26 research outputs found

    Deletion of the glucocorticoid receptor chaperone FKBP51 prevents glucocorticoid-induced skin atrophy

    Get PDF
    FKBP51 (FK506-binding protein 51) is a known co-chaperone and regulator of the glucocorticoid receptor (GR), which usually attenuates its activity. FKBP51 is one of the major GR target genes in skin, but its role in clinical effects of glucocorticoids is not known. Here, we used FKBP51 knockout (KO) mice to determine FKBP51's role in the major adverse effect of topical glucocorticoids, skin atrophy. Unexpectedly, we found that all skin compartments (epidermis, dermis, dermal adipose and CD34+ stem cells) in FKBP51 KO animals were much more resistant to glucocorticoid-induced hypoplasia. Furthermore, despite the absence of inhibitory FKBP51, the basal level of expression and glucocorticoid activation of GR target genes were not increased in FKBP51 KO skin or CRISPR/Cas9-edited FKBP51 KO HaCaT human keratinocytes. FKBP51 is known to negatively regulate Akt and mTOR. We found a significant increase in AktSer473 and mTORSer2448 phosphorylation and downstream pro-growth signaling in FKBP51-deficient keratinocytes in vivo and in vitro. As Akt/mTOR-GR crosstalk is usually negative in skin, our results suggest that Akt/mTOR activation could be responsible for the lack of increased GR function and resistance of FKBP51 KO mice to the steroid-induced skin atrophy

    PPARĪ“ binding to heme oxygenase 1 promoter prevents angiotensin II-induced adipocyte dysfunction in Goldblatt hypertensive rats

    Get PDF
    Abstract: OBJECTIVE: Reninā€“angiotensin system (RAS) regulates adipogenic response with adipocyte hypertrophy by increasing oxidative stress. Recent studies have shown the role of peroxisome proliferator-activated receptor-d (PPARĪ“) agonist in attenuation of angiotensin II-induced oxidative stress. The aim of this study was to explore a potential mechanistic link between PPARĪ“ and the cytoprotective enzyme heme oxygenase-1 (HO-1) and to elucidate the contribution of HO-1 to the adipocyte regulatory effects of PPARĪ“ agonism in an animal model of enhanced RAS, the Goldblatt 2 kidney 1 clip (2K1C) model. METHOD: We first established a direct stimulatory effect of the PPARĪ“ agonist (GW 501516) on the HO-1 gene by demonstrating increased luciferase activity in COS-7 cells transfected with a luciferase-HO-1 promoter construct. Sprague-Dawley rats were divided into four groups: sham-operated animals, 2K1C rats and 2K1C rats treated with GW 501516, in the absence or presence of the HO activity inhibitor, stannous mesoporphyrin (SnMP). RESULTS: 2K1C animals had increased visceral adiposity, adipocyte hypertrophy, increased inflammatory cytokines, increased circulatory and adipose tisssue levels of renin and Ang II along with increased adipose tissue gp91 phox expression (Po0.05) when compared with sham-operated animals. Treatment with GW 501516 increased adipose tissue HO-1 and adiponectin levels (Po0.01) along with enhancement of Wnt10b and b-catenin expression. HO-1 induction was accompanied by the decreased expression of Wnt5b, mesoderm specific transcript (mest) and C/EBPa levels and an increased number of small adipocytes (Po0.05). These effects of GW501516 were reversed in 2K1C animals exposed to SnMP (Po0.05). CONCLUSION: Taken together, our study demonstrates, for the first time, that increased levels of Ang II contribute towards adipose tissue dysregulation, which is abated by PPARĪ“-mediated upregulation of the heme-HO system. These findings highlight the pivotal role and symbiotic relationship of HO-1, adiponectin and PPARĪ“ in the regulation of metabolic homeostasis in adipose tissues

    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

    FKBP51 controls cellular adipogenesis through p38 kinase-mediated phosphorylation of GRĪ± and PPARĪ³

    No full text
    Glucocorticoid receptor-Ī± (GRĪ±) and peroxisome proliferator-activated receptor-Ī³ (PPARĪ³) are critical regulators of adipogenic responses. We have shown that FK506-binding protein 51 (FKBP51) represses the Akt-p38 kinase pathway to reciprocally inhibit GRĪ± but stimulate PPARĪ³ by targeting serine 112 (PPARĪ³) and serines 220 and 234 (GRĪ±). Here, this mechanism is shown to be essential for GRĪ± and PPARĪ³ control of cellular adipogenesis. In 3T3-L1 cells, FKBP51 was a prominent marker of the differentiated state and knockdown of FKBP51 showed reduced lipid accumulation and expression of adipogenic genes. Compared with wild-type (WT), FKBP51 knockout (51KO) mouse embryonic fibroblasts (MEFs) showed dramatic resistance to differentiation, with almost no lipid accumulation and greatly reduced adipogenic gene expression. These features were rescued by reexpression of FKBP51 in 51KO cells. 51KO MEFs exhibited reduced fatty acid synthase activity, increased sensitivity to GRĪ±-induced lipolysis, and reduced PPARĪ³ activity at adipogenic genes (adiponectin, CD36, and perilipin) but elevated GRĪ± transrepression at these same genes. A p38 kinase inhibitor increased lipid content in WT cells and also restored lipid levels in 51KO cells, showing that elevated p38 kinase activity is a major contributor to adipogenic resistance in the 51KO cells. In 51KO cells, the S112A mutant of PPARĪ³ and the triple S212A/S220A/S234A mutant of GRĪ± both increased lipid accumulation, identifying these residues as targets of the FKBP51/p38 axis. Our combined investigations have uncovered FKBP51 as a key regulator of adipogenesis via the Akt-p38 pathway and as a potential target in the treatment of obesity and related disorders

    FKBP51 reciprocally regulates GRĪ± and PPARĪ³ activation via the Akt-p38 pathway

    No full text
    FK506-binding protein 51 (FKBP51) is a negative regulator of glucocorticoid receptor-Ī± (GRĪ±), although the mechanism is unknown. We show here that FKBP51 is also a chaperone to peroxisome proliferator-activated receptor-Ī³ (PPARĪ³), which is essential for activity, and uncover the mechanism underlying this differential regulation. In COS-7 cells, FKBP51 overexpression reduced GRĪ± activity at a glucocorticoid response element-luciferase reporter, while increasing PPARĪ³ activity at a peroxisome proliferator response element reporter. Conversely, FKBP51-deficient (knockout) (51KO) mouse embryonic fibroblasts (MEFs) showed elevated GRĪ± but reduced PPARĪ³ activities compared with those in wild-type MEFs. Phosphorylation is known to exert a similar pattern of reciprocal modulation of GRĪ± and PPARĪ³. Knockdown of FKBP51 in 3T3-L1 preadipocytes increased phosphorylation of PPARĪ³ at serine 112, a phospho-residue that inhibits activity. In 51KO cells, elevated phosphorylation of GRĪ± at serines 220 and 234, phospho-residues that promote activity, was observed. Because FKBP51 is an essential chaperone to the Akt-specific phosphatase PH domain leucine-rich repeat protein phosphatase, Akt signaling was investigated. Elevated Akt activation and increased activation of p38 kinase, a downstream target of Akt that phosphorylates GRĪ± and PPARĪ³, were seen in 51KO MEFs, causing activation and inhibition, respectively. Inactivation of p38 with PD169316 reversed the effects of FKBP51 deficiency on GRĪ± and PPARĪ³ activities and reduced PPARĪ³ phosphorylation. Last, loss of FKBP51 caused a shift of PPARĪ³ from cytoplasm to nucleus, as previously shown for GRĪ±. A model is proposed in which FKBP51 loss reciprocally regulates GRĪ± and PPARĪ³ via 2 complementary mechanisms: activation of Akt-p38-mediated phosphorylation and redistribution of the receptors to the nucleus for direct targeting by p38

    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

    The effect of TEL, LOS, and ROSI on osteoblastic phenotype of U-33/Ī³2 cells.

    No full text
    <p>Cells were treated for 3 days with either vehicle (DMSO) (V), or ROSI (R), or TEL (T), or LOS (L), or in combination (RT or RL). A. Enzymatic activity of alkaline phosphatase (ALP) after treatment with different doses of TEL or 1 ĀµM ROSI. B. Relative expression of osteoblast-specific transcription factors, Runx2 and Osterix, in cells treated with either 1 ĀµM ROSI, or 50 ĀµM TEL, or in combination. C. ALP activity in cells treated with either 1 ĀµM ROSI, or 50 ĀµM TEL, or in combination. D. ALP activity in cells treated with either 1 ĀµM ROSI, or 50 ĀµM LOS, or combination. ALP activity was normalized to the number of cells assessed in MTT proliferation assay (panels A, C, and D). * p<0.05 <i>vs</i>. vehicle.</p

    TEL effect on expression of members of TGFĪ² (A) and BMP (B) signaling pathways.

    No full text
    <p>U-33/Ī³2 cells were treated for 3 days with either vehicle (DMSO) (V), or 1 ĀµM ROSI (R), or 50 ĀµM TEL (T), or in combination (RT). * p<0.05 <i>vs</i>. vehicle.</p
    corecore