scholarly journals Metastasis suppressor 1 controls osteoblast differentiation and bone homeostasis through regulating Src-Wnt/β-catenin signaling

Author(s):  
Meng Chen ◽  
Liying Shan ◽  
Ying Gan ◽  
Lijie Tian ◽  
Jie Zhou ◽  
...  

Abstract Metastasis suppressor 1 (MTSS1) plays an inhibitory role in tumorigenesis and metastasis of a variety of cancers. To date, the function of MTSS1 in the differentiation of marrow stromal progenitor cells is completely unknown. In the current study, we explored whether and how MTSS1 has a role in osteoblast differentiation and bone homeostasis. Our data showed that MTSS1 mRNA was upregulated during osteoblast differentiation and downregulated in the osteoblastic lineage cells of ovariectomized and aged mice. Functional studies revealed that MTSS1 promoted the osteogenic differentiation from marrow stromal progenitor cells. Mechanistic explorations uncovered that the inactivation of Src and afterwards activation of canonical Wnt signaling were involved in osteoblast differentiation induced by MTSS1. The enhanced osteogenic differentiation induced by MTSS1 overexpression was attenuated when Src was simultaneously overexpressed, and conversely, the inhibition of osteogenic differentiation by MTSS1 siRNA was rescued when the Src inhibitor was supplemented to the culture. Finally, the in vivo transfection of MTSS1 siRNA to the marrow of mice significantly reduced the trabecular bone mass, along with the reduction of trabecular osteoblasts, the accumulation of marrow adipocytes, and the increase of phospho-Src positive cells on the trabeculae. No change in the number of osteoclasts was observed. This study has for the first time unraveled that MTSS1 contributes to osteoblast differentiation and bone homeostasis through regulating Src-Wnt/β-catenin signaling. It also suggests the potential of MTSS1 as a new target for the treatment of osteoporosis.

Molecules ◽  
2018 ◽  
Vol 23 (12) ◽  
pp. 3119 ◽  
Author(s):  
Shin-Hye Kim ◽  
Kwang-Jin Kim ◽  
Hyeon Kang ◽  
Young-Jin Son ◽  
Sik-Won Choi ◽  
...  

The number of patients with bone metabolic disorders including osteoporosis is increasing worldwide. These disorders often facilitate bone fractures, which seriously impact the patient’s quality of life and could lead to further health complications. Bone homeostasis is tightly regulated to balance bone resorption and formation. However, many anti-osteoporotic agents are broadly categorized as either bone forming or anti-resorptive, and their therapeutic use is often limited due to unwanted side effects. Therefore, safe and effective therapeutic agents are needed for osteoporosis. This study aims to clarify the bone protecting effects of oat bran water extract (OBWE) and its mode of action. OBWE inhibited RANKL (receptor activator of nuclear factor-κB ligand)-induced osteoclast differentiation by blocking c-Fos/NFATc1 through the alteration of I-κB. Furthermore, we found that OBWE enhanced BMP-2-stimulated osteoblast differentiation by the induction of Runx2 via Smad signaling molecules. In addition, the anti-osteoporotic activity of OBWE was also evaluated using an in vivo model. OBWE significantly restored ovariectomy-induced bone loss. These in vitro and in vivo results showed that OBWE has the potential to prevent and treat bone metabolic disorders including osteoporosis.


Cells ◽  
2020 ◽  
Vol 9 (6) ◽  
pp. 1500
Author(s):  
Feng-Sheng Wang ◽  
Yu-Shan Chen ◽  
Jih-Yang Ko ◽  
Chung-Wen Kuo ◽  
Huei-Jing Ke ◽  
...  

Glucocorticoid provokes bone mass loss and fatty marrow, accelerating osteoporosis development. Bromodomain protein BRD4, an acetyl–histone-binding chromatin reader, regulates stem cell and tissue homeostasis. We uncovered that glucocorticoid inhibited acetyl Lys-9 at the histone 3 (H3K9ac)-binding Runx2 promoter and decreased osteogenic differentiation, whereas bromodomain protein 4 (BRD4) and adipocyte formation were upregulated in bone-marrow mesenchymal progenitor cells. BRD4 knockdown improved H3K9ac occupation at the Runx2 promoter and osteogenesis, but attenuated glucocorticoid-mediated adipocyte formation together with the unaffected H3K9ac-binding PPARγ2 promoter. BRD4 regulated epigenome related to fatty acid metabolism and the forkhead box P1 (Foxp1) pathway, which occupied the PPARγ2 promoter to modulate glucocorticoid-induced adipocytic activity. In vivo, BRD4 inhibitor JQ-1 treatment mitigated methylprednisolone-induced suppression of bone mass, trabecular microstructure, mineral acquisition, and osteogenic differentiation. Foxp1 signaling, marrow fat, and adipocyte formation in glucocorticoid-treated skeleton were reversed upon JQ-1 treatment. Taken together, glucocorticoid-induced H3K9 hypoacetylation augmented BRD4 action to Foxp1, which steered mesenchymal progenitor cells toward adipocytes at the cost of osteogenic differentiation in osteoporotic skeletons. BRD4 inhibition slowed bone mass loss and marrow adiposity. Collective investigations convey a new epigenetic insight into acetyl histone reader BRD4 control of osteogenesis and adipogenesis in skeleton, and highlight the remedial effects of the BRD4 inhibitor on glucocorticoid-induced osteoporosis.


2019 ◽  
Vol 5 (1) ◽  
Author(s):  
Rongze Wang ◽  
Yuanxu Zhang ◽  
Fujun Jin ◽  
Gongchen Li ◽  
Yao Sun ◽  
...  

Abstract Type 1 diabetes mellitus (T1DM) is an autoimmune insulin-dependent disease associated with destructive bone homeostasis. Accumulating evidence has proven that miRNAs are widely involved in the regulation of bone homeostasis. However, whether miRNAs also regulate osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in T1DM mice is under exploration. In this study, miRNA microarray was utilized to screen the differentially expressed miRNAs, which uncovered that miR-214-3p potentially inhibited BMSCs osteogenic differentiation in T1DM mice. We found that high glucose suppressed BMSCs osteogenic differentiation with significant elevation of the miR-214-3p expression. Further study found that the osteogenic differentiation of BMSCs was inhibited by AgomiR-214-3p while enhanced by AntagomiR-214-3p in BMSCs supplemented with high glucose. Moreover, we found that miR-214-3p knockout T1DM mice were resistant to high-glucose-induced bone loss. These results provide a novel insight into an inhibitory role of high-glucose-induced miR-214-3p in BMSCs osteogenic differentiation both in vitro and in vivo. Molecular studies revealed that miR-214-3p inhibits BMSCs osteogenic differentiation by targeting the 3′-UTR of β-catenin, which was further corroborated in human bone specimens and BMSCs of T1DM patients. Taken together, our study discovered that miR-214-3p is a pivotal regulator of BMSCs osteogenic differentiation in T1DM mice. Our findings also suggest that miR-214-3p could be a potential target in the treatment of bone disorders in patients with T1DM.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2776-2776
Author(s):  
Mirle Schemionek ◽  
Ulrich Steidl ◽  
Albrecht Schwab ◽  
Daniel G Tenen ◽  
Copland Mhairi ◽  
...  

Abstract Abstract 2776 The implementation of Bcr-Abl tyrosine kinase inhibitors (TKIs) has greatly improved the outcome of patients with chronic myeloid leukemia (CML). However, discontinuation of TKI therapy often results in relapse suggesting that leukemic stem cells (LSCs) survive despite treatment. More detailed investigations utilizing patient samples and murine CML models have confirmed that the leukemia-initiating cell population is usually not eradicated by inhibiting Bcr-Abl activity and that this is due to a lack of oncogene addiction of LSCs, showing that further research is required aiming to fully understand LSC biology. To identify new Bcr-Abl targets that are involved in LSC persistence, we performed a microarray analysis of the leukemia-initiating cell population in an inducible transgenic SCLtTAxBcr-Abl CML mouse model in which we had previously shown that these cells are not oncogene-addicted (Schemionek et al., BLOOD 2010; Hamilton et al., BLOOD 2012). One of the most downregulated genes in CML vs. normal stem cells was Metastasis Suppressor 1 (Mtss1/MIM). Although the multidomain protein Mtss1 may be involved in carcinogenesis of several solid tumors, its exact physiological role is still unknown. Current findings suggest that Mtss1 interacts with multiple partners and is involved in various signalling pathways that regulate actin dynamics and cell motility. Interestingly, Rac and Src are Mtss1 interacting partners, and both proteins are known to be involved in Bcr-Abl mediated leukemogenesis. We have previously shown that Mtss1 is downregulated in mouse LSCs and demonstrated that this is a Bcr-Abl kinase mediated effect in various human and murine CML cell lines. Moreover, we have shown that Mtss1 overexpression in 32D-Bcr-Abl cells induces a defect in Bcr-Abl mediated migration in vitro and reduces the potential to form solid tumors in vivo. Here we show that Bcr-Abl kinase-dependent regulation of Mtss1 expression was also evident in mononuclear cells and CD34+ progenitor cells from patients with CML upon IM or dasatinib treatment in vitro. Moreover, cells from IM-treated patients with chronic phase CML showed elevated Mtss1 expression levels within one to three weeks of treatment. Increasing Mtss1 expression upon 5-aza-2′-deoxycytidine-treatment of K562 and 32D-Bcr-Abl cells suggested that methylation might be involved in Mtss1 regulation. To determine a potential leukemia suppressing effect of Mtss1 overexpression, we performed colony assays using lineage negative SCLtTAxBcr-Abl (dtg) bone marrow (BM) cells that had been retrovirally infected to overexpress Mtss1 (dtg::Mtss1) or empty-vector (dtg::ev). Successfully transduced BM cells were FACS-sorted via GFP-expression, encoded by the retroviral vector. Mtss1 overexpression led to a 2.3-fold decrease in CFU numbers. In a second set of experiments we transplanted 1.3×105 GFP-FACS-sorted dtg::Mtss1 or dtg::ev cells into 9 Gy-irradiated recipients. While dtg::ev recipients contained 66% (+/−8%) of GFP-positive cells in the BM, these cells were decreased in dtg::Mtss1 transplanted mice to 23% (+/−21%), 12 days after transplantation. A similar effect was evident in the spleen [dtg::ev recipients: 90% (+/− 3%) versus dtg::Mtss1 recipients 59% (+/−20%)] suggesting that Mtss1 confers a disadvantage to Bcr-Abl positive BM cells in the early steps of leukemic cell propagation, compared to Bcr-Abl cells alone. Since the multidomain Mtss1 protein contains a putative Abl-SH2-binding site, we performed co-immunoprecipitations using 32D-Bcr-Abl-Flag-Mtss1 cells. These experiments showed that both proteins were direct binding partners and that Mtss1 was not phosphorylated by Bcr-Abl. Taken together, our data show that Mtss1 is downregulated via a Bcr-Abl kinase mediated mechanism and this might involve methylation. Moreover, additional inhibition of Mtss1 activity might be mediated through direct binding by Bcr-Abl. Forced expression of the potential tumor suppressor in CML stem and progenitor cells reduces leukemic cell propagation in vivo and may thus provide a rationale to contribute to LSC elimination in patients with CML. Disclosures: Mhairi: BMS: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Pfizer: Honoraria. Koschmieder:Novartis / Novartis Foundation: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Honoraria, Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Kai Hang ◽  
Li Ying ◽  
Jinwu Bai ◽  
Yibo Wang ◽  
Zhihui Kuang ◽  
...  

Abstract Background Globally, bone fractures are the most common musculoskeletal trauma, and approximately 8–10% of cases that fall into the categories of delayed or non-union healing. To date, there are no efficient pharmacological agents to accelerate the healing of bone fractures. Thus, it is necessary to find new strategies that accelerate bone healing and reduce the incidence of non-union or delayed fracture healing. Previous studies have revealed that the plasminogen activation system has been demonstrated to play an important role in bone metabolism. However, the function of SERPINB2 in the osteogenesis of hBMSCs remains unclear. Therefore, in this study, we investigated the effects and mechanism of SERPINB2 on osteogenic differentiation. Methods We investigated the osteogenesis effects of hBMSCs by both exogenous SerpinB2 protein and SERPINB2 gene silencing in vitro. Cell proliferation assay was used to assess the effect of exogenous SerpinB2 or SERPINB2 silencing on proliferation of hBMSCs. qPCR and Western blotting analysis detected the expression of target genes and proteins respectively. ALP staining was used to evaluated ALP activity and Alizarin Red staining (ARS) was used to evaluate mineral deposition. In vivo, a murie tibial fracture model was established, histological evaluation and radiographic analysis was used to confirm the therapeutic effects of SERPINB2 silencing in fracture healing. Statistical significance between two groups was determined by Student’s t test, one-way ANOVA or Bonferroni’s post-hoc test according to the distribution of the tested population. Results The addition of exogenous SerpinB2 protein inhibted osteoblast differentiation of hBMSCs in vitro, while SERPINB2 gene silencing significant promote osteoblast differentiation of hBMSCs in vitro. And silenced SERPINB2 gene also increased mineral deposits. Moreover, β-catenin levels were up-regulated by SERPINB2 gene depletion. And the enhancement of osteogenic differentiation induced by SERPINB2 silencing was almost inhibited by specific Wnt/β-catenin signaling pathway inhibitor. In a murine tibial fracture model, local injection of SERPINB2 siRNA improved bone fracture healing. Conclusions Taken together, these findings indicate that SERPINB2 silencing promoted osteogenic differentiation of BMSCs via the Wnt/β-catenin signaling pathway, and silenced SERPINB2 in vivo effectively promotes fracture healing, suggesting that SERPINB2 may be a novel target for bone fracture healing.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Dan Zhang ◽  
Kim De Veirman ◽  
Rong Fan ◽  
Qiang Jian ◽  
Yuchen Zhang ◽  
...  

Abstract Background Bone destruction is a hallmark of multiple myeloma (MM). It has been reported that proteasome inhibitors (PIs) can reduce bone resorption and increase bone formation in MM patients, but the underlying mechanisms remain unclear. Methods Mesenchymal stem cells (MSCs) were treated with various doses of PIs, and the effects of bortezomib or carfilzomib on endoplasmic reticulum (ER) stress signaling pathways were analyzed by western blotting and real-time PCR. Alizarin red S (ARS) and alkaline phosphatase (ALP) staining were used to determine the osteogenic differentiation in vitro. Specific inhibitors targeting different ER stress signaling and a Tet-on inducible overexpressing system were used to validate the roles of key ER stress components in regulating osteogenic differentiation of MSCs. Chromatin immunoprecipitation (ChIP) assay was used to evaluate transcription factor-promoter interaction. MicroCT was applied to measure the microarchitecture of bone in model mice in vivo. Results We found that both PERK-ATF4 and IRE1α-XBP1s ER stress branches are activated during PI-induced osteogenic differentiation. Inhibition of ATF4 or XBP1s signaling can significantly impair PI-induced osteogenic differentiation. Furthermore, we demonstrated that XBP1s can transcriptionally upregulate ATF4 expression and overexpressing XBP1s can induce the expression of ATF4 and other osteogenic differentiation-related genes and therefore drive osteoblast differentiation. MicroCT analysis further demonstrated that inhibition of XBP1s can strikingly abolish bortezomib-induced bone formation in mouse. Conclusions These results demonstrated that XBP1s is a master regulator of PI-induced osteoblast differentiation. Activation of IRE1α-XBP1s ER stress signaling can promote osteogenesis, thus providing a novel strategy for the treatment of myeloma bone disease.


2007 ◽  
Vol 13 (4) ◽  
pp. 412-422 ◽  
Author(s):  
Andrea T. Badillo ◽  
Kirstin J. Beggs ◽  
Elisabeth H. Javazon ◽  
Jessica C. Tebbets ◽  
Alan W. Flake

Author(s):  
Shin-Hye Kim ◽  
Kwang-Jin Kim ◽  
Hyeon Jung Kang ◽  
Young-Jin Son ◽  
Sik-Won Choi ◽  
...  

The number of patients with bone metabolic disorders including osteoporosis is increasing worldwide. These disorders often facilitate bone fractures, which seriously impact the patient’s quality of life and could lead to further health complications. Bone homeostasis is tightly regulated to balance bone resorption and formation. However, many anti-osteoporotic agents are broadly categorized as either bone forming or anti-resorptive, and their therapeutic use is often limited due to unwanted side effects. Therefore, safe and effective therapeutic agents are needed for osteoporosis. This study aims to clarify the bone protecting effects of oat bran water extract (OBWE) and its mode of action. OBWE inhibited RANKL-induced osteoclast differentiation by blocking c-Fos/NFATc1 through the alteration of I-κB. Furthermore, we found that OBWE enhanced BMP-2-stimulated osteoblast differentiation by the induction of Runx2 via Smad signaling molecules. In addition, the anti-osteoporotic activity of OBWE was also evaluated using an in vivo model. OBWE significantly restored ovariectomy-induced bone loss. These in vitro and in vivo results showed that OBWE has the potential to combat bone metabolic disorders including osteoporosis


Sign in / Sign up

Export Citation Format

Share Document