Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2356-2368, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28888991

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2), a multi-domain protein, is a key causative factor in Parkinson's disease (PD). Identification of novel substrates and the molecular mechanisms underlying the effects of LRRK2 are essential for understanding the pathogenesis of PD. In this study, we showed that LRRK2 played an important role in neuronal cell death by directly phosphorylating and activating apoptosis signal-regulating kinase 1 (ASK1). LRRK2 phosphorylated ASK1 at Thr832 that is adjacent to Thr845, which serves as an autophosphorylation site. Moreover, results of binding and kinase assays showed that LRRK2 acted as a scaffolding protein by interacting with each components of the ASK1-MKK3/6-p38 MAPK pathway through its specific domains and increasing the proximity to downstream targets. Furthermore, LRRK2-induced apoptosis was suppressed by ASK1 inhibition in neuronal stem cells derived from patients with PD. These results clearly indicate that LRRK2 acts as an upstream kinase in the ASK1 pathway and plays an important role in the pathogenesis of PD.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , MAP Kinase Kinase Kinase 5/genetics , Neurons/metabolism , Parkinson Disease/genetics , Apoptosis/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Neurons/pathology , Parkinson Disease/pathology , Phosphorylation , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases/genetics
2.
Neuroreport ; 28(4): 181-186, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28169962

ABSTRACT

Parkin, an E3 ubiquitin ligase, is the most frequently mutated gene in hereditary Parkinson's disease. Inactivation of Parkin leads to impairment of the ubiquitin-proteasome system, resulting in the accumulation of misfolded or aggregated proteins and ensuing neurodegeneration. In this study, we show that Parkin positively regulates the Notch1 signaling pathway. Overexpression of Parkin stabilized Notch1-IC protein levels, whereas knockdown of Parkin decreased Notch1-IC protein stability. Notably, overexpression of Parkin disrupted oxidative stress-induced apoptosis in neuronal cells. However, knockdown of Notch1 inhibited Parkin-induced neuronal cell survival. Together, these results indicate that Parkin is a novel regulator of the Notch1 signaling pathway, which promotes neuronal cell survival.

3.
Oncotarget ; 7(48): 79047-79063, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27806347

ABSTRACT

Autophagy is a highly conserved mechanism that degrades long-lived proteins and dysfunctional organelles, and contributes to cell fate. In this study, autophagy attenuates Notch1 signaling by degrading the Notch1 intracellular domain (Notch1-IC). Nutrient-deprivation promotes Notch1-IC phosphorylation by MEKK1 and phosphorylated Notch1-IC is recognized by Fbw7 E3 ligase. The ubiquitination of Notch1-IC by Fbw7 is essential for the interaction between Notch1-IC and p62 and for the formation of aggregates. Inhibition of Notch1 signaling prevents the transformation of breast cancer cells, tumor progression, and metastasis. The expression of Notch1 and p62 is inversely correlated with Beclin1 expression in human breast cancer patients. These results show that autophagy inhibits Notch1 signaling by promoting Notch1-IC degradation and therefore plays a role in tumor suppression.


Subject(s)
Beclin-1/metabolism , Breast Neoplasms/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism , MAP Kinase Kinase Kinase 1/metabolism , RNA-Binding Proteins/metabolism , Receptor, Notch1/chemistry , Receptor, Notch1/metabolism , Autophagy , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , HEK293 Cells , Humans , Neoplasm Metastasis , Phosphorylation , Signal Transduction
4.
Cancer Res ; 76(16): 4728-40, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27335110

ABSTRACT

The receptor Notch1 plays an important role in malignant progression of many cancers, but its regulation is not fully understood. In this study, we report that the kinase HIPK2 is responsible for facilitating the Fbw7-dependent proteasomal degradation of Notch1 by phosphorylating its intracellular domain (Notch1-IC) within the Cdc4 phosphodegron motif. Notch1-IC expression was higher in cancer cells than normal cells. Under genotoxic stress, Notch1-IC was phosphorylated constitutively by HIPK2 and was maintained at a low level through proteasomal degradation. HIPK2 phosphorylated the residue T2512 in Notch1-IC. Somatic mutations near this residue rendered Notch1-IC resistant to degradation, as induced either by HIPK2 overexpression or adriamycin treatment. In revealing an important mechanism of Notch1 stability, the results of this study could offer a therapeutic strategy to block Notch1-dependent progression in many types of cancer. Cancer Res; 76(16); 4728-40. ©2016 AACR.


Subject(s)
Breast Neoplasms/pathology , Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Notch1/metabolism , Animals , Blotting, Western , Breast Neoplasms/metabolism , Cell Proliferation/physiology , Cell Survival/physiology , Female , Fluorescent Antibody Technique , Heterografts , Humans , Immunoblotting , Immunoprecipitation , Mice , Mutation , Neoplasm Invasiveness/pathology , Phosphorylation , Polymerase Chain Reaction , Protein Stability , Receptor, Notch1/genetics
5.
Biochim Biophys Acta ; 1863(2): 179-88, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26547035

ABSTRACT

p21-Activated kinase 1 (PAK1) is a serine/threonine protein kinase implicated in cytoskeletal remodeling and cell motility. Recent studies have shown that it also promotes cell proliferation, regulates apoptosis, and increases cell transformation and invasion. In this study, we showed that NOTCH1 intracellular domain (NOTCH1-IC) negatively regulated PAK1 signaling pathway. We found a novel interaction between NOTCH1-IC and PAK1. Overexpression of NOTCH1-IC decreased PAK1-induced integrin-linked kinase 1 (ILK1) phosphorylation, whereas inhibition of NOTCH1 signaling increased PAK1-induced ILK1 phosphorylation. Notably, ILK1 phosphorylation was higher in PS1,2(-/-) cells than in PS1,2(+/+) cells. As expected, overexpression of NOTCH1-IC decreased ILK1-induced phosphorylation of glycogen synthase kinase 3 beta (GSK-3beta). Furthermore, NOTCH1-IC disrupted the interaction of PAK1 with ILK1 and altered PAK1 localization by directly interacting with it. This inhibitory effect of NOTCH1-IC on the PAK1 signaling pathway was mediated by the binding of NOTCH1-IC to PAK1 and by the alteration of PAK1 localization. Together, these results suggest that NOTCH1-IC is a new regulator of the PAK1 signaling pathway that directly interacts with PAK1 and regulates its shuttling between the nucleus and the cytoplasm.


Subject(s)
Receptor, Notch1/metabolism , Signal Transduction , p21-Activated Kinases/metabolism , Binding Sites/genetics , Cell Movement , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Microscopy, Confocal , Models, Biological , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptor, Notch1/genetics , p21-Activated Kinases/genetics
6.
Biochim Biophys Acta ; 1853(11 Pt A): 2918-28, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26276215

ABSTRACT

Fe65 is a highly conserved adaptor protein that interacts with several binding partners. Fe65 binds proteins to mediate various cellular processes. But the interacting partner and the regulatory mechanisms controlled by Fe65 are largely unknown. In this study, we found that Fe65 interacts with the C-terminus of Jagged1. Furthermore, Fe65 negatively regulates AP1-mediated Jagged1 intercellular domain transactivation in a Tip60-independent manner. We found that Fe65 triggers the degradation of Jagged1, but not the Jagged1 intracellular domain (JICD), through both proteasome and lysosome pathways. We also showed that Fe65 promotes recruitment of the E3 ligase Neuralized-like 1 (Neurl1) to membrane-tethered Jagged1 and monoubiquitination of Jagged1. These three proteins form a stable trimeric complex, thereby decreasing Jagged1 targeting by ubiquitin-mediated degradation. Consequently, Jagged1 is a novel binding partner of Fe65, and Fe65 may act as a novel effector of Jagged1 signaling.


Subject(s)
Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Calcium-Binding Proteins/genetics , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Lysosomes/genetics , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Serrate-Jagged Proteins , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/physiology
7.
Neurosci Lett ; 600: 6-11, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26033182

ABSTRACT

Notch signaling pathway is well known that it is involved in regulating cell fate, proliferation and homeostasis. In this study, we show a novel function of alpha-synuclein (SNCA) to promote degradation of Notch1 intracellular domain (Notch1-IC) through Fbw7, ubiquitin E3 ligase. We identified that SNCA inhibits Notch1 transcription activity and diminishes the interaction between Notch1-IC and RBP-Jk. We also found decrease of Notch1-IC protein stability by exogenous and endogenous SNCA through proteasomal pathway, not through lysosomal pathway. And, we found that SNCA promotes interaction between Notch1-IC and Fbw7. Furthermore, SNCA directly interacts with Fbw7. SNCA increases ubiquitination of Notch-IC by Fbw7 through interaction with Fbw7. Together, these results suggest that SNCA is a novel regulator of Notch1-IC transcriptional activity with acting as an enhancer of the interaction of Notch1-IC and Fbw7 with increasing degradation of Notch1-IC.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Receptor, Notch1/metabolism , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/metabolism , F-Box-WD Repeat-Containing Protein 7 , HEK293 Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Signal Transduction , Transcription, Genetic , Ubiquitination
8.
J Neurochem ; 134(5): 799-810, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25996556

ABSTRACT

The gamma-secretase is a multiprotein complex that cleaves many type-I membrane proteins, such as the Notch receptor and the amyloid precursor protein. Nicastrin (NCT) is an essential component of the multimeric gamma-secretase complex and functions as a receptor for gamma-secretase substrates. In this study, we found that Akt1 markedly regulated the protein stability of NCT. Importantly, the kinase activity of Akt1 was essential for the inhibition of gamma-secretase activity through degradation of NCT. Notably, the protein level of endogenous NCT was higher in shAkt1-expressing cells than in shCon-expressing cells. Akt1 physically interacted with NCT and mediated its degradation through proteasomal and lysosomal pathways. We also found that Akt1 phosphorylates NCT at Ser437, resulting in a significant reduction in NCT protein stability. Importantly, a phospho-deficient mutation in NCT at Ser437 stabilized its protein levels. Collectively, our results reveal that Akt1 functions as a negative regulator of the gamma-secretase activity through phosphorylation and degradation of NCT. Generation of the amyloid peptide (A-beta) and the amyloid precursor protein (APP) intracellular domain (AICD) can happen by sequential proteolysis of APP by beta and gamma-secretase. The gamma-secretase complex consists of four essential proteins: presenilin (PS1 or PS2), presenilin enhancer 2 (PEN-2), anterior pharynx-defective 1 (APH-1), and the Nicastrin (NCT). NCT can interact and be phosphorylated by Akt1, and phosphorylated NCT promotes its proteasomal and lysosomal degradation. As a result, Akt1 plays role in reducing gamma-secretase activity through phosphorylation-dependent regulation of NCT protein degradation.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins c-akt/physiology , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Lysosomes/metabolism , Membrane Glycoproteins/genetics , Models, Biological , Phosphorylation , Phosphoserine/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 110(17): 6865-70, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23569274

ABSTRACT

Notch1 genes encode receptors for a signaling pathway that regulates various aspects of cell growth and differentiation; however, the role of Notch1 signaling in p38 mitogen-activated protein kinase (MAPK) signaling pathway is still not well defined. In this study, we found that Notch1 intracellular domain (Notch1-IC) prevents oxidative stress-induced cell death through the suppression of the Apoptosis signal-regulating kinase (ASK) 1 signaling pathway. Notch1-IC inhibited H2O2-induced activation of ASK1 and the activation of downstream kinases in the p38 MAPK signaling cascade. The results of both in vivo binding and kinase studies have revealed that ASK1 is the direct target of Notch1-IC, whereas it produced no effect on either MAP kinase kinase (MKK) 3 or p38 MAPK. Notch1-IC blocked both the homooligomerization of ASK1 and inhibited ASK1 activity. Furthermore, Notch1-IC facilitated the translocation of activated ASK1 toward the nucleus. Notch1 knockdown was determined to be highly susceptible to oxidative stress-induced activation of ASK1-MKK3/MKK6-p38 MAPK signaling cascade and cell death. Taken together, our findings suggest that Notch1-IC may act as a negative regulator in ASK1 signaling cascades.


Subject(s)
Cell Death/physiology , MAP Kinase Kinase Kinase 5/metabolism , Oxidative Stress/physiology , Receptor, Notch1/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Fractionation , Cell Line , DNA Primers/genetics , Escherichia coli , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Luciferases , Mice , Models, Biological , Mutagenesis, Site-Directed , Protein Binding , Receptor, Notch1/physiology , Signal Transduction/genetics
10.
Biochim Biophys Acta ; 1833(1): 69-79, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23103515

ABSTRACT

The Notch signaling pathway plays a crucial role in the regulation of cell fate decision, and is also a key regulator of cell differentiation, including bone homeostasis, in a variety of contexts. However, the role of Notch1 signaling in osteoclast differentiation is still controversial. In this study, we show that Receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation is promoted by the Notch1 intracellular domain (Notch1-IC) and Ca(2+)/Calmodulin dependent protein kinase IV (CaMKIV) signaling. Notch1-IC protein level was augmented by CaMKIV through escape from ubiquitin dependent protein degradation. In addition, CaMKIV remarkably increased Notch1-IC stability, and the kinase activity of CaMKIV was essential for facilitating Notch1 signaling. CaMKIV directly interacted with Notch1-IC and phosphorylates Notch1-IC, thereby decreasing proteasomal protein degradation through F-box and WD repeat domain-containing 7 (Fbw7). We also found that Notch1-IC prevented inhibition of osteoclast differentiation by KN-93 but not the phosphorylation deficient form of Notch1-IC. These results suggest that phosphorylated Notch1-IC by CaMKIV increases Notch1-IC stability, which enhances osteoclast differentiation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/physiology , Cell Differentiation/genetics , Osteoclasts/physiology , Receptor, Notch1/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cell Differentiation/drug effects , Cells, Cultured , HEK293 Cells , Humans , Mice , Models, Biological , Osteoclasts/drug effects , Osteoclasts/metabolism , Protein Binding , Protein Interaction Domains and Motifs/physiology , Protein Stability/drug effects , RANK Ligand/pharmacology , Receptor, Notch1/chemistry , Receptor, Notch1/physiology , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
11.
J Biol Chem ; 287(44): 36814-29, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22888005

ABSTRACT

Serine-threonine Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is the key component in noncanonical Wnt5a signaling and has been shown to regulate its signaling. In this study, we found that CaMKII induced by Wnt5a remarkably reduced the protein stability of the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT), a co-repressor of Notch signaling, through proteasomal degradation. Wnt5a was found to enhance Notch1 intracellular domain (Notch1-IC) transcription activity, which could be inhibited by treatment with KN93, a CaMKII inhibitor. The kinase activity of CaMKII was essential for the activation of Notch signaling. We also determined that CaMKII could enhance the association between Notch1-IC and RBP-Jk. Furthermore, the physical association between RBP-Jk and SMRT was substantially suppressed by CaMKII. We demonstrated that CaMKII directly bound and phosphorylated SMRT at Ser-1407, thereby facilitating SMRT translocation from the nucleus to the cytoplasm and proteasome-dependent degradation. These results suggest that CaMKII down-regulated the protein stability of SMRT through proteasomal degradation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Proto-Oncogene Proteins/physiology , Receptor, Notch1/metabolism , Wnt Proteins/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , Genes, Reporter , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Luciferases/biosynthesis , Luciferases/genetics , Mice , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proteolysis , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factor HES-1 , Transcriptional Activation , Ubiquitination , Wnt Proteins/metabolism , Wnt Signaling Pathway , Wnt-5a Protein
12.
PLoS One ; 7(5): e37111, 2012.
Article in English | MEDLINE | ID: mdl-22590650

ABSTRACT

The gamma-secretase complex is involved in the intramembranous proteolysis of a variety of substrates, including the amyloid precursor protein and the Notch receptor. Nicastrin (NCT) is an essential component of the gamma-secretase complex and functions as a receptor for gamma-secretase substrates. In this study, we determined that serum- and glucocorticoid-induced protein kinase 1 (SGK1) markedly reduced the protein stability of NCT. The SGK1 kinase activity was decisive for NCT degradation and endogenous SGK1 inhibited gamma-secretase activity. SGK1 downregulates NCT protein levels via proteasomal and lysosomal pathways. Furthermore, SGK1 directly bound to and phosphorylated NCT on Ser437, thereby promoting protein degradation. Collectively, our findings indicate that SGK1 is a gamma-secretase regulator presumably effective through phosphorylation and degradation of NCT.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Immediate-Early Proteins/metabolism , Lysosomes/metabolism , Membrane Glycoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Amyloid Precursor Protein Secretases/genetics , Animals , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Lysosomes/genetics , Membrane Glycoproteins/genetics , Mice , Phosphorylation/physiology , Proteasome Endopeptidase Complex/genetics , Protein Serine-Threonine Kinases/genetics , Protein Stability
13.
J Cell Sci ; 125(Pt 5): 1296-308, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22302987

ABSTRACT

Transcriptional regulation performs a central role in Notch1 signaling by recombining binding protein Suppressor of Hairless (RBP-Jk)--a signaling pathway that is widely involved in determination of cell fate. Our earlier work demonstrated the possible regulation of the Notch1-RBP-Jk pathway through protein degradation of RBP-Jk; however, the potential regulator for the degradation of RBP-Jk remains to be determined. Here, we report that the expression of endogenous and exogenous RBP-Jk was increased significantly in cells treated with proteasome- and lysosome-specific inhibitors. The effects of these inhibitors on RBP-Jk occurred in a dose- and time-dependent manner. The level of RBP-Jk protein was higher in presenilin-2 (PS2)-knockout cells than in presenilin-1 (PS1)-knockout cells. Furthermore, the level of RBP-Jk was decreased by expression of PS2 in PS1 and PS2 double-knockout cells. We also found that PS1-knockout cells treated with a specific inhibitor of p38 mitogen-activated protein kinase ∂ (MAPK) had significantly increased levels of RBP-Jk. p38 MAPK phosphorylates RBP-Jk at Thr339 by physical binding, which subsequently induces the degradation and ubiquitylation of the RBP-Jk protein. Collectively, our results indicate that PS2 modulates the degradation of RBP-Jk through phosphorylation by p38 MAPK.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Presenilin-2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Ammonium Chloride/pharmacology , Cell Line , Chloroquine/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Gene Knockout Techniques , HEK293 Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/biosynthesis , Leupeptins/pharmacology , Lysosomes/metabolism , MAP Kinase Signaling System , Phosphorylation , Presenilin-1/genetics , Presenilin-2/genetics , Proteasome Inhibitors , Proteolysis , Receptor, Notch1/metabolism , Transcription, Genetic , Transcriptional Activation , Ubiquitination , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
14.
J Biol Chem ; 287(7): 4690-701, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22199353

ABSTRACT

Notch1 receptor functions as a critical controller of cell fate decisions and also as a key regulator of cell growth, differentiation, and proliferation in invertebrates and vertebrates. In this study, we have demonstrated that the adaptor protein Fe65 attenuates Notch1 signaling via the accelerated degradation of the membrane-tethered Notch1 in the cytoplasm. Fe65 also suppresses Notch1 transcriptional activity via the dissociation of the Notch1-IC-recombining binding protein suppressor of hairless (RBP)-Jk complex within the nucleus. Fe65 is capable of forming a trimeric complex with Itch and membrane-tethered Notch1, and Fe65 enhances the protein degradation of membrane-tethered Notch1 via an Itch-dependent proteasomal pathway. Collectively, our results demonstrate that Fe65 carries out different functions depending on its location in the regulation of Notch1 signaling.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Proteolysis , Receptor, Notch1/metabolism , Signal Transduction/physiology , Animals , HEK293 Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , NIH 3T3 Cells , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
15.
Exp Cell Res ; 317(17): 2438-46, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21820430

ABSTRACT

Notch signaling involves the proteolytic cleavage of the transmembrane Notch receptor after binding to its transmembrane ligands. Jagged-1 also undergoes proteolytic cleavage by gamma-secretase and releases an intracellular fragment. In this study, we have demonstrated that the Jagged-1 intracellular domain (JICD) inhibits Notch1 signaling via a reduction in the protein stability of the Notch1 intracellular domain (Notch1-IC). The formation of the Notch1-IC-RBP-Jk-Mastermind complex is prevented in the presence of JICD, via a physical interaction. Furthermore, JICD accelerates the protein degradation of Notch1-IC via Fbw7-dependent proteasomal pathway. These results indicate that JICD functions as a negative regulator in Notch1 signaling via the promotion of Notch1-IC degradation.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptor, Notch1/chemistry , Receptor, Notch1/metabolism , Ubiquitin-Protein Ligases/metabolism , Cells, Cultured , Down-Regulation , F-Box-WD Repeat-Containing Protein 7 , HEK293 Cells , Humans , Jagged-1 Protein , Protein Structure, Tertiary , Serrate-Jagged Proteins , Signal Transduction
16.
Mol Cells ; 32(2): 161-5, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21643850

ABSTRACT

Notch signaling involves the proteolytic cleavage of the transmembrane Notch receptor after binding to its transmembrane ligands. The Delta-like ligand 1 also undergoes proteolytic cleavage upon Notch binding, resulting in the production of a free intracellular domain. In this study, we have demonstrated that the Delta-like 1 intracellular domain (Dll1-IC) specifically binds to Notch1-IC in the nucleus, thereby disrupting the association of the Notch1-IC-RBP-Jk-MAM transcription activator complex. Additionally, the Notch1-mediated blockage of the induction of MyoD is abolished by the co-expression of Dll1-IC. Collectively, our results show that Dll1-IC functions as a negative regulator in Notch signaling via the disruption of the Notch1-IC-RBP-Jk complex.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , MyoD Protein/metabolism , Receptor, Notch1/metabolism , Animals , Calcium-Binding Proteins , Cell Differentiation/genetics , HEK293 Cells , Humans , Mice , MyoD Protein/genetics , Protein Binding/genetics , Protein Engineering , Protein Structure, Tertiary/genetics , Receptor, Notch1/genetics , Sequence Deletion/genetics , Signal Transduction/genetics , Transcriptional Activation , Transgenes/genetics
17.
J Cell Sci ; 124(Pt 11): 1831-43, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21558417

ABSTRACT

The Notch1 receptor is a crucial controller of cell fate decisions, and is also a key regulator of cell growth and differentiation in a variety of contexts. In this study, we have demonstrated that the APP intracellular domain (AICD) attenuates Notch1 signaling by accelerated degradation of the Notch1 intracellular domain (Notch1-IC) and RBP-Jk, through different degradation pathways. AICD suppresses Notch1 transcriptional activity by the dissociation of the Notch1-IC-RBP-Jk complex after processing by γ-secretase. Notch1-IC is capable of forming a trimeric complex with Fbw7 and AICD, and AICD enhances the protein degradation of Notch1-IC through an Fbw7-dependent proteasomal pathway. AICD downregulates the levels of RBP-Jk protein through the lysosomal pathway. AICD-mediated degradation is involved in the preferential degradation of non-phosphorylated RBP-Jk. Collectively, our results demonstrate that AICD functions as a negative regulator in Notch1 signaling through the promotion of Notch1-IC and RBP-Jk protein degradation.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Ammonium Chloride/pharmacology , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Line , Down-Regulation/genetics , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Genes, Reporter , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Leupeptins/pharmacology , Luciferases/genetics , Luciferases/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mitogen-Activated Protein Kinase 10/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Multimerization , Protein Stability , Protein Structure, Tertiary , Receptor, Notch1/antagonists & inhibitors , Transcriptional Activation , Ubiquitin-Protein Ligases/metabolism
18.
J Bone Miner Res ; 26(2): 317-30, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20740684

ABSTRACT

Notch1 genes encode receptors for a signaling pathway that regulates cell growth and differentiation in various contexts, but the role of Notch1 signaling in osteogenesis is not well defined. Notch1 controls osteoblast differentiation by affecting Runx2, but the question arises whether normal osteoblastic differentiation can occur regardless of the presence of Notch1. In this study, we observed the downregulation of Notch1 signaling during osteoblastic differentiation. BMPR-IB/Alk6-induced Runx2 proteins reduced Notch1 activity to a marked degree. Accumulated Runx2 suppressed Notch1 transcriptional activity by dissociating the Notch1-IC-RBP-Jk complex. Using deletion mutants, we also determined that the N-terminal domain of Runx2 was crucial to the binding and inhibition of the N-terminus of the Notch1 intracellular domain. Notably, upregulation of the Runx2 protein level paralleled reduced expression of Hes1, which is a downstream target of Notch1, during osteoblast differentiation. Collectively, our data suggest that Runx2 is an inhibitor of the Notch1 signaling pathway during normal osteoblast differentiation.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation , Osteoblasts/cytology , Receptor, Notch1/antagonists & inhibitors , 3T3 Cells , Animals , Animals, Newborn , Cell Differentiation , Humans , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Receptor, Notch1/metabolism , Signal Transduction
19.
J Cell Sci ; 124(Pt 1): 100-12, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21147854

ABSTRACT

Notch is a transmembrane protein that acts as a transcriptional factor in the Notch signaling pathway for cell survival, cell death and cell differentiation. Notch1 and Fbw7 mutations both lead the activation of the Notch1 pathway and are found in the majority of patients with the leukemia T-ALL. However, little is known about the mechanisms and regulators that are responsible for attenuating the Notch signaling pathway through Fbw7. Here, we report that the serum- and glucocorticoid-inducible protein kinase SGK1 remarkably reduced the protein stability of the active form of Notch1 through Fbw7. The protein level and transcriptional activity of the Notch1 intracellular domain (Notch1-IC) were higher in SGK1-deficient cells than in SGK1 wild-type cells. Notch1-IC was able to form a trimeric complex with Fbw7 and SGK1, thereby SGK1 enhanced the protein degradation of Notch1-IC via a Fbw7-dependent proteasomal pathway. Furthermore, activated SGK1 phosphorylated Fbw7 at serine 227, an effect inducing Notch1-IC protein degradation and ubiquitylation. Moreover, accumulated dexamethasone-induced SGK1 facilitated the degradation of Notch1-IC through phosphorylation of Fbw7. Together our results suggest that SGK1 inhibits the Notch1 signaling pathway via phosphorylation of Fbw7.


Subject(s)
Cell Cycle Proteins/metabolism , Down-Regulation , F-Box Proteins/metabolism , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Notch1/chemistry , Receptor, Notch1/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Glucocorticoids/metabolism , Humans , Immediate-Early Proteins/genetics , Mice , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Stability , Receptor, Notch1/genetics , Ubiquitin-Protein Ligases/genetics
20.
J Cell Biochem ; 110(1): 229-37, 2010 May.
Article in English | MEDLINE | ID: mdl-20213747

ABSTRACT

DJ-1 has been reported as a gene linked to early onset familial Parkinson's disease, and is functionally involved in transcriptional regulation and oxidative stress-induced cell death. To understand the role of DJ-1 in cellular stress, this study investigated DJ-1's effect on stress-activated protein kinase signaling and H(2)O(2)-induced activation of apoptosis signal-regulating kinase 1 (ASK1). According to the results, the overexpression of DJ-1 inhibited H(2)O(2)-induced activation of ASK1 as well as the activation of downstream kinases in the p38 mitogen-activated protein kinase (MAPK) signaling cascade. The results of both in vivo binding and kinase studies have revealed that ASK1 is the direct target of DJ-1, whereas it has shown no effect on either MKK3 or p38. DJ-1 blocked both the homo-oligomerization of ASK1 and inhibited ASK1 activity. Taken together, our data strongly suggest that DJ-1, by directly inhibiting ASK1, may act as a negative regulator in ASK1 signaling cascades.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Oncogene Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line , Enzyme Activation , Humans , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Models, Biological , Oxidative Stress , Protein Binding , Protein Deglycase DJ-1 , Protein Multimerization
SELECTION OF CITATIONS
SEARCH DETAIL
...