Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biology (Basel) ; 13(1)2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38248470

ABSTRACT

Loss of function of the Retinoblastoma gene (RB1) due to mutations is commonly seen in human osteosarcomas. One of the Rb1 gene functions is to facilitate cell fate from mesenchymal stem cells to osteoblasts and prevent adipocyte differentiations. In this study, we demonstrate that a stable reduction of Rb1 expression (RbKD) in murine osteoblasts causes them to express higher levels of PPAR-É£ and other adipocyte-specific transcription factors while retaining high expression of osteoblast-specific transcription factors, Runx2/Cbfa1 and SP7/Osterix. Inhibition of gap junctional intercellular communication (GJIC) in osteoblasts is another mechanism that causes osteoblasts to transdifferentiate to adipocytes. We found that preosteoblasts exposed to osteoblast differentiating media (DP media) increased GJIC. RbKD cells showed reduced GJIC along with a reduction in expression of Cx43, the protein that mediates GJIC. Other membrane associated adhesion protein Cadherin 11 (Cad11) was also decreased. Since PPAR-É£ is increased with Rb1 loss, we wondered if the reduction of this transcription factor would reverse the changes observed. Reduction of PPAR-É£ in control osteoblasts slightly increased bone-specific expression and reduced adipocytic expression as expected along with an increase in Cad11 and Cx43 expression. GJIC remained high and was unaffected by a reduction in PPAR-É£ in control cells. Knockdown of PPAR-É£ in RbKD cells reduced adipocyte gene expression, while osteoblast-specific expression showed improvement. Cx43, Cad11 and GJIC remained unaffected by PPAR-É£ reduction. Our observations suggest that increased PPAR-É£ that happens with Rb1 loss only affects osteoblast-adipocyte-specific gene expression but does not completely reverse Cx43 gene expression or GJIC. Therefore, these effects may represent independent events triggered by Rb1loss and/or the differentiation process.

2.
Cell Reprogram ; 25(3): 109-120, 2023 06.
Article in English | MEDLINE | ID: mdl-37200520

ABSTRACT

Dental pulp stem cells (DPSCs) and stem cells from human exfoliated deciduous teeth (SHED) are a source of mesenchymal stem cells with the potential to differentiate into several cell types. We initially isolated SHED cells and compared their osteogenic capacity with commercially available DPSCs. Both cells exhibited similar capacities of growth and osteogenic differentiation. A fourfold to sixfold increase in endogenous microRNA26a (miR26a) expression during osteogenic differentiation of preosteoblasts and a similar but attenuated increase (twofold to fourfold) in differentiating SHED was observed, suggesting a role in the process. We, therefore, overexpressed miR26a in SHED to determine if the osteogenic differentiation capacity can be potentiated in vitro. SHED with a threefold increase in miR26a expression showed increased growth rate when compared with parent cells. When exposed to an osteogenic differentiating promoting medium, the miR26a overexpressing cells showed 100-fold increases in the expression of bone marker genes such as type 1 collagen, alkaline phosphatase, and Runx2. The mineralization capacity of these cells was also increased 15-fold. As miR26a targets regulate several bone-specific genes, we evaluated the effect of miR26a overexpression on established targets. We found a moderate decrease in SMAD1 and a profound decrease in PTEN expression. miR26a could potentiate its effect on osteoblast differentiation by its ability to inhibit PTEN and increase the viability of cells and their numbers, a process essential in osteoblast differentiation. Our studies suggest that the upregulation of miR26a can increase bone formation and may serve as an important target to further investigate its potential in tissue engineering applications.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Humans , Stem Cells , Cell Differentiation , Cells, Cultured , Osteoblasts , Cell Proliferation
3.
Biochem Biophys Rep ; 25: 100920, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33553686

ABSTRACT

In order to understand the role of the p53 tumor suppressor gene in microRNA expression during osteoblast differentiation, we used a screen to identify microRNAs that were altered in a p53-dependent manner. MicroRNAs from MC3T3-E1 preosteoblasts were isolated from day 0 (undifferentiated) and day 4 (differentiating) and compared to a p53 deficient MC3T3-E1 line treated similarly. Overall, one fourth of all the microRNAs tested showed a reduction of 0.6 fold, and a similar number of them were increased 1.7 fold with differentiation. P53 deficiency caused 40% reduction in expression of microRNAs in differentiating cells, while a small percent (0.03%) showed an increase. Changes in microRNAs were validated using real-time PCR and two microRNAs were selected for further analysis (miR-34b and miR-140). These two microRNAs were increased significantly during differentiation but showed a dramatic reduction in expression in a p53 deficient state. Stable expression of miR-34b and miR-140 in MC3T3-E1 cells resulted in decreases in cell proliferation rates when compared to control cells. There was a 4-fold increase in p53 levels with miR-34b expression and a less dramatic increase with miR-140. Putative target binding sites for bone specific transcription factors, Runx2 and Osterix, were found for miR-34b, while Runx2, beta catenin and type 1 collagen were found to be miR-140 targets. Western blot analyses and functional assays for the transcription factors Runx2, Osterix and Beta-catenin confirmed microRNA specific interactions. These studies provide evidence that p53 mediated regulation of osteoblast differentiation can also occur through specific microRNAs such as miR-34b and miR-140 that also directly target important bone specific genes.

4.
In Vitro Cell Dev Biol Anim ; 54(1): 71-84, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29197031

ABSTRACT

Liposarcoma is a malignant soft tissue tumor that originates from adipose tissue and is one of the most frequently diagnosed soft tissue sarcomas in humans. There is great interest in identifying novel chemotherapeutic options for treating liposarcoma based upon molecular alterations in the cancer cells. The Aurora kinases have been identified as promising chemotherapeutic targets based on their altered expression in many human cancers and cellular roles in mitosis and cytokinesis. In this study, we investigated the effects of an Aurora kinase A inhibitor (MK-5108), an Aurora kinase B inhibitor (AZD1152-HQPA), and a pan-Aurora kinase inhibitor (AMG 900) on undifferentiated SW-872 and well-differentiated 93T449 human liposarcoma cells. Treatment of the SW-872 and 93T449 cells with MK-5108 (0-1000 nM), AZD1152-HQPA (0-1000 nM), and AMG 900 (0-1000 nM) for 72 h resulted in a dose-dependent decrease in the total viable cell number. Based upon the EC50 values, the potency of the three Aurora kinase inhibitors in the SW-872 cells was as follows: AMG 900 (EC50 = 3.7 nM) > AZD1152-HQPA (EC50 = 43.4 nM) > MK-5108 (EC50 = 309.0 nM), while the potency in the 93T449 cells was as follows: AMG 900 (EC50 = 6.5 nM) > AZD1152-HQPA (EC50 = 74.5 nM) > MK-5108 (EC50 = 283.6 nM). The percentage of polyploidy after 72 h of drug treatment (0-1000 nM) was determined by propidium iodide staining and flow cytometric analysis. AMG 900 caused a significant increase in polyploidy starting at 25 nM in the SW-872 and 93T449 cells, and AZD1152-HQPA caused a significant increase starting at 100 nM in the SW-872 cells and 250 nM in the 93T449 cells. The Aurora kinase A inhibitor MK-5108 did not significantly increase the percentage of polyploid cells at any of the doses tested in either cell line. The expression of Aurora kinase A and B was evaluated in the SW-872 cells versus differentiated adipocytes and human mesenchymal stem cells by real-time RT-PCR and Western blot analysis. Aurora kinase A and B mRNA expression was significantly increased in the SW-872 cells versus the differentiated adipocytes and human mesenchymal stem cells. Western blot analysis revealed a ~ 48 kDa immunoreactive band for Aurora kinase A that was not present in the differentiated adipocytes or the human mesenchymal stem cells. A ~ 39 kDa immunoreactive band for Aurora kinase B was detected in the SW-872 cells, differentiated adipocytes, and human mesenchymal stem cells. A smaller immunoreactive band for Aurora kinase B was detected in the SW-872 cells but not in the differentiated adipocytes and human mesenchymal stem cells, and this may reflect the expression of a truncated splice variant of Aurora kinase B that has been associated with poor patient prognosis. The 93T449 cells demonstrated decreased expression of Aurora kinase A and B mRNA and protein compared to the SW-872 cells, and also expressed the truncated form of Aurora kinase B. The results of these in vitro studies indicate that Aurora kinase inhibitors should be further investigated as possible chemotherapeutic agents for human liposarcoma.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinases/antagonists & inhibitors , Cyclohexanecarboxylic Acids/pharmacology , Phthalazines/pharmacology , Thiazoles/pharmacology , Adipocytes/drug effects , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Aurora Kinase B/antagonists & inhibitors , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Aurora Kinases/genetics , Aurora Kinases/metabolism , Cell Differentiation , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liposarcoma , Mesenchymal Stem Cells/drug effects , Polyploidy , Protein Kinase Inhibitors/pharmacology
5.
Article in English | MEDLINE | ID: mdl-28670298

ABSTRACT

Osteoblasts and adipocytes originate from common mesenchymal progenitor cells and are controlled by specific transcription factors. While 1,25-dihydroxyvitamin D3 (vitamin D) is known to be an important factor for osteoblast differentiation, there are conflicting reports regarding its effect on adipogenesis. In this study, we attempted to understand the effect of exposure of preosteoblasts (MC3T3-E1) to adipogenic media with and without vitamin D and determined the expression of adipogenic genes during this process. Our studies show that while transdifferentiation of preosteoblasts occurred on exposure to adipogenic media, the effect of vitamin D treatment was synergistic resulting in several hundred fold increase in adipocyte transcription factors C/EBPα and peroxisome proliferator-activated receptor-gamma (P < 0.001) along with an increase in markers of adipogenesis and accumulation of lipid droplets in cells. Vitamin D treatment was also accompanied by 100-fold to 700-fold increases in vitamin D receptor expression during the treatment period (P < 0.001). To determine how the effect of vitamin D might compare to other genetic manipulations that promote adipogenic differentiation, we stably knocked down retinoblastoma expression in MC3T3-E1 cells. Recent studies have suggested retinoblastoma (Rb1) tumor suppressor gene function to be critical to maintain osteoblasts function and inhibit adipocyte differentiation. We exposed MC3T3-E1 cells with reduced Rb1 expression to adipogenic media and found an increase in adipogenic differentiation when compared to cells with a full complement of Rb dosage. However, the extent of the change was not as dramatic as seen with vitamin D. These studies show that preosteoblasts are sensitive and respond to these manipulations that favor the adipocytic phenotype. While vitamin D is not known to directly affect targets in adipogenesis, our observations may have resulted from the malleability of preosteoblast genome in MC3T3-E1 cells, which allowed adipocyte specific gene expression under appropriate stimuli. Why this pathway is influenced and subverted by an anabolic bone factor such as vitamin D remains to be determined.

6.
PLoS One ; 8(11): e82494, 2013.
Article in English | MEDLINE | ID: mdl-24312426

ABSTRACT

The tumor suppressor gene p53 is involved in a variety of cellular activities such as cellular stress responses, cell cycle regulation and differentiation. In our previous studies we have shown p53's transcription activating role to be important in osteoblast differentiation. There is still a debate in the literature as to whether p53 inhibits or promotes differentiation. We have found p53 heterozygous mice to show a p53 dependency on some bone marker gene expression that is absent in knockout mice. Mice heterozygous for p53 also show a higher incidence of osteosarcomas than p53 knockout mice. This suggests that p53 is able to modify the environment within osteoblasts. In this study we compare changes in gene expression resulting after either a transient or stable reduction in p53. Accordingly we reduced p53 levels transiently and stably in C2C12 cells, which are capable of both myoblast and osteoblast differentiation, and compared the changes in gene expression of candidate genes regulated by the p53 pathway. Using a PCR array to assay for p53 target genes, we have found different expression profiles when comparing stable versus transient knockdown of p53. As expected, several genes with profound changes after transient p53 loss were related to apoptosis and cell cycle regulation. In contrast, stable p53 loss produced a greater change in MyoD and other transcription factors with tissue specific roles, suggesting that long term loss of p53 affects tissue homeostasis to a greater degree than changes resulting from acute loss of p53. These differences in gene expression were validated by measuring promoter activity of different pathway specific genes involved in differentiation. These studies suggest that an important role for p53 is context dependent, with a stable reduction in p53 expression affecting normal tissue physiology more than acute loss of p53.


Subject(s)
Cell Differentiation/genetics , Gene Deletion , Gene Expression Profiling , Genes, p53 , Animals , Base Sequence , Cell Line , DNA Primers , Homeostasis , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction
7.
Am J Pharm Educ ; 77(8): 169, 2013 Oct 14.
Article in English | MEDLINE | ID: mdl-24159210

ABSTRACT

OBJECTIVE: To determine the impact of performing critical-thinking and reflection assignments within interdisciplinary learning teams in a biochemistry course on pharmacy students' and prospective health professions students' collaboration scores. DESIGN: Pharmacy students and prospective medical, dental, and other health professions students enrolled in a sequence of 2 required biochemistry courses. They were randomly assigned to interdisciplinary learning teams in which they were required to complete case assignments, thinking and reflection exercises, and a team service-learning project. ASSESSMENT: Students were asked to complete the Scale of Attitudes Toward Physician-Pharmacist Collaboration prior to the first course, following the first course, and following the second course. The physician-pharmacist collaboration scores of prospective health professions students increased significantly (p<0.001). CONCLUSIONS: Having prospective health professions students work in teams with pharmacy students to think and reflect in and outside the classroom improves their attitudes toward physician-pharmacist collaboration.


Subject(s)
Attitude of Health Personnel , Biochemistry , Interprofessional Relations , Pharmacists , Physicians , Students, Pharmacy/psychology , Thinking , Cooperative Behavior , Humans
8.
Biometals ; 26(1): 33-42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23080431

ABSTRACT

The E-cadherin/ß-catenin complex is a structural component of adherens-type junctions in epithelial cells. Moreover, ß-catenin acts as an intracellular signaling molecule that can influence the expression of a variety of genes that regulate apoptosis and cell cycle control. Cadmium (Cd) is an environmental toxicant that causes renal dysfunction and disrupts cadherin-dependent cell-cell adhesion in various types of epithelial cells. In this study, we examined the effects of Cd on the subcellular localization of ß-catenin, the cadherin/ß-catenin complex and ß-catenin-mediated gene transcription in rat proximal tubule NRK-52E cells. Exposure to 5-10 µM Cd for 4 h caused the NRK cells to separate from each other without killing the cells or causing them to detach from the growing surface. This effect was associated with the loss of ß-catenin and E-cadherin from the cell-cell contacts and apparent changes in the accumulation of ß-catenin in the nuclear cell subfraction. The expression of the ß-catenin-sensitive gene, c-jun was significantly increased in cells exposed to 5 µM Cd. However, there was no change in the expression of several other ß-catenin-regulated genes including: c-myc, cyclin D1 and matrilysin. Additional studies utilizing the TOPFLASH ß-catenin reporter gene construct showed that Cd caused a 2-3 fold increase in the expression of the luciferase reporter gene. Overall, these results indicate that Cd disrupts the cadherin/ß-catenin complex in NRK-52E cells, but this effect leads to only partial activation of ß-catenin-mediated gene transcription.


Subject(s)
Cadmium Chloride/toxicity , Environmental Pollutants/toxicity , beta Catenin/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Kidney/cytology , Lithium/pharmacology , Protein Transport/drug effects , Rats , Transcriptional Activation/drug effects , Transcriptome/drug effects , beta Catenin/genetics
9.
Biochem Biophys Res Commun ; 430(1): 370-4, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23149414

ABSTRACT

While Mdm2 is an important negative regulator of the p53 tumor suppressor, it also possesses p53-independent functions in cellular differentiation processes. Mdm2 expression is alternatively regulated by two P1 and P2 promoters. In this study we show that the P2-intiated transcription of Mdm2 gene is activated by 1,25-dihydroxy vitamin D3 in MC3T3 cells. By using P1 and P2-specific reporters, we demonstrate that only the P2-promoter responds to vitamin D treatment. We have further identified a potential vitamin D receptor responsive element proximal to the two p53 response elements within the Mdm2 P2 promoter. Using cell lines that are p53-temperature sensitive and p53-null, we show requirement of p53 for VDR-mediated up regulation of Mdm2 expression. Our results indicate that 1,25-dihydroxy vitamin D3 and its receptor have a role in the regulation of P2-initiated Mdm2 gene expression in a p53-dependent way.


Subject(s)
Calcitriol/metabolism , Gene Expression Regulation , Osteoblasts/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Vitamin D Response Element/genetics , Animals , Calcitriol/pharmacology , Cell Line , Mice , Osteoblasts/drug effects , Receptors, Calcitriol/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Vitamin D Response Element/drug effects
10.
Am J Pharm Educ ; 76(8): 150, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23129849

ABSTRACT

OBJECTIVE: To measure changes in pharmacy and medical students' physician-pharmacist collaboration scores resulting from a workshop designed to promote understanding of the others' roles in health care. METHODS: More than 88% of first-year pharmacy (n = 215) and medical (n = 205) students completed the Scale of Attitudes Toward Physician-Pharmacist Collaboration on 3 occasions in order to establish a baseline of median scores and to determine whether the scores were influenced by an interprofessional workshop. RESULTS: Participation in the interprofessional workshop increased pharmacy students' collaboration scores above baseline (p=0.02) and raised the scores of medical students on the education component of the collaboration survey instrument (p=0.015). The collaboration scores of pharmacy students greatly exceeded those of medical students (p<0.0001). CONCLUSION: A workshop designed to foster interprofessional understanding between pharmacy and medical students raised the physician-pharmacist collaboration scores of both. Crucial practical goals for the future include raising the collaboration scores of medical students to those of pharmacy students.


Subject(s)
Education, Medical/methods , Education, Pharmacy/methods , Students, Medical/psychology , Students, Pharmacy/psychology , Cooperative Behavior , Data Collection , Humans , Interprofessional Relations , Professional Role
11.
Exp Cell Res ; 318(8): 867-76, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22405968

ABSTRACT

Osteocalcin (OC) is a major noncollagenous bone matrix protein and an osteoblast marker whose expression is limited to mature osteoblasts during the late differentiation stage. In previous studies we have shown osteosarcomas to lose p53 function with a corresponding loss of osteocalcin gene expression. Introduction of wild type p53 resulted in re expression of the osteocalcin gene. Using gel shift and chromatin immunoprecipitation assays, we have identified a putative p53 binding site within the rat OC promoter region and observed an increase in OC promoter activity when p53 accumulates using a CAT assay. The p53 inducible gene Mdm2 is a well-known downstream regulator of p53 levels. Our results showed a synergistic increase in the OC promoter activity when both p53 and MDM2 were transiently overexpressed. We further demonstrate that p53 is not degraded during overexpression of MDM2 protein. Increased OC expression was observed with concomitantly increased p53, VDR, and MDM2 levels in ROS17/2.8 cells during treatment with differentiation promoting (DP) media, but was significantly decreased when co-treated with DP media and the small molecule inhibitor of MDM2-p53 interaction, Nutlin-3. We have also observed a dramatic increase of the OC promoter activity in the presence of p53 and Mdm2 with inclusion of Cbfa-1 and p300 factors. Our results suggest that under some physiological conditions the oncoprotein MDM2 may cooperate with p53 to regulate the osteocalcin gene during osteoblastic differentiation.


Subject(s)
Gene Expression Regulation , Osteoblasts/metabolism , Osteocalcin/genetics , Osteogenesis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein/pharmacology , Genes, p53 , Imidazoles/pharmacology , Osteocalcin/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/genetics , Rats , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
12.
Calcif Tissue Int ; 89(5): 411-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21964930

ABSTRACT

The tumor-suppressor p53 is a transcription factor that regulates a number of genes in the process of cell-cycle inhibition, apoptosis, and DNA damage. Recent studies have revealed a crucial role for p53 in bone remodeling. In our previous studies we have shown that p53 is an important regulator of osteoblast differentiation. In this study we investigated the role of p53 in the regulation of human osteocalcin gene expression. We observed that osteocalcin promoter activity could be upregulated by both exogenous and endogenous p53 and downregulated by p53-specific small interfering RNA. DNA affinity immunoblotting assay showed that p53 can bind to the human osteocalcin promoter in vitro. We further identified a p53 response element within the osteocalcin promoter region using a chromatin immunoprecipitation assay. Furthermore, we observed an additive effect of p53 and VDR on the regulation of osteocalcin promoter activity. Our findings suggest that p53 may directly target the human osteocalcin gene and positively affect osteocalcin gene expression.


Subject(s)
Gene Expression Regulation , Osteocalcin/genetics , Tumor Suppressor Protein p53/genetics , Animals , Base Sequence , Bone Remodeling/genetics , Cell Line, Tumor , Down-Regulation , Humans , Molecular Sequence Data , Osteocalcin/metabolism , Promoter Regions, Genetic , Rats , Transfection , Tumor Suppressor Protein p53/metabolism , Up-Regulation
13.
Porto Alegre; Artmed; 2011. 236 p.
Monography in Portuguese | LILACS, Coleciona SUS | ID: biblio-941460
15.
In Vitro Cell Dev Biol Anim ; 45(7): 361-70, 2009.
Article in English | MEDLINE | ID: mdl-19347411

ABSTRACT

Selective knockdown of phosphatase and tensin homolog (PTEN) has been recently shown to increase life long accumulation of bone and its ability to increase osteoblast lifespan. In order to determine how loss of PTEN function affects osteoblast differentiation, we created cell lines with stable knockdown of PTEN expression using short hairpin RNA vectors and characterized several clones. The effect of deregulated PTEN in osteoblasts was studied in relationship to cell proliferation and differentiation. Downregulation of PTEN initially affected the cell's attachment and spreading on plastic but cells recovered after a brief period of time. When cell proliferation was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, we noticed a small but significant increase in growth rates with PTEN reduction. The size of individual cells appeared larger when compared to control cells. Differentiation properties of these osteoblasts were increased as evidenced by higher expression of several of the bone markers tested (alkaline phosphatase, osteocalcin, osterix, bone morphogenetic protein 2, Cbfa1, osteoprotegerin, and receptor activator of NF-kappaB ligand) and their mineralization capacity in culture. As stabilization of beta-catenin is known to be responsible for growth deregulation with PTEN loss in other cell types, we investigated the activation of the canonical Wnt pathway in our cell lines. Immunofluorescence staining, protein expression in subcellular fractions for beta-catenin, and assays for activation of the canonical Wnt/beta-catenin signaling were studied in the PTEN downregulated cells. There was an overall decrease in beta-catenin expression in cells with PTEN knockdown. The distribution of beta-catenin was more diffuse within the cell in the PTEN-reduced clones when compared to controls where they were mostly present in cell borders. Signaling through the canonical pathway was also reduced in the PTEN knockdown cells when compared to control. The results of this study suggest that while decreased PTEN augments cell proliferation and positively affects differentiation, there is a decrease in beta-catenin levels and activity in osteoblasts. Therefore, at least in osteoblasts, beta-catenin is not responsible for mediating the activation of osteoblast differentiation with reduction in PTEN function.


Subject(s)
Down-Regulation , Osteoblasts/metabolism , PTEN Phosphohydrolase/genetics , beta Catenin/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation , Cell Shape , Osteoblasts/cytology , PTEN Phosphohydrolase/metabolism , Rats
16.
Cancer Cell Int ; 5: 24, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-16053526

ABSTRACT

BACKGROUND: This study was undertaken to examine the relationship between the tumor suppressor gene p53 and the nuclear signaling protein beta-catenin during bone differentiation. Cross talk between p53 and beta-catenin pathways has been demonstrated and is important during tumorigenesis and DNA damage, where deregulation of beta catenin activates p53. In this study, we used estrogen treatment of osteoblasts as a paradigm to study the relationship between the two proteins during osteoblast differentiation. RESULTS: We exposed osteoblast-like ROS17/2.8 cells to 17-beta estradiol (E2), in a short term assay, and studied the cellular distribution and expression of beta-catenin. We found beta-catenin to be up regulated several fold following E2 treatment. Levels of p53 and its functional activity mirrored the quantitative changes seen in beta-catenin. Alkaline phosphatase, an early marker of osteoblast differentiation, was increased in a manner similar to beta-catenin and p53. In order to determine if there was a direct relationship between alkaline phosphatase expression and beta-catenin, we used two different approaches. In the first approach, treatment with LiCl, which is known to activate beta-catenin, caused a several fold increase in alkaline phosphatase activity. In the second approach, transient transfection of wild type beta-catenin into osteoblasts increased alkaline phosphatase activity two fold over basal levels, showing that beta catenin expression can directly affect alkaline phosphatase expression. However increase in beta catenin activity was not associated with an increase in its signaling activity through TCF/LEF mediated transcription. Immunofluorescence analyses of p53 and beta-catenin localization showed that E2 first caused an increase in cytosolic beta-catenin followed by the accumulation of beta-catenin in the nucleus. Nuclear p53 localization was detected in several cells. Expression of p53 was accompanied by distribution of beta-catenin to the cytoplasm and cell borders. A sub population of cells staining strongly for both proteins appeared to be apoptotic. CONCLUSION: These results suggest that interactions between p53 and beta-catenin signaling pathways may play a key role in osteoblast differentiation and maintenance of tissue homeostasis.

17.
J Orthop Res ; 23(6): 1345-53, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15994055

ABSTRACT

We have previously shown p53 to have a specific role in osteoblast differentiation by its ability to regulate expression of certain bone specific proteins. In this study, we show mineralized matrix formation in vivo to be directly related to the presence of wild type p53 in osteoblastic osteosarcoma cells. In order to further understand the importance of p53 in differentiation, we investigated the relationship between p53 and Bone Morphogenetic Proteins (BMPs) (BMP 1, 2, 3A, 3B (GDF-10), 4, 5, 6, 7, 8A and 8B) during osteoblast differentiation. The expression of several BMPs were tested using RNase Protection Assay in differentiating ROS17/2.8 osteoblastic osteosarcoma cells. The expression of BMPs 1, 2, 3a, 3b and 7 showed time dependent modulation during in vitro differentiation. In order to determine if p53 has a role in this process, we used a murine osteosarcoma cell line stably expressing a temperature sensitive p53. Cells were exposed to ascorbic acid and glycerophosphates to hasten in vitro osteoblast differentiation and maintained either at 32 or 37 degrees C for expression of the wild type or mutant p53 phenotype. The expression of BMP-2, BMP-4 and BMP-7 were modulated in a p53 dependent fashion. We were able to confirm the p53 dependency of BMP-2 independently by RT-PCR. While BMP-2 expression was evident in the presence of both wild type and mutant p53, regulated expression was seen only in cells expressing wild type p53. Transient over expression of wild type p53 did not result in the same BMP-2 response as stable expression showing that the presence of p53 may be important for an orderly development of osteoblast differentiation rather than a direct effect on gene expression. The functional relationship between p53 and these bone specific markers is discussed.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation , Osteoblasts/cytology , Tumor Suppressor Protein p53/physiology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/genetics , Cell Line, Tumor , Osteocalcin/genetics , Osteosarcoma/chemistry , Osteosarcoma/pathology , Promoter Regions, Genetic , RNA, Messenger/analysis , Rats , Transfection , Transforming Growth Factor beta/genetics , Tumor Suppressor Protein p53/analysis
18.
Life Sci ; 75(17): 2045-55, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15312749

ABSTRACT

Estrogen is known to be anabolic for bone and we have used estrogen treatment as a paradigm to understand how p53 may affect osteoblast differentiation. In previous studies we have shown estrogen treatment to increase p53 functional activity in osteoblasts. Estrogen has been suggested to inhibit apoptosis in osteoblasts. Since the significance of a p53 increase during estrogen treatment is not apparent, we investigated the environment within osteoblasts after treatment with estrogen. We observed two peaks of p53 activity during continuous treatment of 17-[beta]-estradiol (E2) for 72h. The gene expression profile of different cell cycle regulators and apoptosis related genes at different times during treatment with 17-[beta]-estradiol were tested using gene arrays. There was an early increase in expression of several genes involved in apoptosis. This was followed by changes in expression of several genes involved in cell survival and stress response. The second peak of activity was associated with increase in expression of cell cycle regulators. Our results suggest that p53 activity may be a result of activation of several signaling pathways involving apoptosis, cell survival and cell cycle arrest. P53 may have a role in integrating these responses, which eventually results in cell cycle arrest and expression of differentiation markers.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Osteoblasts/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Survival/physiology , Chromatography, Thin Layer , Estradiol/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Osteoblasts/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...