Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1871(4): 119690, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38367915

ABSTRACT

The scaffold protein 14-3-3ζ is an established regulator of adipogenesis and postnatal adiposity. We and others have demonstrated the 14-3-3ζ interactome to be diverse and dynamic, and it can be examined to identify novel regulators of physiological processes, including adipogenesis. In the present study, we sought to determine if factors that influence adipogenesis during the development of obesity could be identified in the 14-3-3ζ interactome found in white adipose tissue of lean or obese TAP-tagged-14-3-3ζ overexpressing mice. Using mass spectrometry, differences in the abundance of novel, as well as established, adipogenic factors within the 14-3-3ζ interactome could be detected in adipose tissues. One novel candidate was revealed to be plakoglobin, the homolog of the known adipogenic inhibitor, ß-catenin, and herein, we report that plakoglobin is involved in adipocyte differentiation. Plakoglobin is expressed in murine 3T3-L1 cells and is primarily localized to the nucleus, where its abundance decreases during adipogenesis. Depletion of plakoglobin by siRNA inhibited adipogenesis and reduced PPARγ2 expression, and similarly, plakoglobin depletion in human adipose-derived stem cells also impaired adipogenesis and reduced lipid accumulation post-differentiation. Transcriptional assays indicated that plakoglobin does not participate in Wnt/ß-catenin signaling, as its depletion did not affect Wnt3a-mediated transcriptional activity. Taken together, our results establish plakoglobin as a novel regulator of adipogenesis in vitro and highlights the ability of using the 14-3-3ζ interactome to identify potential pro-obesogenic factors.


Subject(s)
14-3-3 Proteins , Adipocytes , gamma Catenin , Animals , Humans , Mice , 14-3-3 Proteins/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , beta Catenin/genetics , beta Catenin/metabolism , gamma Catenin/genetics , gamma Catenin/metabolism , Obesity/metabolism , Wnt Signaling Pathway
2.
Oncogene ; 33(13): 1690-9, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-23584478

ABSTRACT

Protein synthesis has a key role in the control of cell proliferation, and its deregulation is associated with pathological conditions, notably cancer. Rapamycin, an inhibitor of mammalian target of rapamycin complex 1 (mTORC1), was known to inhibit protein synthesis. However, it does not substantially inhibit protein synthesis and cell proliferation in many cancer types. We were interested in finding a novel target in rapamycin-resistant cancer. The rate-limiting factor for translation is eukaryotic translation initiation factor 4E (eIF4E), which is negatively regulated by eIF4E-binding protein 1 (4E-BP1). Here, we provide evidence that glycogen synthase kinase (GSK)-3ß promotes cell proliferation through positive regulation of protein synthesis. We found that GSK-3ß phosphorylates and inactivates 4E-BP1, thereby increasing eIF4E-dependent protein synthesis. Considering the clinical relevance of pathways regulating protein synthesis, our study provides a promising new strategy and target for cancer therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Glycogen Synthase Kinase 3/metabolism , Phosphoproteins/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Cell Cycle Proteins , Cell Growth Processes/physiology , Enzyme Inhibitors/pharmacology , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Nude , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Phosphoproteins/biosynthesis , Phosphorylation , Protein Binding , Protein Biosynthesis , Random Allocation , Thiazoles/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Urea/analogs & derivatives , Urea/pharmacology , Xenograft Model Antitumor Assays
3.
Oncogene ; 32(38): 4480-9, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23108403

ABSTRACT

The incidence of malignant melanoma is growing rapidly worldwide and there is still no effective therapy for metastatic disease. This type of cancer is highly resistant to conventional DNA-damaging chemotherapeutics, and intense research has been dedicated for understanding the molecular pathways underlying chemoresistance. The Ras/mitogen-activated protein kinase (MAPK) signalling pathway is often deregulated in melanoma, which frequently harbours activating mutations in NRAS or BRAF. Herein, we demonstrate that the MAPK-activated protein kinase RSK (p90 ribosomal S6 kinase) contributes to melanoma chemoresistance by altering their response to chemotherapeutic agents. We find that RSK phosphorylates checkpoint kinase 1 (Chk1) at an inhibitory site, Ser280, both in vitro and in vivo. Our results indicate that RSK is the predominant protein kinase operating downstream of mitogens and oncogenes of the Ras/MAPK pathway, and consistent with this, we find that RSK constitutively phosphorylates Chk1 in melanoma. We show that RSK inhibition increases Chk1 activity in response to DNA-damaging agents, suggesting that the Ras/MAPK pathway modulates Chk1 function and the response to DNA damage. Accordingly, we demonstrate that RSK promotes G2 DNA damage checkpoint silencing in a Chk1-dependent manner, and find that RSK inhibitors sensitize melanoma cells to DNA-damaging agents. Together, our results identify a novel link between the Ras/MAPK pathway and the DNA damage response, and suggest that RSK inhibitors may be used to modulate chemosensitivity, which is one of the major obstacles to melanoma treatment.


Subject(s)
DNA Damage , Drug Resistance, Neoplasm/genetics , G2 Phase Cell Cycle Checkpoints , Gene Silencing , Melanoma/genetics , Melanoma/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Cell Line , Checkpoint Kinase 1 , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Signal Transduction
4.
J Biol Chem ; 276(25): 23097-104, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11312266

ABSTRACT

The Akt kinase plays a crucial role in supporting Trk-dependent cell survival, whereas the p75 neurotrophin receptor (p75NTR) facilitates cellular apoptosis. The precise mechanism that p75NTR uses to promote cell death is not certain, but one possibility is that p75NTR-dependent ceramide accumulation inhibits phosphatidylinositol 3-kinase-mediated Akt activation. To test this hypothesis, we developed a system for examining p75NTR-dependent apoptosis and determined the effect of p75NTR on Akt activation. Surprisingly, p75NTR increased, rather than decreased, Akt phosphorylation in a variety of cell types, including human Niemann-Pick fibroblasts, which lack acidic sphingomyelinase activity. The p75NTR expression level required to elicit Akt phosphorylation was much lower than that required to activate the JNK pathway or to mediate apoptosis. We show that p75NTR-dependent Akt phosphorylation was independent of TrkA signaling, required active phosphatidylinositol 3-kinase, and was associated with increased tyrosine phosphorylation of p85 and Shc and with reduced cytosolic tyrosine phosphatase activity. Finally, we show that p75NTR expression increased survival in cells exposed to staurosporine or subjected to serum withdrawal. These findings indicate that p75NTR facilitates cell survival through novel signaling cascades that result in Akt activation.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptors, Nerve Growth Factor/physiology , Adenoviridae/genetics , Animals , Apoptosis , COS Cells , Cell Survival/physiology , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , PC12 Cells , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt , Rats , Receptor, Nerve Growth Factor , Recombination, Genetic , Tyrosine/metabolism
5.
Neuron ; 27(2): 279-88, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10985348

ABSTRACT

The mechanisms employed by the p75 neurotrophin receptor (p75NTR) to mediate neurotrophin-dependent apoptosis are poorly defined. Two-hybrid analyses were used to identify proteins involved in p75NTR apoptotic signaling, and a p75NTR binding partner termed NRAGE (for neurotrophin receptor-interacting MAGE homolog) was identified. NRAGE binds p75NTR in vitro and in vivo, and NRAGE associates with the plasma membrane when NGF is bound to p75NTR. NRAGE blocks the physical association of p75NTR with TrkA, and, conversely, TrkA overexpression eliminates NRAGE-mediated NGF-dependent death, indicating that interactions of NRAGE or TrkA with p75NTR are functionally and physically exclusive. NRAGE overexpression facilitates cell cycle arrest and permits NGF-dependent apoptosis within sympathetic neuron precursors cells. Our results show that NRAGE contributes to p75NTR-dependent cell death and suggest novel functions for MAGE family proteins.


Subject(s)
Apoptosis/physiology , DNA-Binding Proteins/metabolism , Neoplasm Proteins/genetics , Nerve Growth Factor/metabolism , Receptor, Nerve Growth Factor/metabolism , Adrenergic Fibers/metabolism , Animals , Antigens, Neoplasm , Brain/embryology , Brain/metabolism , Cell Compartmentation , Cell Cycle/genetics , Cell Membrane/metabolism , Cloning, Molecular , Humans , Melanoma-Specific Antigens , Molecular Sequence Data , Organ Specificity , RNA, Messenger/biosynthesis , Rats , Receptor, trkA/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Signal Transduction/genetics , Spinal Cord/embryology , Spinal Cord/metabolism , Two-Hybrid System Techniques
6.
J Neurosci ; 19(16): 6887-96, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10436046

ABSTRACT

Seizure causes neuronal cell loss in both animal models and human epilepsy. To determine the contribution of apoptotic mechanisms to seizure-induced neuronal cell death, rat brains were examined for the occurrence of terminal deoxynucleotidyl transferase-mediated UTP nick end labeling (TUNEL)-positive nuclei after pilocarpine-induced seizure. Numerous TUNEL-positive cells were observed throughout the postseizure hippocampus, piriform cortex, and entorhinal cortex. Combined TUNEL/NeuN immunocytochemistry demonstrated that the vast majority of TUNEL-positive cells were neurons. To identify components of the signal transduction cascade promoting postseizure apoptosis, the expression of the p75 neurotrophin receptor (p75NTR) was examined. Seizure-induced increases in p75NTR protein and mRNA were detected in hippocampus, piriform cortex, and entorhinal cortex. Immunohistochemical double labeling revealed almost complete correspondence between TUNEL-positive and p75NTR-expressing cells, suggesting that seizure-induced neuronal loss within the CNS occurs through apoptotic signaling cascades involving p75NTR.


Subject(s)
Apoptosis/physiology , Neurons/metabolism , Pilocarpine/toxicity , Receptors, Nerve Growth Factor/biosynthesis , Seizures/metabolism , Animals , Entorhinal Cortex/drug effects , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Male , Neurons/pathology , Olfactory Pathways/drug effects , Olfactory Pathways/metabolism , Olfactory Pathways/pathology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor , Seizures/chemically induced , Seizures/pathology
7.
J Biol Chem ; 274(30): 21443-9, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10409708

ABSTRACT

The p75 neurotrophin receptor (p75NTR) has been linked to activation of the NF-kappaB transcriptional complex in oligodendrocytes, Schwann cells, and PCNA cells. In this report, tumor necrosis factor (TNF)- and neurotrophin-mediated NF (nuclear factor)-kappaB activation were compared in several cell lines. All cell types showed TNF-mediated activation of NF-kappaB, but direct neurotrophin-dependent activation of NF-kappaB was never observed under normal growth conditions. In PCNA cells, a modest nerve growth factor (NGF)-dependent induction of NF-kappaB was detected but only after cells were subjected to severe stress. Although NGF binding did not directly activate NF-kappaB under normal conditions, NGF consistently altered TNF-dependent NF-kappaB activation in each cell type examined, and extended exposure to NGF and TNF always increased NF-kappaB activation over that achieved with TNF alone. The increase in NF-kappaB activity mediated by NGF correlated with reduced levels of IkappaBalpha; NGF added alone had no effect on IkappaBalpha levels, but when added with TNF, NGF treatment significantly reduced IkappaBalpha levels. We propose that modulation of cytokine receptor signaling is a significant physiological function of the p75 neurotrophin receptor and that previous reports of direct NF-kappaB activation through p75NTR reflect this modulatory activity.


Subject(s)
NF-kappa B/metabolism , Receptors, Nerve Growth Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , HeLa Cells , Humans , Oligodendroglia/metabolism , Oligodendroglia/pathology , Receptor, Nerve Growth Factor , Schwann Cells/metabolism , Schwann Cells/pathology , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...