A novel pathological role of p53 in kidney development revealed by gene-environment interactions

2005 ◽  
Vol 288 (1) ◽  
pp. F98-F107 ◽  
Author(s):  
Hao Fan ◽  
Jessica R. Harrell ◽  
Susana Dipp ◽  
Zubaida Saifudeen ◽  
Samir S. El-Dahr

Gene-environment interactions are implicated in congenital human disorders. Accordingly, there is a pressing need to develop animal models of human disease, which are the product of defined gene-environment interactions. Previously, our laboratory demonstrated that gestational salt stress of bradykinin B2 receptor (B2R)-null mice induces renal dysgenesis and early death of the offspring (El-Dahr SS, Harrison-Bernard LM, Dipp S, Yosipiv IV, and Meleg-Smith S. Physiol Genomics 3: 121–131, 2000). In contrast, salt-stressed B2R +/+ or +/− littermates have normal development. The present study investigates the mechanisms underlying the susceptibility of B2R-null mice to renal dysgenesis. Proteomic and conventional Western blot screens identified E-cadherin among the differentially repressed proteins in B2R−/− kidneys, whereas the checkpoint kinase Chk1 and its substrate P-Ser20 p53 were induced. We tested the hypothesis that p53 mediates repression of E-cadherin gene expression and is causally linked to the renal dysgenesis. Genetic crosses between B2R −/− and p53+/− mice revealed that germline reduction of p53 gene dosage rescues B2R−/− mice from renal dysgenesis and restores kidney E-cadherin gene expression. Furthermore, γ-irradiation induces repression of E-cadherin gene expression in p53+/+ but not −/− cells. In transient transfection assays, p53 repressed human E-cadherin promoter-driven reporter activity, whereas a mutant p53, which cannot bind DNA, did not. Functional promoter analysis indicated the presence of a p53-responsive element in exon 1, which partially mediates p53-induced repression. Chromatin immunoprecipitation assays revealed that p53 inhibits histone acetylation of the E-cadherin promoter. Treatment with a histone deacetylase inhibitor reversed both p53-mediated promoter repression and deacetylation. In conclusion, this study demonstrates that gene-environment interactions cooperate to induce congenital defects through p53 activation.

Epigenomics ◽  
2019 ◽  
Vol 11 (16) ◽  
pp. 1827-1837 ◽  
Author(s):  
Shihoko Kojima ◽  
Daniela Cimini

Aneuploidy (i.e., abnormal chromosome number) is the leading cause of miscarriage and congenital defects in humans. Moreover, aneuploidy is ubiquitous in cancer. The deleterious phenotypes associated with aneuploidy are likely a result of the imbalance in the levels of gene products derived from the additional chromosome(s). Here, we summarize the current knowledge on how the presence of extra chromosomes impacts gene expression. We describe studies that have found a strict correlation between gene dosage and transcript levels as wells as studies that have found a less stringent correlation, hinting at the possible existence of dosage compensation mechanisms. We conclude by peering into the epigenetic changes found in aneuploid cells and outlining current knowledge gaps and potential areas of future investigation.


2005 ◽  
Vol 37 (3) ◽  
pp. 485-492 ◽  
Author(s):  
Arisan Serdar ◽  
Caskurlu Turhan ◽  
Guney Soner ◽  
Sonmez Nurettin Cem ◽  
Keles Bayram ◽  
...  

1999 ◽  
Vol 276 (4) ◽  
pp. C946-C954 ◽  
Author(s):  
Li Li ◽  
Ji Li ◽  
Jaladanki N. Rao ◽  
Minglin Li ◽  
Barbara L. Bass ◽  
...  

The nuclear phosphoprotein p53 acts as a transcription factor and is involved in growth inhibition and apoptosis. The present study was designed to examine the effect of decreasing cellular polyamines on p53 gene expression and apoptosis in small intestinal epithelial (IEC-6) cells. Cells were grown in DMEM containing 5% dialyzed fetal bovine serum in the presence or absence of α-difluoromethylornithine (DFMO), a specific inhibitor of polyamine biosynthesis, for 4, 6, and 12 days. The cellular polyamines putrescine, spermidine, and spermine in DFMO-treated cells decreased dramatically at 4 days and remained depleted thereafter. Polyamine depletion by DFMO was accompanied by a significant increase in expression of the p53 gene. The p53 mRNA levels increased 4 days after exposure to DFMO, and the maximum increases occurred at 6 and 12 days after exposure. Increased levels of p53 mRNA in DFMO-treated cells were paralleled by increases in p53 protein. Polyamines given together with DFMO completely prevented increased expression of the p53 gene. Increased expression of the p53 gene in DFMO-treated cells was associated with a significant increase in G1 phase growth arrest. In contrast, no features of programmmed cell death were identified after polyamine depletion: no internucleosomal DNA fragmentation was observed, and no morphological features of apoptosis were evident in cells exposed to DFMO for 4, 6, and 12 days. These results indicate that 1) decreasing cellular polyamines increases expression of the p53 gene and 2) activation of p53 gene expression after polyamine depletion does not induce apoptosis in intestinal crypt cells. These findings suggest that increased expression of the p53 gene may play an important role in growth inhibition caused by polyamine depletion.


2008 ◽  
Vol 121 (13) ◽  
pp. 2224-2234 ◽  
Author(s):  
G. Solanas ◽  
M. Porta-de-la-Riva ◽  
C. Agusti ◽  
D. Casagolda ◽  
F. Sanchez-Aguilera ◽  
...  

2001 ◽  
Vol 34 (5) ◽  
pp. 415-420 ◽  
Author(s):  
George J Soleas ◽  
David M Goldberg ◽  
Linda Grass ◽  
Michael Levesque ◽  
Eleftherios P Diamandis

PLoS Genetics ◽  
2010 ◽  
Vol 6 (5) ◽  
pp. e1000944 ◽  
Author(s):  
Jean-François Gout ◽  
Daniel Kahn ◽  
Laurent Duret ◽  

2010 ◽  
Vol 17 (12) ◽  
pp. 3379-3385 ◽  
Author(s):  
Ming-Te Huang ◽  
Po-Li Wei ◽  
Jun-Jen Liu ◽  
Der-Zen Liu ◽  
Huang Huey-Chun ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document