Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters











Publication year range
1.
Mol Cell ; 68(4): 645-658.e5, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149593

ABSTRACT

Hajdu-Cheney syndrome (HCS), a rare autosomal disorder caused by heterozygous mutations in NOTCH2, is clinically characterized by acro-osteolysis, severe osteoporosis, short stature, neurological symptoms, cardiovascular defects, and polycystic kidneys. Recent studies identified that aberrant NOTCH2 signaling and consequent osteoclast hyperactivity are closely associated with the bone-related disorder pathogenesis, but the exact molecular mechanisms remain unclear. Here, we demonstrate that sustained osteoclast activity is largely due to accumulation of NOTCH2 carrying a truncated C terminus that escapes FBW7-mediated ubiquitination and degradation. Mice with osteoclast-specific Fbw7 ablation revealed osteoporotic phenotypes reminiscent of HCS, due to elevated Notch2 signaling. Importantly, administration of Notch inhibitors in Fbw7 conditional knockout mice alleviated progressive bone resorption. These findings highlight the molecular basis of HCS pathogenesis and provide clinical insights into potential targeted therapeutic strategies for skeletal disorders associated with the aberrant FBW7/NOTCH2 pathway as observed in patients with HCS.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Hajdu-Cheney Syndrome , Mutation , Osteoporosis , Proteolysis , Receptor, Notch2 , Animals , Cell Line , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Hajdu-Cheney Syndrome/genetics , Hajdu-Cheney Syndrome/metabolism , Mice, Knockout , Osteoporosis/genetics , Osteoporosis/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Ubiquitination/genetics
2.
Sci Signal ; 10(460)2017 01 03.
Article in English | MEDLINE | ID: mdl-28049764

ABSTRACT

The SCFß-TRCP E3 ubiquitin ligase complex plays pivotal roles in normal cellular physiology and in pathophysiological conditions. Identification of ß-transducin repeat-containing protein (ß-TRCP) substrates is therefore critical to understand SCFß-TRCP biology and function. We used a ß-TRCP-phosphodegron motif-specific antibody in a ß-TRCP substrate screen coupled with tandem mass spectrometry and identified multiple ß-TRCP substrates. One of these substrates was Lipin1, an enzyme and suppressor of the family of sterol regulatory element-binding protein (SREBP) transcription factors, which activate genes encoding lipogenic factors. We showed that SCFß-TRCP specifically interacted with and promoted the polyubiquitination of Lipin1 in a manner that required phosphorylation of Lipin1 by mechanistic target of rapamycin 1 (mTORC1) and casein kinase I (CKI). ß-TRCP depletion in HepG2 hepatocellular carcinoma cells resulted in increased Lipin1 protein abundance, suppression of SREBP-dependent gene expression, and attenuation of triglyceride synthesis. Moreover, ß-TRCP1 knockout mice showed increased Lipin1 protein abundance and were protected from hepatic steatosis induced by a high-fat diet. Together, these data reveal a critical physiological function of ß-TRCP in regulating hepatic lipid metabolic homeostasis in part through modulating Lipin1 stability.


Subject(s)
Lipogenesis , Liver/metabolism , Nuclear Proteins/metabolism , Phosphatidate Phosphatase/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Immunoblotting , Mice , Mice, Knockout , NIH 3T3 Cells , Nuclear Proteins/genetics , Phosphatidate Phosphatase/genetics , Phosphorylation , Protein Binding , Proteolysis , Reverse Transcriptase Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases/genetics , Substrate Specificity , Ubiquitination
3.
Oncotarget ; 8(6): 9947-9960, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28039480

ABSTRACT

Folliculin-interacting protein 1 and 2 (FNIP1 and FNIP2) play critical roles in preventing renal malignancy through their association with the tumor suppressor FLCN. Mutations in FLCN are associated with Birt-Hogg-Dubé (BHD) syndrome, a rare disorder with increased risk of renal cancer. Recent studies indicated that FNIP1/FNIP2 double knockout mice display enlarged polycystic kidneys and renal carcinoma, which phenocopies FLCN knockout mice, suggesting that these two proteins function together to suppress renal cancer. However, the molecular mechanism functionally linking FNIP1/FNIP2 and FLCN remains largely elusive. Here, we demonstrated that FNIP2 protein is unstable and subjected to proteasome-dependent degradation via ß-TRCP and Casein Kinase 1 (CK1)-directed ubiquitination in a nutrition-dependent manner. Degradation of FNIP2 leads to lysosomal dissociation of FLCN and subsequent lysosomal association of mTOR, which in turn promotes the proliferation of renal cancer cells. These results indicate that SCFß-TRCP negatively regulates the FLCN complex by promoting FNIP degradation and provide molecular insight into the pathogenesis of BHD-associated renal cancer.


Subject(s)
Birt-Hogg-Dube Syndrome/enzymology , Carcinoma, Renal Cell/enzymology , Carrier Proteins/metabolism , Cell Proliferation , Kidney Neoplasms/enzymology , Nutritional Status , Proto-Oncogene Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Birt-Hogg-Dube Syndrome/genetics , Birt-Hogg-Dube Syndrome/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carrier Proteins/genetics , Casein Kinase I/metabolism , Energy Metabolism , HEK293 Cells , HeLa Cells , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Lysosomes/metabolism , Mice, Nude , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Proto-Oncogene Proteins/genetics , RNA Interference , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection , Tumor Burden , Tumor Suppressor Proteins/genetics , Ubiquitination
4.
Clin Calcium ; 26(2): 298-304, 2016 Feb.
Article in Japanese | MEDLINE | ID: mdl-26813510

ABSTRACT

The transcriptional factor nuclear factor κB(NF-κB)regulates the expression of a wide variety of genes that are involved in immune and inflammatory responses, proliferation, and tumorigenesis. NF-κB consists of five members, such as p65(RelA), RelB, c-Rel, p50/p105(NF-κB1), and p52/p100(NF-κB2). There are two distinct NF-κB activation pathways, termed the classical and alternative NF-κB signaling pathways. Since mice lacking both p50 and p52 subunits developed typical osteopetrosis, due to total lack of osteoclasts, NF-κB is also important osteoclast differentiation. A selective NF-κB inhibitor blocked receptor activator of NF-κB ligand(RANKL)-induced osteoclastogenesis both in vitro and in vivo. Recent findings have shown that inactivation of NF-κB enhances osteoblast differentiation in vitro and bone formation in vivo. NF-κB is constitutively activated in many cancers including oral squamous cell carcinoma(OSCC), and is involved in the invasive characteristics of OSCC. A selective NF-κB inhibitor also prevented jaw bone destruction by OSCC by reduced osteoclast numbers in animal model. Thus the inhibition of NF-κB might useful for the treatment of bone diseases, such as arthritis, osteoporosis, periodontitis, and bone invasion by OSCC by inhibiting bone resorption and by stimulating bone formation.


Subject(s)
Benzamides , Carcinoma, Squamous Cell/drug therapy , Cell Differentiation/drug effects , Cell Differentiation/genetics , Jaw Neoplasms/drug therapy , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Osteoblasts/cytology , Osteoclasts/cytology , Osteogenesis/drug effects , Osteogenesis/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Animals , Benzamides/administration & dosage , Benzamides/pharmacology , Benzamides/therapeutic use , Carcinoma, Squamous Cell/pathology , Disease Models, Animal , Humans , Jaw/pathology , Jaw Neoplasms/pathology , Mice , Neoplasm Invasiveness , RANK Ligand
5.
Oncotarget ; 5(23): 12317-30, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25373602

ABSTRACT

Oral squamous cell carcinoma (OSCC) cells display significantly augmented nuclear factor-κB (NF-κB) activity, and inhibiting this activity suppresses malignant tumor characteristics. Thus, we evaluated the effect of IMD-0560, a novel inhibitor of IκB kinase (IKK) ß that is under assessment in a clinical trial of rheumatoid arthritis, on bone invasion by the mouse OSCC cell line SCCVII. We examined the inhibitory effects of IMD-0560 on NF-κB activity and tumor invasion using human OSCC cell lines and SCCVII cells in vitro. Using a mouse model of jaw bone invasion by SCCVII cells, we assessed the inhibitory effect of IMD-0560 on jaw bone invasion, tumor growth, and matrix degradation in vivo. IMD-0560 suppressed the nuclear translocation of NF-κB and the degradation of IκBα in OSCC cells. IMD-0560 also inhibited invasion by suppressing matrix metalloproteinase-9 (MMP-9) production in OSCC cells. IMD-0560 protected against zygoma and mandible destruction by SCCVII cells, reduced the number of osteoclasts by inhibiting receptor activator of NF-κB ligand (RANKL) expression in osteoblastic cells and SCCVII cells, increased SCCVII cell death and suppressed cell proliferation and MMP-9 production in SCCVII cells. Based on these results, IMD-0560 may represent a new therapeutic agent for bone invasion by OSCC cells.


Subject(s)
Benzamides/pharmacology , Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Carcinoma, Squamous Cell/secondary , Enzyme Inhibitors/pharmacology , Mouth Neoplasms/pathology , Neoplasm Metastasis/prevention & control , Animals , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Humans , I-kappa B Proteins/antagonists & inhibitors , Male , Mice , Microscopy, Fluorescence , Neoplasm Invasiveness/prevention & control , Real-Time Polymerase Chain Reaction
6.
Mol Endocrinol ; 28(9): 1460-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25029242

ABSTRACT

Bone morphogenic proteins (BMPs) stimulate bone formation in vivo and osteoblast differentiation in vitro via a Smad signaling pathway. Recent findings revealed that the activation of nuclear factor-κB (NF-κB) inhibits BMP-induced osteoblast differentiation. Here, we show that NF-κB inhibits BMP signaling by directly targeting the Smad pathway. A selective inhibitor of the classic NF-κB pathway, BAY11-770682, enhanced BMP2-induced ectopic bone formation in vivo. In mouse embryonic fibroblasts (MEFs) prepared from mice deficient in p65, the main subunit of NF-κB, BMP2, induced osteoblastic differentiation via the Smad complex to a greater extent than that in wild-type MEFs. In p65(-/-) MEFs, the BMP2-activated Smad complex bound much more stably to the target element than that in wild-type MEFs without affecting the phosphorylation levels of Smad1/5/8. Overexpression of p65 inhibited BMP2 activity by decreasing the DNA binding of the Smad complex. The C-terminal region, including the TA2 domain, of p65 was essential for inhibiting the BMP-Smad pathway. The C-terminal TA2 domain of p65 associated with the MH1 domain of Smad4 but not Smad1. Taken together, our results suggest that p65 inhibits BMP signaling by blocking the DNA binding of the Smad complex via an interaction with Smad4. Our study also suggests that targeting the association between p65 and Smad4 may help to promote bone regeneration in the treatment of bone diseases.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Gene Expression Regulation , Smad4 Protein/metabolism , Transcription Factor RelA/metabolism , Animals , Bone Development , Bone Diseases/metabolism , Cell Differentiation/genetics , Fibroblasts/metabolism , Humans , Mice , Osteoblasts/metabolism , Osteogenesis , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Signal Transduction/genetics
7.
Oncotarget ; 5(12): 4211-21, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24961988

ABSTRACT

CYLD negatively regulates the NF-κB signaling pathway and osteoclast differentiation largely through antagonizing TNF receptor-associated factor (TRAF)-mediated K63-linkage polyubiquitination in osteoclast precursor cells. CYLD activity is controlled by IκB kinase (IKK), but the molecular mechanism(s) governing CYLD protein stability remains largely undefined. Here, we report that SCFß-TRCP regulates the ubiquitination and degradation of CYLD, a process dependent on prior phosphorylation of CYLD at Ser432/Ser436 by IKK. Furthermore, depletion of ß-TRCP induced CYLD accumulation and TRAF6 deubiquitination in osteoclast precursor cells, leading to suppression of RANKL-induced osteoclast differentiation. Therefore, these data pinpoint the IKK/ß-TRCP/CYLD signaling pathway as an important modulator of osteoclastogenesis.


Subject(s)
SKP Cullin F-Box Protein Ligases/genetics , Tumor Suppressor Proteins/genetics , Animals , Deubiquitinating Enzyme CYLD , HeLa Cells , Humans , Male , Mice , Osteoclasts , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Transfection , Tumor Suppressor Proteins/metabolism , Ubiquitination
8.
J Biol Chem ; 289(11): 7349-61, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24488495

ABSTRACT

The alternative nuclear factor-κB (NF-κB) pathway, mainly the RelB-p52 heterodimer, plays important roles in bone metabolism through an unknown mechanism. We have previously reported that alymphoplasia (aly/aly) mice, which lack active NF-κB-inducing kinase (NIK), show mild osteopetrosis due to the inhibition of osteoclastogenesis. p100 retains RelB in the cytoplasm and inhibits RANKL-induced osteoclastogenesis in aly/aly cells. Furthermore, the overexpression of RelB in aly/aly cells rescues RANKL-induced osteoclastogenesis by inducing p100 processing. In contrast, the overexpression of p65 in aly/aly cells has no effect. However, the overexpression of RelB fails to rescue RANKL-induced osteoclastogenesis in the presence of p100ΔGRR, which cannot be processed to p52, suggesting that p100 processing is a key step in RelB-rescued, RANKL-induced osteoclastogenesis in aly/aly cells. In this study, Cot (cancer Osaka thyroid), an MAP3K, was up-regulated by RelB overexpression. Analysis of the Cot promoter demonstrated that p65 and RelB bound to the distal NF-κB-binding site and that RelB but not p65 bound to the proximal NF-κB-binding site in the Cot promoter. The knocking down of Cot expression significantly reduced the RANKL-induced osteoclastogenesis induced by RelB overexpression. The phosphorylation of IKKα at threonine 23 and its kinase activity were indispensable for the processing of p100 and osteoclastogenesis by RelB-induced Cot. Finally, constitutively activated Akt enhanced osteoclastogenesis by RelB-induced Cot, and a dominant-negative form of Akt significantly inhibited it. Taken together, these results indicate that the overexpression of RelB restores RANKL-induced osteoclastogenesis by activation of Akt/Cot/IKKα-induced p100 processing.


Subject(s)
I-kappa B Kinase/metabolism , MAP Kinase Kinase Kinases/metabolism , NF-kappa B p52 Subunit/metabolism , Osteoclasts/cytology , Proto-Oncogene Proteins/metabolism , Transcription Factor RelB/metabolism , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cells, Cultured , Female , Gene Expression Regulation , Glutathione Transferase/metabolism , MAP Kinase Signaling System , Macrophages/cytology , Male , Mice , Mice, Transgenic , Osteogenesis , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/metabolism , Retroviridae/metabolism , Signal Transduction
9.
Cell Rep ; 4(4): 803-16, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23972993

ABSTRACT

Proper cell-cycle transitions are driven by waves of ubiquitin-dependent degradation of key regulators by the anaphase-promoting complex (APC) and Skp1-Cullin1-F-box (SCF) E3 ubiquitin ligase complexes. But precisely how APC and SCF activities are coordinated to regulate cell-cycle progression remains largely unclear. We previously showed that APC/Cdh1 earmarks the SCF component Skp2 for degradation. Here, we continue to report that SCF(ß-TRCP) reciprocally controls APC/Cdh1 activity by governing Cdh1 ubiquitination and subsequent degradation. Furthermore, we define both cyclin A and Plk1, two well-known Cdh1 substrates, as upstream modifying enzymes that promote Cdh1 phosphorylation to trigger Cdh1 ubiquitination and subsequent degradation by SCF(ß-TRCP). Thus, our work reveals a negative repression mechanism for SCF to control APC, thereby illustrating an elegant dual repression system between these two E3 ligase complexes to create the ordered cascade of APC and SCF activities governing timely cell-cycle transitions.


Subject(s)
Cdh1 Proteins/metabolism , Cell Cycle , Feedback, Physiological , Proteolysis , SKP Cullin F-Box Protein Ligases/metabolism , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , Cell Cycle Proteins/metabolism , Cyclin A/metabolism , HEK293 Cells , HeLa Cells , Humans , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Ubiquitination , Xenopus , Polo-Like Kinase 1
10.
Cell Res ; 23(7): 947-61, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23670162

ABSTRACT

Fbw7 and Cdh1 are substrate-recognition subunits of the SCF- and APC-type E3 ubiquitin ligases, respectively. There is emerging evidence suggesting that both Fbw7 and Cdh1 function as tumor suppressors by targeting oncoproteins for destruction. Loss of Fbw7, but not Cdh1, is frequently observed in various human tumors. However, it remains largely unknown how Fbw7 mechanistically functions as a tumor suppressor and whether there is a signaling crosstalk between Fbw7 and Cdh1. Here, we report that Fbw7-deficient cells not only display elevated expression levels of SCF(Fbw7) substrates, including cyclin E, but also have increased expression of various APC(Cdh1) substrates. We further defined cyclin E as the critical signaling link by which Fbw7 governs APC(Cdh1) activity, as depletion of cyclin E in Fbw7-deficient cells results in decreased expression of APC(Cdh1) substrates to levels comparable to those in wild-type (WT) cells. Conversely, ectopic expression of cyclin E recapitulates the aberrant APC(Cdh1) substrate expression observed in Fbw7-deficient cells. More importantly, 4A-Cdh1 that is resistant to Cdk2/cyclin E-mediated phosphorylation, but not WT-Cdh1, reversed the elevated expression of various APC(Cdh1) substrates in Fbw7-deficient cells. Overexpression of 4A-Cdh1 also resulted in retarded cell growth and decreased anchorage-independent colony formation. Altogether, we have identified a novel regulatory mechanism by which Fbw7 governs Cdh1 activity in a cyclin E-dependent manner. As a result, loss of Fbw7 can lead to aberrant increase in the expression of both SCF(Fbw7) and APC(Cdh1) substrates. Our study provides a better understanding of the tumor suppressor function of Fbw7, and suggests that Cdk2/cyclin E inhibitors could serve as effective therapeutic agents for treating Fbw7-deficient tumors.


Subject(s)
Cdh1 Proteins/metabolism , Cyclin E/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cdh1 Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cyclin E/genetics , Humans , Immunoblotting , Immunoprecipitation , Male , Mice , Models, Biological , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/physiology
11.
J Bone Miner Res ; 28(12): 2449-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23526406

ABSTRACT

p130Cas, Crk-associated substrate (Cas), is an adaptor/scaffold protein that plays a central role in actin cytoskeletal reorganization. We previously reported that p130Cas is not tyrosine-phosphorylated in osteoclasts derived from Src-deficient mice, which are congenitally osteopetrotic, suggesting that p130Cas serves as a downstream molecule of c-Src and is involved in osteoclastic bone resorption. However, the physiological role of p130Cas in osteoclasts has not yet been confirmed because the p130Cas-deficient mice displayed embryonic lethality. Osteoclast-specific p130Cas conditional knockout (p130Cas(ΔOCL-) ) mice exhibit a high bone mass phenotype caused by defect in multinucleation and cytoskeleton organization causing bone resorption deficiency. Bone marrow cells from p130Cas(ΔOCL-) mice were able to differentiate into osteoclasts and wild-type cells in vitro. However, osteoclasts from p130Cas(ΔOCL-) mice failed to form actin rings and resorb pits on dentine slices. Although the initial events of osteoclast attachment, such as ß3-integrin or Src phosphorylation, were intact, the Rac1 activity that organizes the actin cytoskeleton was reduced, and its distribution was disrupted in p130Cas(ΔOCL-) osteoclasts. Dedicator of cytokinesis 5 (Dock5), a Rho family guanine nucleotide exchanger, failed to associate with Src or Pyk2 in osteoclasts in the absence of p130Cas. These results strongly indicate that p130Cas plays pivotal roles in osteoclastic bone resorption.


Subject(s)
Bone Resorption/metabolism , Bone Resorption/pathology , Crk-Associated Substrate Protein/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Actins/metabolism , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Bone and Bones/ultrastructure , Cell Differentiation , Guanine Nucleotide Exchange Factors/metabolism , Integrins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Organ Size , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoblasts/ultrastructure , Osteoclasts/ultrastructure , Osteogenesis , Phenotype , Signal Transduction , rac1 GTP-Binding Protein/metabolism
12.
J Bone Miner Res ; 28(6): 1457-67, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23322687

ABSTRACT

Mechanical unloading, such as in a microgravity environment in space or during bed rest (for patients who require prolonged bed rest), leads to a decrease in bone mass because of the suppression of bone formation and the stimulation of bone resorption. To address the challenges presented by a prolonged stay in space and the forthcoming era of a super-aged society, it will be important to prevent the bone loss caused by prolonged mechanical unloading. Nuclear factor κB (NF-κB) transcription factors are activated by mechanical loading and inflammatory cytokines. Our objective was to elucidate the role of NF-κB pathways in bone loss that are caused by mechanical unloading. Eight-week-old wild-type (WT) and NF-κB1-deficient mice were randomly assigned to a control or mechanically unloaded with tail suspension group. After 2 weeks, a radiographic analysis indicated a decrease in bone mass in the tibias and femurs of the unloaded WT mice but not in the NF-κB1-deficient mice. An NF-κB1 deficiency suppressed the unloading-induced reduction in bone formation by maintaining the proportion and/or potential of osteoprogenitors or immature osteoblasts, and by suppression of bone resorption through the inhibition of intracellular signaling through the receptor activator of NF-κB ligand (RANKL) in osteoclast precursors. Thus, NF-κB1 is involved in two aspects of rapid reduction in bone mass that are induced by disuse osteoporosis in space or bed rest.


Subject(s)
Bone Resorption/metabolism , NF-kappa B p50 Subunit/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/metabolism , Weightlessness/adverse effects , Animals , Bone Resorption/genetics , Bone Resorption/pathology , Femur/metabolism , Femur/pathology , Mice , Mice, Mutant Strains , NF-kappa B p50 Subunit/genetics , Osteoblasts/pathology , Osteoclasts/pathology , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/pathology , RANK Ligand/genetics , RANK Ligand/metabolism , Tibia/metabolism , Tibia/pathology , Time Factors
13.
Oncotarget ; 3(9): 1026-35, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22976441

ABSTRACT

The Mdm2 oncoprotein promotes p53 ubiquitination and destruction. Yet, exact molecular mechanisms of Mdm2 destruction itself, under DNA damaging conditions, remain unclear. Recently, we identified SCFß-TRCP as a novel E3 ligase that targets Mdm2 for ubiquitination and destruction in a Casein Kinase Iδ (CKIδ)-dependent manner. However, it remains elusive how the ß-TRCP/CKIδ/Mdm2 signaling axis is regulated by DNA damage signals to govern p53 activity. Consistent with previous studies, we found that inactivation of the Ataxia Telangiectasia Mutated (ATM) kinase, in turn, impaired DNA damage-induced Mdm2 destruction. Although phosphorylation of Mdm2 at Ser395 (an ATM phosphorylation site) facilitated Mdm2 interaction with ß-TRCP, Ser395A-Mdm2 was degraded non-distinguishably from WT-Mdm2 by SCFß-TRCP upon DNA damaging treatments. This indicates that in addition to phosphorylating Mdm2 at Ser395, ATM may govern Mdm2 stability through other unknown mechanisms. We further demonstrated that DNA damage-induced activation of ATM directly phosphorylated CKIδ at two well-conserved S/TQ sites, which promotes CKIδ nuclear localization to increase CKIδ-mediated phosphorylation of Mdm2, thereby facilitating subsequent Mdm2 ubiquitination by SCFß-TRCP. Our studies provide a molecular mechanism of how ATM could govern DNA damage-induced destruction of Mdm2 in part by phosphorylating both Mdm2 and CKIδ to modulate SCFß-TRCP-mediated Mdm2 ubiquitination. Given the pivotal role of Mdm2 in the negative regulation of p53, this work will also provide a rationale for developing CKIδ or ATM agonists as anti-cancer agents.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Proteins/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Amino Acid Sequence , Ataxia Telangiectasia Mutated Proteins , Casein Kinase Idelta/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Humans , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Signal Transduction , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , beta-Transducin Repeat-Containing Proteins/genetics
14.
Cell ; 150(1): 179-93, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770219

ABSTRACT

Aberrant Skp2 signaling has been implicated as a driving event in tumorigenesis. Although the underlying molecular mechanisms remain elusive, cytoplasmic Skp2 correlates with more aggressive forms of breast and prostate cancers. Here, we report that Skp2 is acetylated by p300 at K68 and K71, which is a process that can be antagonized by the SIRT3 deacetylase. Inactivation of SIRT3 leads to elevated Skp2 acetylation, which leads to increased Skp2 stability through impairment of the Cdh1-mediated proteolysis pathway. As a result, Skp2 oncogenic function is increased, whereby cells expressing an acetylation-mimetic mutant display enhanced cellular proliferation and tumorigenesis in vivo. Moreover, acetylation of Skp2 in the nuclear localization signal (NLS) promotes its cytoplasmic retention, and cytoplasmic Skp2 enhances cellular migration through ubiquitination and destruction of E-cadherin. Thus, our study identifies an acetylation-dependent regulatory mechanism governing Skp2 oncogenic function and provides insight into how cytoplasmic Skp2 controls cellular migration.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Prostatic Neoplasms/pathology , S-Phase Kinase-Associated Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Cadherins/metabolism , Casein Kinase I/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , Disease Models, Animal , Humans , Lysine/metabolism , Male , Mice , Molecular Sequence Data , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , Protein Sorting Signals , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/genetics , Sequence Alignment , Ubiquitination
15.
Front Biosci (Landmark Ed) ; 17(6): 2197-212, 2012 06 01.
Article in English | MEDLINE | ID: mdl-22652772

ABSTRACT

The Ubiquitin Proteasome System (UPS) is a major regulator of protein abundance in the cell. The UPS influences the functions of multiple biological processes by targeting key regulators for destruction. E3 ubiquitin ligases are a vital component of the UPS machinery, working with E1 and E2 enzymes to bind substrates and facilitate the transfer of ubiquitin molecules onto the target protein. This poly-ubiquitination, in turn, directs the modified proteins for proteolysis by the 26S proteasome. As the UPS regulates the degradation of multiple oncogenes and tumor suppressors, the dysregulation of this pathway is known to promote various diseases including cancer. While E1 and E2 enzymes have only been minimally linked to cancer development, burgeoning amounts of evidence have implicated loss or gain of E3 function as a key factor in cancer initiation and progression. This review will examine the literature on two SCF-type E3 ligases, SCFFbw7 and SCFbeta-TRCP. In particular, we will highlight novel substrates recently identified for these two E3 ligases, and further discuss how UPS regulation of these targets may promote carcinogenesis.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Neoplasms/enzymology , Neoplasms/etiology , Ubiquitin-Protein Ligases/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Genes, Tumor Suppressor , Humans , Models, Biological , Mutation , Neoplasms/prevention & control , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Substrate Specificity , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , beta-Transducin Repeat-Containing Proteins/genetics
16.
FEBS Lett ; 586(10): 1409-18, 2012 May 21.
Article in English | MEDLINE | ID: mdl-22673505

ABSTRACT

FBW7 (F-box and WD repeat domain-containing 7) has been characterized as an onco-suppressor protein in human cancers. Recent studies have also shown that FBW7 exerts its anti-tumor function primarily by promoting the degradation of various oncoproteins, through which FBW7 regulates cellular proliferation, differentiation and causes genetic instability. In this review, we will discuss the role of FBW7 downstream substrates and how dysregulation of Fbw7-mediated proteolysis of these substrates contributes to tumorigenesis. Additionally, we will also summarize the currently available various Fbw7-knockout mouse models that support Fbw7 as a tumor suppressor gene in the development and progression of human malignancies.


Subject(s)
Cell Cycle Proteins/physiology , F-Box Proteins/physiology , Genes, Tumor Suppressor , Neoplasms/pathology , Ubiquitin-Protein Ligases/physiology , Animals , Cell Cycle Proteins/genetics , Cell Differentiation/physiology , Cell Proliferation , Disease Progression , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Humans , Mice , Mice, Knockout , Models, Animal , Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics
17.
J Exp Med ; 209(7): 1289-307, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22711876

ABSTRACT

The incidence of human papillary thyroid cancer (PTC) is increasing and an aggressive subtype of this disease is resistant to treatment with vascular endothelial growth factor receptor 2 (VEGFR2) inhibitor. VEGFR2 promotes angiogenesis by triggering endothelial cell proliferation and migration. However, the molecular mechanisms governing VEGFR2 stability in vivo remain unknown. Additionally, whether VEGFR2 influences PTC cell migration is not clear. We show that the ubiquitin E3 ligase SCF(ß-TRCP) promotes ubiquitination and destruction of VEGFR2 in a casein kinase I (CKI)-dependent manner. ß-TRCP knockdown or CKI inhibition causes accumulation of VEGFR2, resulting in increased activity of signaling pathways downstream of VEGFR2. ß-TRCP-depleted endothelial cells exhibit enhanced migration and angiogenesis in vitro. Furthermore, ß-TRCP knockdown increased angiogenesis and vessel branching in zebrafish. Importantly, we found an inverse correlation between ß-TRCP protein levels and angiogenesis in PTC. We also show that ß-TRCP inhibits cell migration and decreases sensitivity to the VEGFR2 inhibitor sorafenib in poorly differentiated PTC cells. These results provide a new biomarker that may aid a rational use of tyrosine kinase inhibitors to treat refractory PTC.


Subject(s)
Cell Movement , Neovascularization, Pathologic/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Thyroid Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adult , Aged , Amino Acid Sequence , Animals , Cell Line, Tumor , Cells, Cultured , Female , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Male , Middle Aged , Molecular Sequence Data , Neovascularization, Pathologic/genetics , Protein Binding , RNA Interference , SKP Cullin F-Box Protein Ligases/genetics , Sequence Homology, Amino Acid , Signal Transduction , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Ubiquitination , Vascular Endothelial Growth Factor Receptor-2/genetics , Young Adult , Zebrafish
18.
Cell Rep ; 1(5): 434-43, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22708077

ABSTRACT

The NFkB/Rel family of proteins play critical roles in a variety of cellular processes. Thus, their physiological activation is tightly controlled. Recently, the NFkB2/p100 precursor has been characterized as the fourth IkB type of suppressor for NFkB. However, the molecular mechanism(s) underlying regulated destruction of NFkB2 remains largely unknown. Here, we report that, unlike other IkBs, ubiquitination and destruction of NFkB2 are governed by SCF(Fbw7) in a GSK3-dependent manner. In Fbw(7-/-) cells, elevated expression of NFkB2/p100 leads to a subsequent reduction in NFkB signaling pathways and elevated sensitivity to TNFa-induced cell death. Reintroducing wild-type Fbw7, but not disease-derived mutant forms of Fbw7, rescues NFkB activity. Furthermore, T cell-specific depletion of Fbw7 also leads to reduced NFkB activity and perturbed T cell differentiation. Therefore, our work identifies Fbw7 as a physiological E3 ligase controlling NFkB20s stability. It further implicates that Fbw7 might exert its tumor-suppressor function by regulating NFkB activity.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , NF-kappa B p52 Subunit/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Amino Acid Sequence , Animals , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Glycogen Synthase Kinase 3/metabolism , Humans , Mice , Mice, Knockout , Molecular Sequence Data , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
19.
Int J Dent ; 2012: 148261, 2012.
Article in English | MEDLINE | ID: mdl-22505894

ABSTRACT

Bone defects often result from tumor resection, congenital malformation, trauma, fractures, surgery, or periodontitis in dentistry. Although dental implants serve as an effective treatment to recover mouth function from tooth defects, many patients do not have the adequate bone volume to build an implant. The gold standard for the reconstruction of large bone defects is the use of autogenous bone grafts. While autogenous bone graft is the most effective clinical method, surgical stress to the part of the bone being extracted and the quantity of extractable bone limit this method. Recently mesenchymal stem cell-based therapies have the potential to provide an effective treatment of osseous defects. In this paper, we discuss both the current therapy for bone regeneration and the perspectives in the field of stem cell-based regenerative medicine, addressing the sources of stem cells and growth factors used to induce bone regeneration effectively and reproducibly.

20.
Mol Endocrinol ; 26(3): 414-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22282470

ABSTRACT

We previously reported that alymphoplasia (aly/aly) mice, which have a natural loss-of-function mutation in the Nik gene, which encodes a kinase essential for the processing of p100 to p52 in the alternative nuclear factor-κB (NF-κB) pathway, show mild osteopetrosis with an increase in several parameters of bone formation: bone formation rate, mineral apposition rate, and osteoblast number. We therefore investigated the molecular mechanisms triggered by the alternative NF-κB pathway in the regulation of osteoblast differentiation using primary osteoblasts (POB) prepared from aly/aly mice. Alkaline phosphatase (ALP) activity and mineralization induced by the presence of ß-glycerophosphate and ascorbic acid were enhanced in POB from aly/aly compared with wild-type (WT) mice. Furthermore, osteoblastic differentiation induced by bone morphogenetic protein 2 (BMP2), as shown by ALP activity, mRNA expression of osteocalcin, Id1, Osterix and Runx2, and Sma- and Mad-related protein (Smad)1/5/8 phosphorylation, was also enhanced in POB from aly/aly mice. The ectopic bone formation in vivo that was induced by BMP2 was enhanced in aly/aly mice compared with controls. Transfection of a mutant form of p100, p100ΔGRR, which cannot be processed to p52, stimulated ALP activity and Smad phosphorylation. In contrast to p100ΔGRR, overexpression of p52 inhibited these events. Both BMP2-induced ALP activity and Smad phosphorylation were reduced in POB from p100-deficient mice, which carry a homozygous deletion of the COOH-terminal ankyrin repeats of p100 but still express functional p52 protein. p52 and p100ΔGRR interacted with a BMP receptor, ALK2, in overexpressed COS7 cells and changed the ALK2 protein levels in opposite directions: p52 reduced ALK2 and p100 increased it. Thus, the alternative the NF-κB pathway via the processing of p52 from p100 negatively regulates osteoblastic differentiation and bone formation by modifying BMP activity.


Subject(s)
Cell Differentiation , NF-kappa B p52 Subunit/physiology , Osteoblasts/physiology , Osteogenesis , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/physiology , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/metabolism , Cells, Cultured , Female , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , NF-kappa B p52 Subunit/metabolism , Osteoblasts/enzymology , Osteoblasts/metabolism , Phosphorylation , Primary Cell Culture , Prostheses and Implants , Protein Binding , Skull/cytology , Smad Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL