Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Res ; 24(4): 433-50, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24662486

ABSTRACT

Activation of Notch signaling requires intracellular routing of the receptor, but the mechanisms controlling the distinct steps in the routing process is poorly understood. We identify PKCζ as a key regulator of Notch receptor intracellular routing. When PKCζ was inhibited in the developing chick central nervous system and in cultured myoblasts, Notch-stimulated cells were allowed to undergo differentiation. PKCζ phosphorylates membrane-tethered forms of Notch and regulates two distinct routing steps, depending on the Notch activation state. When Notch is activated, PKCζ promotes re-localization of Notch from late endosomes to the nucleus and enhances production of the Notch intracellular domain, which leads to increased Notch activity. In the non-activated state, PKCζ instead facilitates Notch receptor internalization, accompanied with increased ubiquitylation and interaction with the endosomal sorting protein Hrs. Collectively, these data identify PKCζ as a key regulator of Notch trafficking and demonstrate that distinct steps in intracellular routing are differentially modulated depending on Notch signaling status.


Subject(s)
Protein Kinase C/physiology , Receptor, Notch1/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Chick Embryo , HEK293 Cells , HeLa Cells , Humans , Mice , Molecular Sequence Data , Protein Transport , Receptor, Notch1/genetics , Sequence Homology, Amino Acid , Signal Transduction/genetics
2.
Mol Biol Cell ; 23(21): 4323-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22933572

ABSTRACT

The AP-1 transcription factor c-Jun has been shown to be essential for stress-induced apoptosis in several models. However, the molecular mechanisms underlying the proapoptotic activity of c-Jun are poorly understood. We identify the apoptosis-antagonizing transcription factor (AATF) as a novel nucleolar stress sensor, which is required as a cofactor for c-Jun-mediated apoptosis. Overexpression or down-regulation of AATF expression levels led to a respective increase or decrease in the amount of activated and phosphorylated c-Jun with a proportional alteration in the induction levels of the proapoptotic c-Jun target genes FasL and TNF-α. Accordingly, AATF promoted commitment of ultraviolet (UV)-irradiated cells to c-Jun-dependent apoptosis. Whereas AATF overexpression potentiated UV-induced apoptosis in wild-type cells, c-Jun-deficient mouse embryonic fibroblasts were resistant to AATF-mediated apoptosis induction. Furthermore, AATF mutants defective in c-Jun binding were also defective in inducing AP-1 activity and c-Jun-mediated apoptosis. UV irradiation induced a translocation of AATF from the nucleolus to the nucleus, thereby enabling its physical association to c-Jun. Analysis of AATF deletion mutants revealed that the AATF domains required for compartmentalization, c-Jun binding, and enhancement of c-Jun transcriptional activity were all also required to induce c-Jun-dependent apoptosis. These results identify AATF as a nucleolar-confined c-Jun cofactor whose expression levels and spatial distribution determine the stress-induced activity of c-Jun and the levels of c-Jun-mediated apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Cell Nucleolus/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/chemistry , Cell Nucleolus/radiation effects , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , HEK293 Cells , Humans , Mice , Mice, Knockout , Nuclear Proteins/chemistry , Protein Binding/radiation effects , Protein Structure, Tertiary , Protein Transport/radiation effects , Repressor Proteins/chemistry , Ultraviolet Rays
3.
Mol Biol Cell ; 22(9): 1539-49, 2011 May.
Article in English | MEDLINE | ID: mdl-21346193

ABSTRACT

Many types of progenitor cells are distinguished by the expression of the intermediate filament protein nestin, a frequently used stem cell marker, the physiological roles of which are still unknown. Whereas myogenesis is characterized by dynamically regulated nestin levels, we studied how altering nestin levels affects myoblast differentiation. Nestin determined both the onset and pace of differentiation. Whereas depletion of nestin by RNAi strikingly accelerated the process, overexpression of nestin completely inhibited differentiation. Nestin down-regulation augmented the early stages of differentiation, at the level of cell-cycle withdrawal and expression of myogenic markers, but did not affect proliferation of undifferentiated dividing myoblasts. Nestin regulated the cleavage of the Cdk5 activator protein p35 to its degradation-resistant form, p25. In this way, nestin has the capacity to halt myoblast differentiation by inhibiting sustained activation of Cdk5 by p25, which is critical for the progress of differentiation. Our results imply that nestin regulates the early stages of myogenesis rather than maintains the undifferentiated state of progenitor cells. In the bidirectional interrelationship between nestin and Cdk5, Cdk5 regulates the organization and stability of its own nestin scaffold, which in turn controls the effects of Cdk5. This nestin-Cdk5 cross-talk sets the pace of muscle differentiation.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase 5/metabolism , Intermediate Filament Proteins/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Nerve Tissue Proteins/metabolism , Stem Cells/metabolism , Animals , Cell Cycle , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase 5/genetics , Humans , Intermediate Filament Proteins/genetics , Mice , Muscle Development/genetics , Nerve Tissue Proteins/genetics , Nestin , Phosphotransferases/metabolism , Polymerase Chain Reaction , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics , Stem Cells/cytology
4.
Mol Biol Cell ; 21(8): 1423-34, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20200223

ABSTRACT

Atypical protein kinase Czeta (PKCzeta) is emerging as a mediator of differentiation. Here, we describe a novel role for PKCzeta in myogenic differentiation, demonstrating that PKCzeta activity is indispensable for differentiation of both C2C12 and mouse primary myoblasts. PKCzeta was found to be associated with and to regulate the Cdk5/p35 signaling complex, an essential factor for both neuronal and myogenic differentiation. Inhibition of PKCzeta activity prevented both myotube formation and simultaneous reorganization of the nestin intermediate filament cytoskeleton, which is known to be regulated by Cdk5 during myogenesis. p35, the Cdk5 activator, was shown to be a specific phosphorylation target of PKCzeta. PKCzeta-mediated phosphorylation of Ser-33 on p35 promoted calpain-mediated cleavage of p35 to its more active and stable fragment, p25. Strikingly, both calpain activation and the calpain-mediated cleavage of p35 were shown to be PKCzeta-dependent in differentiating myoblasts. Overall, our results identify PKCzeta as a controller of myogenic differentiation by its regulation of the phosphorylation-dependent and calpain-mediated p35 cleavage, which is crucial for the amplification of the Cdk5 activity that is required during differentiation.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Muscle Development , Protein Kinase C/metabolism , Signal Transduction , Animals , COS Cells , Calpain/metabolism , Cell Differentiation/drug effects , Chlorocebus aethiops , Intermediate Filament Proteins/metabolism , Mice , Models, Biological , Muscle Development/drug effects , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/enzymology , Nerve Tissue Proteins/metabolism , Nestin , Phosphorylation/drug effects , Phosphotransferases/metabolism , Protein Binding/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects
5.
Mol Cell Proteomics ; 6(8): 1380-91, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17510049

ABSTRACT

Recent advances in instrument control and enrichment procedures have enabled us to quantify large numbers of phosphoproteins and record site-specific phosphorylation events. An intriguing problem that has arisen with these advances is to accurately validate where phosphorylation events occur, if possible, in an automated manner. The problem is difficult because MS/MS spectra of phosphopeptides are generally more complicated than those of unmodified peptides. For large scale studies, the problem is even more evident because phosphorylation sites are based on single peptide identifications in contrast to protein identifications where at least two peptides from the same protein are required for identification. To address this problem we have developed an integrated strategy that increases the reliability and ease for phosphopeptide validation. We have developed an off-line titanium dioxide (TiO(2)) selective phosphopeptide enrichment procedure for crude cell lysates. Following enrichment, half of the phosphopeptide fractionated sample is enzymatically dephosphorylated, after which both samples are subjected to LC-MS/MS. From the resulting MS/MS analyses, the dephosphorylated peptide is used as a reference spectrum against the original phosphopeptide spectrum, in effect generating two peptide spectra for the same amino acid sequence, thereby enhancing the probability of a correct identification. The integrated procedure is summarized as follows: 1) enrichment for phosphopeptides by TiO(2) chromatography, 2) dephosphorylation of half the sample, 3) LC-MS/MS-based analysis of phosphopeptides and corresponding dephosphorylated peptides, 4) comparison of peptide elution profiles before and after dephosphorylation to confirm phosphorylation, and 5) comparison of MS/MS spectra before and after dephosphorylation to validate the phosphopeptide and its phosphorylation site. This phosphopeptide identification represents a major improvement as compared with identifications based only on single MS/MS spectra and probability-based database searches. We investigated an applicability of this method to crude cell lysates and demonstrate its application on the large scale analysis of phosphorylation sites in differentiating mouse myoblast cells.


Subject(s)
Phosphopeptides/analysis , Amino Acid Sequence , Chromatography, Liquid/methods , Molecular Sequence Data , Phosphorylation , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Titanium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...