apparent sonographically in the second trimester. Prenatal diagnosis provides a potential opportunity to begin treatment in utero. We hypothesize that by analyzing and integrating dysregulated gene expression and pathways common to humans with DS and mouse models we can discover novel targets for therapy. Here, we tested the safety and efficacy of apigenin (4’, 5, 7-trihydroxyflavone), identified using this approach, in both human amniocytes from fetuses with T21 and in the Ts1Cje mouse model. The experiments compared treated to untreated results in T21 and euploid cells, as well as in Ts1Cje mice and their wild-type littermate controls. T21 cells cultured with apigenin (2µM) had significantly reduced oxidative stress and improved antioxidant defense response in vitro. Apigenin (333-400 mg/kg/day), mixed with chow, was initiated prenatally to the dams and fed to the pups over their lifetimes. There was no significant increase in birth defects or pup deaths resulting from prenatal apigenin treatment. Apigenin significantly improved several developmental milestones and spatial olfactory memory in Ts1Cje neonates. In addition, we noted sex-specific effects on exploratory behavior and long-term hippocampal memory in adult mice, with males showing significantly more improvement than females. Global gene expression analyses demonstrated that apigenin targets similar signaling pathways through common upstream regulators both in vitro and in vivo. These studies provide proof-of-principle that apigenin has therapeutic effects in preclinical models of Down syndrome.ONE SENTENCE SUMMARYAs a candidate prenatal treatment for Down syndrome, apigenin improved oxidative stress/antioxidant capacity imbalance and reduced pathways associated with inflammation in human cells while improving aspects of behavior in the Ts1Cje mouse model.


2013 ◽  
Vol 208 (1) ◽  
pp. S41 ◽  
Author(s):  
Alissa Carver ◽  
Maged Costantine ◽  
Esther Tamayo ◽  
Huaizhi Yin ◽  
J. Regino Perez-Polo ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document