30 research outputs found

    Ī²-Catenin Directly Sequesters Adipocytic and Insulin Sensitizing Activities but Not Osteoblastic Activity of PPARĪ³2 in Marrow Mesenchymal Stem Cells

    Get PDF
    <div><p>Lineage allocation of the marrow mesenchymal stem cells (MSCs) to osteoblasts and adipocytes is dependent on both Wnt signaling and PPARĪ³2 activity. Activation of PPARĪ³2, an essential regulator of energy metabolism and insulin sensitivity, stimulates adipocyte and suppresses osteoblast differentiation and bone formation, and correlates with decreased bone mass and increased fracture rate. In contrast, activation of Wnt signaling promotes osteoblast differentiation, augments bone accrual and reduces total body fat. This study examined the cross-talk between PPARĪ³2 and Ī²-catenin, a key mediator of canonical Wnt signaling, on MSC lineage determination. Rosiglitazone-activated PPARĪ³2 induced rapid proteolytic degradation of Ī²-catenin, which was prevented by either inhibiting glycogen synthase kinase 3 beta (GSK3Ī²) activity, or blocking pro-adipocytic activity of PPARĪ³2 using selective antagonist GW9662 or mutation within PPARĪ³2 protein. Stabilization of Ī²-catenin suppressed PPARĪ³2 pro-adipocytic but not anti-osteoblastic activity. Moreover, Ī²-catenin stabilization decreased PPARĪ³2-mediated insulin signaling as measured by insulin receptor and FoxO1 gene expression, and protein levels of phosphorylated Akt (pAkt). Cellular knockdown of Ī²-catenin with siRNA increased expression of adipocyte but did not affect osteoblast gene markers. Interestingly, the expression of Wnt10b was suppressed by anti-osteoblastic, but not by pro-adipocytic activity of PPARĪ³2. Moreover, Ī²-catenin stabilization in the presence of activated PPARĪ³2 did not restore Wnt10b expression indicating a dominant role of PPARĪ³2 in negative regulation of pro-osteoblastic activity of Wnt signaling. In conclusion, Ī²-catenin and PPARĪ³2 are in cross-talk which results in sequestration of pro-adipocytic and insulin sensitizing activity. The anti-osteoblastic activity of PPARĪ³2 is independent of this interaction.</p> </div

    PPARĪ³2 mutation, abrogating the pro-adipocytic but not the anti-osteoblastic activity, protects Ī²-catenin protein from degradation.

    No full text
    <p>A. Western blot analysis of protein levels of non-mutated (WT) and mutated (D409A) forms of PPARĪ³2 analyzed 72 h after transfection of HEK293 cells. Ī²-actin was used as a loading control. Each lane was loaded with 50 Āµg of total protein lysate. EV ā€“ empty vector control. B. Western blot analysis of Ī²-catenin protein levels after treatment with 1 ĀµM Rosi for 72 h. Hek293 cells were transfected with Ī²-catenin expression construct and either empty expression vectors (pSPORT6 and pEF-BOS), or non-mutated (WT), or mutated (D409A) PPARĪ³2 expression constructs. Each lane was loaded with 50 Āµg of total protein lysate. C. Effect of D409A mutation on transcriptional activity of PPARĪ³2. Hek293 cells were transiently transfected with above constructs and co-transfected with p2AOx luciferase reporter gene construct under the control PPARĪ³ response elements. Cells were treated with either vehicle or 1 ĀµM Rosi for 48 h and lysates were analyzed for luciferase activity. Promoter activity of firefly luciferase was normalized to renilla luciferase which was used as a transfection control. D ā€“ G. Effect of D409A mutation on expression of adipocyte-specific (D) and osteoblast-specific (E ā€“ G) gene markers, and Wnt10b (H). U-33/c cells were transiently transfected with either empty vector (pEF-BOS), or non-mutated (WT), or mutated (D409A) PPARĪ³2 expression constructs and treated with either vehicle or 1 ĀµM Rosi for 72 h. Relative transcript levels were calculated as fold change as compared to vehicle treated cells in each transfection. V ā€“ vehicle; R- Rosi; * p<0.05 V <i>vs.</i> R.</p

    Stabilization of Ī²-catenin protein using LiCl does not affect PPARĪ³2 anti-osteoblastic activity.

    No full text
    <p>U-33/Ī³2 cells were treated with either vehicle, 1 ĀµM Rosi, 10 mM LiCl, or in combination for 72 h. A. Alkaline phosphatase activity. Bā€“D. Relative expression of osteoblast-specific gene markers and Wnt10b. Fold change in transcript levels was calculated as compared to vehicle treated cells. Statistical differences are shown between Rosi-treated samples and samples receiving combined treatment (NS ā€“ non-significant). V ā€“ vehicle; R ā€“ Rosi; L ā€“ LiCl; LR ā€“ LiCl+Rosi.</p

    Marrow Adipose Tissue: Skeletal Location, Sexual Dimorphism, and Response to Sex Steroid Deficiency

    No full text
    Marrow adipose tissue (MAT) is unique with respect to origin, metabolism, and function. MAT is characterized with high heterogeneity which correlates with skeletal location and bone metabolism. This fat depot is also highly sensitive to various hormonal, environmental, and pharmacologic cues to which it responds with changes in volume and/or metabolic phenotype. We have demonstrated previously that MAT has characteristics of both white (WAT) and brown (BAT)-like or beige adipose tissue, and that beige phenotype is attenuated with aging and in diabetes. Here, we extended our analysis by comparing MAT phenotype in different locations within a tibia bone of mature C57BL/6 mice and with respect to the presence of sex steroids in males and females. We report that MAT juxtaposed to trabecular bone of proximal tibia (pMAT) is characterized by elevated expression of beige fat markers including Ucp1, HoxC9, Prdm16, Tbx1, and Dio2, when compared with MAT located in distal tibia (dMAT). There is also a difference in tissue organization with adipocytes in proximal tibia being dispersed between trabeculae, while adipocytes in distal tibia being densely packed. Higher trabecular bone mass (BV/TV) in males correlates with lower pMAT volume and higher expression of beige markers in the same location, when compared with females. However, there is no sexual divergence in the volume and transcriptional profile of dMAT. A removal of ovaries in females resulted in decreased cortical bone mass and increased volume of both pMAT and dMAT, as well as volume of gonadal WAT (gWAT). Increase in pMAT volume was associated with marked increase in Fabp4 and Adiponectin expression and relative decrease in beige fat gene markers. A removal of testes in males resulted in cortical and trabecular bone loss and the tendency to increased volume of both pMAT and dMAT, despite a loss of gWAT. Orchiectomy did not affect the expression of white and beige adipocyte gene markers. In conclusion, expression profile of beige adipocyte gene markers correlates with skeletal location of active bone remodeling and higher BV/TV, however bone loss resulted from sex steroid deficiency is not proportional to MAT expansion at the same skeletal location

    The effect of Ī²-catenin silencing on the expression of adipocyte, osteoblast, and Wnt-signaling gene markers.

    No full text
    <p>U-33/Ī³2 cells were transiently transfected with either 200 ng of Ī²-cat siRNA, which consisted of four Ī²-catenin-specific 20ā€“25 nt oligonucleotides, or 200 ng of scrambled siRNA (sc siRNA) for negative control. Seventy two hours after transfection proteins and RNA were extracted. A. Western blot analysis of Ī²-catenin protein levels. Each lane was loaded with 50 Āµg of total protein lysate. Bā€“D. Analysis of gene expression of adipocyte-specific (B), osteoblast-specific (C), and Wnt signaling (D) gene markers. All values are expressed as fold change compared to control transfected with sc siRNA and represented by value 1. * p<0.05.</p

    GSK3Ī² antagonist LiCl protects Ī²-catenin protein from PPARĪ³2-mediated degradation and preserves Ī²-catenin transcriptional activity.

    No full text
    <p>U-33/Ī³2 cells were treated with either vehicle, 1 ĀµM Rosi, 10 mM LiCl, or in combination for 72 h. A. Western blot analysis of total levels of Ī²-catenin protein. Ī²-actin was used as a loading control. Each lane was loaded with 50 Āµg of total protein lysate. B. Relative expression of Ī²-catenin mRNA as compared to vehicle treated cells. C. Immunocytochemistry of Ī²-catenin protein. Green: Ī²-catenin; purple: DAPI staining of nuclei. D. Percentage of Ī²-catenin positive cells (T) and cells positive for Ī²-catenin in the nucleus (N). E. Transcriptional activity of Ī²-catenin measured in U-33/Ī³2 cells treated as above for 48 h using TOP-Flash construct in luciferase gene reporter assay. Promoter activity of firefly luciferase was normalized to renilla luciferase which was used as a transfection control (* p<0.05). V ā€“ vehicle; R ā€“ Rosi; L ā€“ LiCl; L+R or LR ā€“ LiCl+Rosi.</p

    Rosi-mediated activation of PPARĪ³2 degrades the pool of active, protein-unbound Ī²-catenin.

    No full text
    <p>A. Western blot analysis of protein-unbound (Active) and protein-bound (Inactive) fractions of Ī²-catenin isolated from U-33/Ī³2 cells treated with either vehicle (DMSO) or 1 ĀµM Rosi for 1 h. Protein loading per lane: 3 Āµg of protein-bound and 50 Āµg of protein-unbound fraction. B. Relative expression of Ī²-catenin mRNA analyzed after 1 h treatment of U-33/Ī³2 cells with either vehicle or 1 ĀµM Rosi. C. Western blot analysis of total Ī²-catenin protein levels isolated from U-33/c cells and U-33/Ī³2 cells treated with either vehicle or 1 ĀµM Rosi for 72 h. Each lane was loaded with 50 Āµg of total protein lysate. D. Relative expression of Ī²-catenin mRNA analyzed in U-33/c and U-33/Ī³2 cells after 72 h treatment with either vehicle or 1 ĀµM Rosi. Gene expression is presented as fold difference as compared to levels of Ī²-catenin transcript in vehicle treated U-33/c cells (* p<0.05). E. Immunofluorescent visualization of Ī²-catenin and PPARĪ³2 proteins in untreated U-33/Ī³2. V ā€“ vehicle; R ā€“ Rosi; A ā€“ active; I ā€“ inactive.</p

    Selective antagonist GW9662 of PPARĪ³2 pro-adipocytic activity increases Ī²-catenin protein stability.

    No full text
    <p>U-33/Ī³2 cells were treated with either vehicle, 1 ĀµM Rosi, 10 ĀµM GW9662, or in combination for 72 h. A. Adipocyte differentiation was assessed by measuring the number of Oil Red O positive cells. B ā€“ C. Relative expression of adipocyte-specific gene markers. D. Osteoblast differentiation was assessed by measuring alkaline phosphatase activity. E ā€“ G. Relative expression of osteoblast-specific gene markers and Wnt10b. Fold change in transcript levels was calculated as compared to vehicle treated cells. H. Immunocytochemistry of Ī²-catenin protein. Green: Ī²-catenin; purple: DAPI staining of nuclei. I. Percentage of Ī²-catenin positive cells (T) and cells positive for Ī²-catenin in the nucleus (N). J. Western blot analysis of total Ī²-catenin protein levels. Each lane was loaded with 50 Āµg of total protein lysate. K. Transcriptional activity of Ī²-catenin measured with luciferase gene reporter assay using TOP-Flash construct. Promoter activity of firefly luciferase was normalized to renilla luciferase which was used as a transfection control. Statistically significant differences are shown between Rosi-treated samples and samples receiving combined treatment (* p<0.05; NS ā€“ non-significant). V ā€“ vehicle; R ā€“ Rosi; G ā€“ GW9662; GR ā€“ GW9662+ Rosi.</p

    Partial Agonist, Telmisartan, Maintains PPARĪ³ Serine 112 Phosphorylation, and Does Not Affect Osteoblast Differentiation and Bone Mass

    No full text
    <div><p>Peroxisome proliferator activated receptor gamma (PPARĪ³) controls both glucose metabolism and an allocation of marrow mesenchymal stem cells (MSCs) toward osteoblast and adipocyte lineages. Its activity is determined by interaction with a ligand which directs posttranscriptional modifications of PPARĪ³ protein including dephosphorylation of Ser112 and Ser273, which results in acquiring of pro-adipocytic and insulin-sensitizing activities, respectively. PPARĪ³ full agonist TZD rosiglitazone (ROSI) decreases phosphorylation of both Ser112 and Ser273 and its prolonged use causes bone loss in part due to diversion of MSCs differentiation from osteoblastic toward adipocytic lineage. Telmisartan (TEL), an anti-hypertensive drug from the class of angiotensin receptor blockers, also acts as a partial PPARĪ³ agonist with insulin-sensitizing and a weak pro-adipocytic activity. TEL decreased <sup>S273</sup>pPPARĪ³ and did not affect <sup>S112</sup>pPPARĪ³ levels in a model of marrow MSC differentiation, U-33/Ī³2 cells. In contrast to ROSI, TEL did not affect osteoblast phenotype and actively blocked ROSI-induced anti-osteoblastic activity and dephosphorylation of <sup>S112</sup>pPPARĪ³. The effect of TEL on bone was tested side-by-side with ROSI. In contrast to ROSI, TEL administration did not affect bone mass and bone biomechanical properties measured by micro-indentation method and did not induce fat accumulation in bone, and it partially protected from ROSI-induced bone loss. In addition, TEL induced ā€œbrowningā€ of epididymal white adipose tissue marked by increased expression of UCP1, FoxC2, Wnt10b and IGFBP2 and increased overall energy expenditure. These studies point to the complexity of mechanisms by which PPARĪ³ acquires anti-osteoblastic and pro-adipocytic activities and suggest an importance of Ser112 phosphorylation status as being a part of the mechanism regulating this process. These studies showed that TEL acts as a full PPARĪ³ agonist for insulin-sensitizing activity and as a partial agonist/partial antagonist for pro-adipocytic and anti-osteoblastic activities. They also suggest a relationship between PPARĪ³ fat ā€œbrowningā€ activity and a lack of anti-osteoblastic activity.</p></div
    corecore