Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cell ; 162(4): 780-94, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26276632

ABSTRACT

The transcriptional co-activators YAP and TAZ are key regulators of organ size and tissue homeostasis, and their dysregulation contributes to human cancer. Here, we discover YAP/TAZ as bona fide downstream effectors of the alternative Wnt signaling pathway. Wnt5a/b and Wnt3a induce YAP/TAZ activation independent of canonical Wnt/ß-catenin signaling. Mechanistically, we delineate the "alternative Wnt-YAP/TAZ signaling axis" that consists of Wnt-FZD/ROR-Gα12/13-Rho GTPases-Lats1/2 to promote YAP/TAZ activation and TEAD-mediated transcription. YAP/TAZ mediate the biological functions of alternative Wnt signaling, including gene expression, osteogenic differentiation, cell migration, and antagonism of Wnt/ß-catenin signaling. Together, our work establishes YAP/TAZ as critical mediators of alternative Wnt signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Wnt Signaling Pathway , Animals , Cell Cycle Proteins , Cell Line , Frizzled Receptors/metabolism , Humans , Mice , Mice, Transgenic , Trans-Activators , Transcription Factors , YAP-Signaling Proteins , beta Catenin/metabolism
2.
Mol Cell ; 73(6): 1138-1149.e6, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30901564

ABSTRACT

The nuclear factor (NF)-κB pathway plays a central role in inflammatory and immune responses, with aberrant activation of NF-κB signaling being implicated in various human disorders. Here, we show that mammalian ste20-like kinase 1 (MST1) is a previously unrecognized component of the tumor necrosis factor α (TNFα) receptor 1 signaling complex (TNF-RSC) and attenuates TNFα-induced NF-κB signaling. Genetic ablation of MST1 in mouse embryonic fibroblasts and bone marrow-derived macrophages potentiated the TNFα-induced increase in IκB kinase (IKK) activity, as well as the expression of NF-κB target genes. TNFα induced the recruitment of MST1 to TNF-RSC and its interaction with HOIP, the catalytic component of the E3 ligase linear ubiquitin assembly complex (LUBAC). Furthermore, MST1 activated in response to TNFα stimulation mediates the phosphorylation of HOIP and thereby inhibited LUBAC-dependent linear ubiquitination of NEMO/IKKγ. Together, our findings suggest that MST1 negatively regulates TNFα-induced NF-κB signaling by targeting LUBAC.


Subject(s)
Fibroblasts/drug effects , Macrophages/drug effects , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases/metabolism , Animals , Fibroblasts/enzymology , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/enzymology , Mice, Inbred C57BL , Mice, Knockout , Multienzyme Complexes , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
Nucleic Acids Res ; 48(7): 3678-3691, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32123907

ABSTRACT

Genomic instability resulting from defective DNA damage responses or repair causes several abnormalities, including progressive cerebellar ataxia, for which the molecular mechanisms are not well understood. Here, we report a new murine model of cerebellar ataxia resulting from concomitant inactivation of POLB and ATM. POLB is one of key enzymes for the repair of damaged or chemically modified bases, including methylated cytosine, but selective inactivation of Polb during neurogenesis affects only a subpopulation of cortical interneurons despite the accumulation of DNA damage throughout the brain. However, dual inactivation of Polb and Atm resulted in ataxia without significant neuropathological defects in the cerebellum. ATM is a protein kinase that responds to DNA strand breaks, and mutations in ATM are responsible for Ataxia Telangiectasia, which is characterized by progressive cerebellar ataxia. In the cerebella of mice deficient for both Polb and Atm, the most downregulated gene was Itpr1, likely because of misregulated DNA methylation cycle. ITPR1 is known to mediate calcium homeostasis, and ITPR1 mutations result in genetic diseases with cerebellar ataxia. Our data suggest that dysregulation of ITPR1 in the cerebellum could be one of contributing factors to progressive ataxia observed in human genomic instability syndromes.


Subject(s)
Cerebellar Ataxia/genetics , Cerebellum/metabolism , DNA Methylation , DNA Polymerase beta/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Brain/embryology , Brain/pathology , Cerebellum/abnormalities , Cerebellum/pathology , Cytosine/metabolism , DNA Damage , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Knockout , Neurogenesis/genetics
4.
Biochem Biophys Res Commun ; 528(1): 154-159, 2020 07 12.
Article in English | MEDLINE | ID: mdl-32451085

ABSTRACT

Renal fibrosis is one of the characteristic features of chronic kidney disease (CKD). Fibrotic change not only impairs the filtration function of the kidney but is also recognized as a marker of end-stage renal disease (ESRD). The epithelial to mesenchymal transition (EMT) is known to play a role in embryonic development and organ formation, but it is getting much attention for its pathological role in the invasion and metastasis of carcinoma. Recently, it has also been reported that EMT plays a role in the formation of fibrosis during chronic inflammation. EMT contribute to the development of the fibrosis in CKD. Moreover, glomerular podocytes and tubular epithelial cells can also undergo mesenchymal transition in CKD. Hesperetin is a flavonoid present in citrus and is well known for its antioxidant and anti-inflammatory properties. In this study, we investigated the effects of hesperetin on the EMT-elicited podocytes. First, we generated an EMT model by treating transforming growth factor (TGF)-ß1, a potent inducer of EMT to the podocytes. TGF-ß1 decreased the expression of epithelial markers such as nephrin, zonula occludens-1 (ZO-1), while it increased the mesenchymal markers, including fibronectin (FN), vimentin, and α-smooth muscle actin (α-SMA) in the podocytes. Hesperetin suppressed EMT-like changes elicited by TGF-ß1. Interestingly, hesperetin did not interfere with the Smad signaling-the classical TGF-ß signaling-pathway, which was confirmed by the experiment with smad 2/3 -/- podocytes. Instead, hesperetin suppressed EMT-like changes by inhibiting the mTOR pathway-one of the alternative TGF-ß signaling pathways. In conclusion, hesperetin has a protective effect on the TGF-ß1 elicited EMT-like changes of podocytes through regulation of mTOR pathway. It could be a good candidate for the suppression of kidney fibrosis in various CKD.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Hesperidin/pharmacology , Podocytes/metabolism , Podocytes/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/toxicity , Cell Death/drug effects , Hesperidin/chemistry , Humans , Podocytes/drug effects , Signal Transduction/drug effects , Smad Proteins/metabolism
5.
Genes Dev ; 26(19): 2138-43, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22972936

ABSTRACT

The Hippo signaling pathway plays a crucial role in tissue growth and tumorigenesis. Core components of the Hippo pathway include the MST1/2 and Lats1/2 kinases. Acting downstream from the Hippo pathway are the YAP/TAZ transcription coactivators, which are inhibited through phosphorylation by Lats. However, upstream signals that regulate the Hippo pathway have not been well delineated. Here we report that stimulation of protease-activated receptors (PARs) activates YAP/TAZ by decreasing phosphorylation and increasing nuclear localization. PAR1 acts through G(12/13) and Rho GTPase to inhibit the Lats1/2 kinase. Our observations establish thrombin as a physiological signal for the Hippo pathway and implicate Hippo-YAP as a key downstream signaling branch of PAR activation.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Proteinase-Activated/metabolism , Transcription Factors/metabolism , Actin Cytoskeleton , Acyltransferases , Cell Cycle Proteins , Cell Line, Tumor , Cell Nucleus/enzymology , Enzyme Activation/physiology , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Oligopeptides/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , RNA, Small Interfering/metabolism , Receptors, Proteinase-Activated/agonists , Transcription Factors/genetics
6.
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
7.
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
8.
EMBO Rep ; 15(6): 642-56, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24825474

ABSTRACT

The Hippo signaling pathway, consisting of a highly conserved kinase cascade (MST and Lats) and downstream transcription coactivators (YAP and TAZ), plays a key role in tissue homeostasis and organ size control by regulating tissue-specific stem cells. Moreover, this pathway plays a prominent role in tissue repair and regeneration. Dysregulation of the Hippo pathway is associated with cancer development. Recent studies have revealed a complex network of upstream inputs, including cell density, mechanical sensation, and G-protein-coupled receptor (GPCR) signaling, that modulate Hippo pathway activity. This review focuses on the role of the Hippo pathway in stem cell biology and its potential implications in tissue homeostasis and cancer.


Subject(s)
Embryonic Development/physiology , Models, Biological , Neoplasms/physiopathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Stem Cells/physiology , Hippo Signaling Pathway , Homeostasis/physiology , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Nervous System/metabolism , Receptors, G-Protein-Coupled/metabolism , Regeneration/physiology , Skin/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.
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
11.
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
12.
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
13.
Endocrinology ; 165(3)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38366363

ABSTRACT

Histone deacetylase 11 (HDAC11) has been implicated in the pathogenesis of metabolic diseases characterized by chronic low-grade inflammation, such as obesity. However, the influence of HDAC11 on inflammation and the specific effect of HDAC11 on the palmitic acid (PA)-induced NLR family pyrin domain containing 3 (NLRP3) inflammasome activation are poorly understood. The effect of PA treatment on HDAC11 activity and the NLRP3 inflammasome was investigated in human peripheral blood mononuclear cells and THP-1 cells. The PA-induced responses of key markers of NLRP3 inflammasome activation, including NLRP3 gene expression, caspase-1 p10 activation, cleaved IL-1ß production, and extracellular IL-1ß release, were assessed as well. The role of HDAC11 was explored using a specific inhibitor of HDAC11 and by knockdown using small interfering (si)HDAC11 RNA. The relationship between HDAC11 and yes-associated protein (YAP) in the PA-induced NLRP3 inflammasome was investigated in THP-1 cells with HDAC11 or YAP knockdown. Following PA treatment, HDAC11 activity and protein levels increased significantly, concomitant with activation of the NLRP3 inflammasome. Notably, PA-induced the upregulation of NLRP3, caspase-1 p10 activation, the production of cleaved IL-1ß, and the release of IL-1ß into the extracellular space, all of which were attenuated by FT895 treatment and by HDAC11 knockdown. In THP-1 cells, PA induced the expression of YAP and its interaction with NLRP3, resulting in NLRP3 inflammasome activation, whereas both were inhibited by FT895 and siHDAC11 RNA. These findings demonstrate a pivotal role for HDAC11 in the PA-induced activation of the NLRP3 inflammasome. HDAC11 inhibition thus represents a promising therapeutic strategy for mitigating NLRP3 inflammasome-related inflammation in the context of obesity.


Subject(s)
Histone Deacetylases , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Caspase 1/genetics , Caspase 1/metabolism , Histone Deacetylases/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Interleukin-1beta/genetics , Leukocytes, Mononuclear , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity , Palmitates , Palmitic Acid/pharmacology , RNA , THP-1 Cells , YAP-Signaling Proteins/metabolism
14.
Transl Oncol ; 46: 101971, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38797019

ABSTRACT

Cholangiocarcinoma (CCA) is a devastating malignancy characterized by aggressive tumor growth and limited treatment options. Dysregulation of the Hippo signaling pathway and its downstream effector, Yes-associated protein (YAP), has been implicated in CCA development and progression. In this study, we investigated the effects of Isoalantolactone (IALT) on CCA cells to elucidate its effect on YAP activity and its potential clinical significance. Our findings demonstrate that IALT exerts cytotoxic effects, induces apoptosis, and modulates YAP signaling in SNU478 cells. We further confirmed the involvement of the canonical Hippo pathway by generating LATS1/LATS2 knockout cells, highlighting the dependence of IALT-mediated apoptosis and YAP phosphorylation on the Hippo-LATS signaling axis. In addition, IALT suppressed cell growth and migration, partially dependent on YAP-TEAD activity. These results provide insights into the therapeutic potential of targeting YAP in CCA and provide a rationale for developing of YAP-targeted therapies for this challenging malignancy.

15.
Cell Death Dis ; 15(1): 76, 2024 01 20.
Article in English | MEDLINE | ID: mdl-38245531

ABSTRACT

The Hippo pathway's main effector, Yes-associated protein (YAP), plays a crucial role in tumorigenesis as a transcriptional coactivator. YAP's phosphorylation by core upstream components of the Hippo pathway, such as mammalian Ste20 kinase 1/2 (MST1/2), mitogen-activated protein kinase kinase kinase kinases (MAP4Ks), and their substrate, large tumor suppressor 1/2 (LATS1/2), influences YAP's subcellular localization, stability, and transcriptional activity. However, recent research suggests the existence of alternative pathways that phosphorylate YAP, independent of these core upstream Hippo pathway components, raising questions about additional means to inactivate YAP. In this study, we present evidence demonstrating that TSSK1B, a calcium/calmodulin-dependent protein kinase (CAMK) superfamily member, is a negative regulator of YAP, suppressing cellular proliferation and oncogenic transformation. Mechanistically, TSSK1B inhibits YAP through two distinct pathways. Firstly, the LKB1-TSSK1B axis directly phosphorylates YAP at Ser94, inhibiting the YAP-TEAD complex's formation and suppressing its target genes' expression. Secondly, the TSSK1B-LATS1/2 axis inhibits YAP via phosphorylation at Ser127. Our findings reveal the involvement of TSSK1B-mediated molecular mechanisms in the Hippo-YAP pathway, emphasizing the importance of multilevel regulation in critical cellular decision-making processes.


Subject(s)
Hippo Signaling Pathway , Signal Transduction , Animals , Humans , Phosphorylation , YAP-Signaling Proteins , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Proliferation/physiology , Phosphoproteins/metabolism , Mammals
16.
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
17.
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
18.
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
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.
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
SELECTION OF CITATIONS
SEARCH DETAIL