Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
FEBS Lett ; 597(9): 1233-1245, 2023 05.
Article in English | MEDLINE | ID: mdl-36445168

ABSTRACT

Prolonged metabolic stress can lead to severe pathologies. In metabolically challenged primary fibroblasts, we assigned a novel role for the poorly characterized miR-4734 in restricting ATF4 and IRE1-mediated upregulation of a set of proinflammatory cytokines and endoplasmic reticulum stress-associated genes. Conversely, inhibition of this miRNA augmented the expression of those genes. Mechanistically, miR-4734 was found to restrict the expression of the transcriptional activator NF-kappa-B inhibitor zeta (NFKBIZ), which is required for optimal expression of the proinflammatory genes and whose mRNA is targeted directly by miR-4734. Concordantly, overexpression of NFKBIZ compromised the effects of miR-4734, underscoring the importance of this direct targeting. As the effects of miR-4734 were evident under stress but not under basal conditions, it may possess therapeutic utility towards alleviating stress-induced pathologies.


Subject(s)
MicroRNAs , Cytokines/genetics , Cytokines/metabolism , Endoplasmic Reticulum Stress/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Transcription Factors/metabolism , Up-Regulation , Humans
2.
Proc Natl Acad Sci U S A ; 107(43): 18511-6, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20937856

ABSTRACT

The p53 tumor suppressor is mutated in a high percentage of human tumors. However, many other tumors retain wild-type (wt) p53 expression, raising the intriguing possibility that they actually benefit from it. Recent studies imply a role for p53 in regulation of autophagy, a catabolic pathway by which eukaryotic cells degrade and recycle macromolecules and organelles, particularly under conditions of nutrient deprivation. Here, we show that, in many cell types, p53 confers increased survival in the face of chronic starvation. We implicate regulation of autophagy in this effect. In HCT116 human colorectal cancer cells exposed to prolonged nutrient deprivation, the endogenous wt p53 posttranscriptionally down-regulates LC3, a pivotal component of the autophagic machinery. This enables reduced, yet sustainable autophagic flux. Loss of p53 impairs autophagic flux and causes excessive LC3 accumulation upon starvation, culminating in apoptosis. Thus, p53 increases cell fitness by maintaining better autophagic homeostasis, adjusting the rate of autophagy to changing circumstances. We propose that some cancer cells retain wt p53 to benefit from the resultant increased fitness under limited nutrient supply.


Subject(s)
Cell Survival/physiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Autophagy/physiology , Base Sequence , Cell Line, Tumor , Culture Media , DNA Primers/genetics , Down-Regulation , Gene Knockdown Techniques , Gene Knockout Techniques , Genes, p53 , Humans , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Phagosomes/metabolism , Phagosomes/ultrastructure , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL