Hes1 stimulates transcriptional activity of Runx2 by increasing protein stabilization during osteoblast differentiation

2008 ◽  
Vol 367 (1) ◽  
pp. 97-102 ◽  
Author(s):  
Jung Hee Suh ◽  
Hyun Woo Lee ◽  
Joo-Won Lee ◽  
Jae Bum Kim
Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3429-3429 ◽  
Author(s):  
Ya-Wei Qiang ◽  
Yu Chen ◽  
Owen Stephens ◽  
Jeff Rubin ◽  
Stuart Rudikoff ◽  
...  

Abstract Lytic bone destruction is a severe complication of multiple myeloma growth in the bone. We have demonstrated that Dkk1, an antagonist of Wnt signaling which is essential for osteoblast growth and differentiation, is expressed by myeloma plasma cells leading to increased levels of this protein in serum. Myeloma serum can block osteoblast differentiation in-vitro and a neutralizing anti-DKK1 antibody can inhibit this. These data suggest that MM cells disrupt Wnt signaling and that this promotes uncoupled bone turnover. However, the molecular mechanism responsible for this processes remains to be elucidated. Here we investigated Wnt signaling in osteoblast differentiation and identified the molecular mechanism regulating this process. Using qRT-PCR and sequence confirmation we showed that multiple Frizzled (Fz) mRNAs and LRP5/6 co-receptors were expressed in C2C12 cells and three human osteoblast cell lines, hFOB1.19, MG63 and Saos-2. Furthermore, these cells had high levels of total beta-catenin an indicator of active Wnt signaling. Wnt3a conditioned medium (CM) and recombinant Wnt3a protein increased uncomplexed beta-catenin level as assessed by GST-E-cadherin binding assay. Blocking autocrine Wnt signaling with Dkk1 blocked Wnt-3a-induced increase in beta-catenin levels in dose-dependent fashion. Co-culturing of OPM-2 MM cells ectopically expressing Dkk1 inhibited the Wnt-3a induced beta-catenin stabilization in C2C12 cells. Transfection of C2C12 cells with a dominant negative beta-catenin (DNBC) completely blocked endogenous and Wnt-3a-induced TCF/LEF transcriptional activity as evidenced by TOPflash luciferase activity. Furthermore, BMP-2 induced increase in alkaline phosphatase (ALP) activity was completed abrogated in C2C12 cells expressing DNCB. Silencing LRP5/6 mRNA with small inference RNAs blocked BMP-2 induced increase in ALP expression, suggesting that canonical Wnt pathway contributes to BPM-2 induced OB differentiation. BMP-2 induced phosphorylation of Smad 1, 5 and 8 whereas Wnt-3a had no effect on phosphorylation of these Smads. Moreover, Dkk1 did not block BMP-2 induced phosphorylation of Smad-1, −5 and −8 and did not alter transcriptional activity of Cbfa1, an essential transcriptional factor for OB differentiation. Taken together, these data suggest that Wnt induction of OB cell differentiation through canonical Wnt-beta-catenin pathway is independent of the activation of the Smad pathway. Our results suggested that canonical Wnt signaling pathway plays an important role in osteoblastogenesis and that Dkk1 inhibits OB cell differentiation in a way that may contribute to MM bone lesions. New therapies targeting DKK1 in myeloma may represent a novel treatment strategy.


2019 ◽  
Vol 11 (19) ◽  
pp. 2491-2504 ◽  
Author(s):  
Matthias R Bauer ◽  
Rhiannon N Jones ◽  
Raysa K Tareque ◽  
Bradley Springett ◽  
Felix A Dingler ◽  
...  

Aim: The p53 cancer mutation Y220C creates a conformationally unstable protein with a unique elongated surface crevice that can be targeted by molecular chaperones. We report the structure-guided optimization of the carbazole-based stabilizer PK083. Materials & methods: Biophysical, cellular and x-ray crystallographic techniques have been employed to elucidate the mode of action of the carbazole scaffolds. Results: Targeting an unoccupied subsite of the surface crevice with heterocycle-substituted PK083 analogs resulted in a 70-fold affinity increase to single-digit micromolar levels, increased thermal stability and decreased rate of aggregation of the mutant protein. PK9318, one of the most potent binders, restored p53 signaling in the liver cancer cell line HUH-7 with homozygous Y220C mutation. Conclusion: The p53-Y220C mutant is an excellent paradigm for the development of mutant p53 rescue drugs via protein stabilization. Similar rescue strategies may be applicable to other cavity-creating p53 cancer mutations.


2020 ◽  
Author(s):  
Özge Cicek Sener ◽  
Adrian Stender ◽  
Luisa Klemke ◽  
Nadine Stark ◽  
Tamara Isermann ◽  
...  

AbstractA prerequisite for gain-of-function (GOF) p53 missense mutants (mutp53) is protein stabilization. Moreover, a prerequisite for mutp53 stabilization is loss of the remaining wildtype (WT) p53 allele (loss-of-heterozygosity, p53LOH) in mutp53/+ tumors. Thus, GOF, mutp53 stabilization and p53LOH are strictly linked. However, the driving force for p53LOH is unknown. Typically, heterozygous tumors are an instable transition state. Here we identify the repressive WTp53-HSF1 axis as the driver of p53LOH.We find that the WTp53 allele in AOM/DSS-induced colorectal tumors (CRC) of p53R248Q/+ mice retains its haploid transcriptional activity. Notably, WTp53 represses heat-shock factor 1 (HSF1) activity, the master transcription factor of the proteotoxic stress defense response (HSR) that is ubiquitously and constitutively activated in cancer tissues. HSR is critical for stabilizing oncogenic proteins including mutp53. WTp53-retaining murine CRC tumors and tumor-derived organoids and human CRC cells all suppress the tumor-promoting HSF1 transcriptional program.Mechanistically, the retained WTp53 allele activates CDKN1A/p21, leading to cell cycle inhibition and suppression of the E2F target gene MLK3. MLK3 links cell cycle to the MAPK stress pathway to activate the HSR response. We show that in p53R248Q/+ tumors WTp53 activation by constitutive stress (emanating from proliferative/metabolic stresses and genomic instability) represses MLK3, consequently inactivating the MAPK-HSF1 response necessary to ensure tumor survival. This creates strong selection pressure for p53LOH which eliminates the repressive WTp53-HSF1 axis and unleashes the tumor-promoting HSF1 functions, inducing mutp53 stabilization and enabling invasion.HIGHLIGHTSheterozygous p53R248Q/+ tumors retain p53 transcriptional activity in a mouse model of colorectal cancer (CRC)wildtype p53 actively represses the tumor-promoting HSF1-regulated chaperone system and proteotoxic stress responsethe repressive WTp53 – HSF1 axis creates a selective pressure for WTp53 loss-of-heterozygosity in CRC tumorsp53 loss-of-heterozygosity enables stabilization of the gain-of-function p53R248Q mutant protein which in turn enables CRC invasion


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 644-644
Author(s):  
Ya-Wei Qiang ◽  
Bo Hu ◽  
Yu Chen ◽  
Ying Zhong ◽  
Bingyin Shi ◽  
...  

Abstract In multiple myeloma (MM), the proteasome inhibitor bortezomib induces mesenchymal stem cells (MSC) toward osteoblast differentiation. However, it is full unclear about the mechanism(s) underlying bortezomib in this process. Wnt/beta-catenin pathway plays a pivotal role in osteoblast differenciation and bone development. We have demonstrated that inhibition of Wnt/beta-catenin signaling by MM-derived Dkk1 suppresses osteoblast progenitor cell differentiation into osteoblasts (Qiang et al, Bone 2008) and deregulate RANKL and OPG expression in osteoblast cells (Qiang et al Blood 2008a). Increase Wnt signaling by overexpression of Wnt3a in myeloma cells diminished MM-trigged bone lesion in mouse model (Qiang et al Blood 2008b). In the present study we revealed that bortezomib promotes MSC differentiation into osteoblast cells via Wnt-independent activation of beta-catenin/TCF signaling. E-cadherin pull-down assay and subsequently immunoblotting analysis demonstrated that bortezomib induced increases in both free and active forms of beta-catenin protein in cytoplasm and nuclear in bell-shaped dose- and time-dependent manner in mouse and human osteoblast progenitor cell lines including C2C12, C3H10T1/2, Saos-2 and MG63. Similar results were illustrated in primary human 2 cases of normal MSC and MSC from 8 cases of MM pateints. Bortezomib induced increase in ubiquitinated beta-catenin was evidenced by obvious seen slow migration bands of beta-catenin protein in SDS-PAGE gel analysis indicating that bortezomib increased beta-catenin protein by modification of proteasome-mediated degradation of beta-catenin. Increase in cytoplasm and nuclear beta-catenin protein response to bortezomib treatment in the osteoblast cell lines and 4 cases MM derived MSC was further confirmed by immunofluorescent analysis. RT-PCT analysis of TCF family revealed that abundant TCF1 and TCF4 mRNA were expressed in all tested cell lines and in a primary normal MSC, and MM-derived MSC. Bortezomib treatment also resulted in TCF transcriptional activity in bell-shaped, dose-dependent pattern as determined by luciferase activity in these cells transfected with TOPflash plasmid DNAs. Maximal responses to bortezomib were seen at 12.5 nM for both C2C12 (p<0.001) and MG63 (P<0.01), 25 nM for C3H10T1/2 (p<0.001) (p<0.00001) compared with non-stimulation control. These results suggest that transcriptional activation was a downstream effect of bortezomib in osteoblast progenitor cell lines and MM derived MSC. Bortezomib induced increases in beta-catenin protein and TCF transcriptional activity were independent of modification expression of extracellular 19 members of Wnt family ligands, 10 members of Frizzled receptor family, LRP5/6 co-receptors, and antagonists of 4 members of Dkk and sFRP family, respectively, as determined by RT-PCR analysis. Bortezomib did not increase intracellular Dvl-3 proteins, a downstream target of Wnt pathway. Lithium chloride, an inhibitor of GSK3beta did not synergized bortezomib induced increases in beta-catenin protein or TCF transcriptional activity indicating that bortezomib active beta-catenin/TCF signaling independent of activity of GSK3beta. Blocking the catenin/TCF signaling by expressing dominant–activation of beta- negative TFC attenuated bortezomib-induced matrix mineralization indicating that bortezomib induced MSC differentiation into osteoblast through activation of beta-catenin/TCF signaling. Data from experiments in comparison with biological effect of bortezomib with Wnt3a demonstrated that bortezomib did not have effect on OPG and RANKL in these cells, while Wnt3a induces OPG mRNA and protein, but inhibited RANKL expression indicating that bortezomib may have not effect on osteoclastogensis. These results provide insights into a clinically relevant mechanism of action of bortezomib and as such a rationale for its use in the treatment of diseases related to suppression of Wnt/beta-catenin/TCF signaling.


2019 ◽  
Vol 294 (20) ◽  
pp. 8184-8196 ◽  
Author(s):  
William N. Addison ◽  
Martin Pellicelli ◽  
René St-Arnaud

The transcriptional cofactor nascent polypeptide-associated complex and co-regulator α (NACA) regulates osteoblast maturation and activity. NACA functions, at least in part, by binding to Jun proto-oncogene, AP-1 transcription factor subunit (cJUN) and potentiating the transactivation of AP-1 targets such as osteocalcin (Bglap) and matrix metallopeptidase 9 (Mmp9). NACA activity is modulated by phosphorylation carried out by several kinases, but a phosphatase regulating NACA's activity remains to be identified. Here, we used affinity purification with MS in HEK293T cells to isolate NACA complexes and identified protein phosphatase 1 catalytic subunit α (PP1A) as a NACA-associated Ser/Thr phosphatase. NACA interacted with multiple components of the PP1A holoenzyme complex: the PPP1CA catalytic subunit and the regulatory subunits PPP1R9B, PPP1R12A and PPP1R18. MS analysis revealed that NACA co-expression with PPP1CA causes dephosphorylation of NACA at Thr-89, Ser-151, and Thr-174. NACA Ser/Thr-to-alanine variants displayed increased nuclear localization, and NACA dephosphorylation was associated with specific recruitment of novel NACA interactants, such as basic transcription factor 3 (BTF3) and its homolog BTF3L4. NACA and PP1A cooperatively potentiated cJUN transcriptional activity of the AP-1–responsive MMP9-luciferase reporter, which was abolished when Thr-89, Ser-151, or Thr-174 were substituted with phosphomimetic aspartate residues. We confirmed the NACA–PP1A interaction in MC3T3-E1 osteoblastic cells and observed that NACA phosphorylation status at PP1A-sensitive sites is important for the regulation of AP-1 pathway genes and for osteogenic differentiation and matrix mineralization. These results suggest that PP1A dephosphorylates NACA at specific residues, impacting cJUN transcriptional activity and osteoblast differentiation and function.


2013 ◽  
Vol 288 (23) ◽  
pp. 16430-16437 ◽  
Author(s):  
Yue Tao ◽  
Minhao Wu ◽  
Xing Zhou ◽  
Wu Yin ◽  
Bin Hu ◽  
...  

Osterix (Osx) is an osteoblast-specific transcriptional factor and is required for osteoblast differentiation and bone formation. A JmjC domain-containing protein NO66 was previously found to participate in regulation of Osx transcriptional activity and plays an important role in osteoblast differentiation through interaction with Osx. Here, we report the crystal structure of NO66 forming in a functional tetramer. A hinge domain links the N-terminal JmjC domain and C-terminal winged helix-turn-helix domain of NO66, and both domains are essential for tetrameric assembly. The oligomerization interface of NO66 interacts with a conserved fragment of Osx. We show that the hinge domain-dependent oligomerization of NO66 is essential for inhibition of Osx-dependent gene activation. Our findings suggest that homo-oligomerization of JmjC domain containing proteins might play a physiological role through interactions with other regulatory factors during gene expression.


2020 ◽  
Vol 6 (43) ◽  
pp. eaba4147
Author(s):  
Jinlong Suo ◽  
Xue Feng ◽  
Jiayi Li ◽  
Jinghui Wang ◽  
Zuoyun Wang ◽  
...  

VGLL4 has been identified as a YAP inhibitor. However, the exact function of VGLL4 in bone development and bone homeostasis remains unclear. In this study, we demonstrated that VGLL4 breaks TEADs-mediated transcriptional inhibition of RUNX2 to promote osteoblast differentiation and bone development. We found that knockout of VGLL4 in mesenchymal stem cells and preosteoblasts showed osteoporosis and a cleidocranial dysplasia–like phenotype due to osteoblast differentiation disorders. Mechanistically, we showed that the TEAD transcriptional factors severely inhibited osteoblast differentiation in a YAP binding–independent manner. TEADs interacted with RUNX2 to repress RUNX2 transcriptional activity. Furthermore, VGLL4 relieved the transcriptional inhibition of TEADs by directly competing with RUNX2 to bind TEADs through its two TDU domains. Collectively, our studies demonstrate that VGLL4 plays an important role in regulating osteoblast differentiation and bone development, and that TEADs regulate the transcriptional activity of RUNX2, which may shed light on treatment of cleidocranial dysplasia and osteoporosis.


Oncotarget ◽  
2016 ◽  
Vol 7 (25) ◽  
pp. 37471-37486 ◽  
Author(s):  
Jianlei Lu ◽  
Shuang Qu ◽  
Bing Yao ◽  
Yuexin Xu ◽  
Yucui Jin ◽  
...  

2018 ◽  
Vol 46 (1) ◽  
pp. 269-278 ◽  
Author(s):  
Wenjun You ◽  
Lijuan Song ◽  
Kun Wang

Background/Aims: GATA4, a protein related to osteoblast differentiation and mineralization, whose acetylation is essential for cardiac defects. Here, we aimed to explore the functional impacts of GATA4 acetylation on osteoporosis (OS). Methods: GATA4 acetylation in hFOB1.19 and 293T cells was detected after exposure of HDAC inhibitors (TSA and SAHA). Co-immunoprecipitation was conducted to determine which HATs and HDACs was involved in the modulation of GATA4 acetylation/deacetylation, and to identify the acetylation site. The transcriptional activity of GATA4 was measured in the presence or absence of cycloheximide. Furthermore, hFOB1.19 cells viability and apoptosis were evaluated after transfection with acetylation-defective mutant of GATA4. Results: As a result, GATA4 acetylation was identified as a pivotal event in hFOB1.19 cells. GATA4 can be acetylated by P300/CBP, and the acetylation site was on lysine residue K313. Besides, the acetylation of GATA4 can be impaired by HDAC1, rather than by HDAC2-5. GATA4 acetylation contributed to the stability and transcription of GATA4. Moreover, GATA4 acetylation activated CCND2 transcription, and mutation of GATA4 on K-313 reduced cell viability and increased a mitochondria-dependent apoptosis in hFOB1.19 cells. Conclusion: Our data suggest that GATA4 exists as an acetylated protein in hFOB1.19 cells. Acetylation regulates the stability and transcription of GATA4, and activates CCND2 transcription, which may explain the growth-promoting functions of GATA4 in hFOB1.19 cells.


Sign in / Sign up

Export Citation Format

Share Document