Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Bone ; 49(4): 662-72, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704206

ABSTRACT

Although androgen is considered an anabolic hormone, the consequences of androgen receptor (AR) overexpression in skeletally-targeted AR-transgenic lines highlight the detrimental effect of enhanced androgen sensitivity on cortical bone quality. A compartment-specific anabolic response is observed only in male and not in female AR3.6-transgenic (tg) mice, with increased periosteal bone formation and calvarial thickening. To identify anabolic signaling cascades that have the potential to increase bone formation, qPCR array analysis was employed to define expression differences between AR3.6-tg and wild-type (WT) periosteal tissue. Notably, categories that were significantly different between the two genotypes included axonal guidance, CNS development and negative regulation of Wnt signaling with a node centered on stem cell pathways. Further, fine mapping of AR3.6-tg calvaria revealed that anabolic thickening in vivo is not uniform across the calvaria, occurring only in frontal and in not parietal bones. Multipotent fraction 1 progenitor populations from both genotypes were cultured separately as frontal bone neural crest stem-like cells (fNCSC) and parietal bone mesenchymal stem-like cells (pMSC). Both osteoblastic and adipogenic differentiation in these progenitor populations was influenced by embryonic lineage and by genotype. Adipogenesis was enhanced in WT fNCSC compared to pMSC, but transgenic cultures showed strong suppression of lipid accumulation only in fNCSC cells. Osteoblastogenesis was significantly increased in transgenic fNCSC cultures compared to WT, with elevated alkaline phosphatase (ALP) activity and induction of mineralization and nodule formation assessed by alizarin red and von Kossa staining. Osteocalcin (OC) and ALP mRNA levels were also increased in fNCSC cultures from AR3.6-tg vs. WT, but in pMSC cultures ALP mRNA levels, mineralization and nodule formation were decreased in AR3.6-tg cells. Expression differences identified by array in long bone periosteal tissue from AR3.6-tg vs. WT were recapitulated in the fNCSC samples while pMSC profiles reflected cortical expression. These observations reveal the opposing effects of androgen signaling on lineage commitment and osteoblast differentiation that is enhanced in cells derived from a neural crest origin but inhibited in cells derived from a mesodermal origin, consistent with in vivo compartment-specific responses to androgen. Combined, these results highlight the complex action of androgen in the body that is dependent on the embryonic lineage and developmental origin of the cell. Further, these data these data suggest that the periosteum surrounding long bone is derived from neural crest.


Subject(s)
Adipocytes/drug effects , Adiposity/drug effects , Androgens/pharmacology , Bone and Bones/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Osteoblasts/drug effects , Adipocytes/cytology , Adipocytes/metabolism , Anabolic Agents/pharmacology , Animals , Bone and Bones/drug effects , Cells, Cultured , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Neural Crest/cytology , Neural Crest/drug effects , Neural Crest/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteogenesis/genetics , Periosteum/metabolism , Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Skull/drug effects , Skull/metabolism
2.
J Cell Biochem ; 112(7): 1773-86, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21381083

ABSTRACT

Androgens regulate body composition in youth and declining testosterone that occurs with aging is associated with muscle wasting, increased fat mass and osteopenia. Transgenic mice with targeted androgen receptor (AR) over-expression in mesenchymal stem cells (MSC) were generated to explore the role of androgen signaling in the regulation of body composition. Transgenic males, but not females, were shorter and have reduced body weight and visceral fat accumulation. Dual-energy X-ray absorptiometry (DXA) revealed significant reductions in fat mass with a reciprocal increase in lean mass, yet no difference in food consumption or locomotor activity was observed. Adipose tissue weight was normal in brown fat but reduced in both gonadal and perirenal depots, and reduced hyperplasia was observed with smaller adipocyte size in visceral and subcutaneous white adipose tissue. Although serum leptin, adiponectin, triglyceride, and insulin levels were no different between the genotypes, intraperitoneal glucose tolerance testing (IPGTT) showed improved glucose clearance in transgenic males. High levels of the AR transgene are detected in MSCs but not in mature fat tissue. Reduced fibroblast colony forming units indicate fewer progenitor cells resident in the marrow in vivo. Precocious expression of glucose transporter 4 (GLUT4), peroxisome proliferator-activated receptor γ (PPARγ), and CCAAT enhancer-binding protein α (C/EBPα) was observed in proliferating precursor cultures from transgenic mice compared to controls. In more mature cultures, there was little difference between the genotypes. We propose a mechanism where enhanced androgen sensitivity can alter lineage commitment in vivo to reduce progenitor number and fat development, while increasing the expression of key factors to promote smaller adipocytes with improved glucose clearance.


Subject(s)
Adipogenesis/genetics , Body Composition , Cell Lineage , Mesenchymal Stem Cells/physiology , Receptors, Androgen/genetics , Adiposity/genetics , Animals , Blood Glucose/genetics , Body Weight , Bone Marrow Cells/cytology , Cell Differentiation , Cell Size , Colony-Forming Units Assay , Eating , Female , Glucose Tolerance Test , Insulin/blood , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Transgenic , Motor Activity , Receptors, Androgen/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin/cytology , Testis/cytology
3.
Bone ; 46(3): 710-23, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19895913

ABSTRACT

Periosteal expansion is a recognized response to androgen exposure during bone development and in profoundly hypogonadal adults. However, androgen also suppresses endocortical bone formation, indicating that its effects on bone are dichotomous and envelope-specific. In fact, enhanced androgen signaling has been shown to have dramatic detrimental effects on whole bone biomechanical properties in two different transgenic models with skeletally targeted androgen receptor (AR) overexpression. As the mechanisms underlying this response are uncharacterized, we compared patterns of gene expression in periosteum-free cortical bone samples derived from AR-overexpressing transgenic male mice and their wild-type counterparts. We then assessed direct androgen effects in both wild-type and AR-overexpressing osteoblasts in primary culture. Among major signaling pathways associated with bone formation, focused quantitative RT-PCR (qPCR) array-based analysis of endocortical bone gene expression from wild-type vs. transgenic males identified the transforming growth factor-beta (TGF-beta) superfamily and bone morphogenetic protein (BMP) signaling as significantly altered by androgen in vivo. Bioinformatic analyses indicated proliferation, osteoblast differentiation and mineralization as major biological processes affected. Consistent with the in vivo array data and bioinformatic analyses, inhibition of differentiation observed with androgen exposure was reduced by exogenous BMP2 treatment of AR-overexpressing cultures to stimulate BMP signaling, confirming array pathway analysis. In addition, nonaromatizable dihydrotestosterone (DHT) inhibited osteoblast proliferation, differentiation and several indices of mineralization, including mineral accumulation and mineralized nodule formation in primary cultures from both wild-type and AR-transgenic mice. These findings identify a molecular mechanism based on altered BMP signaling that contributes to androgen inhibition of osteoblast differentiation and mineralization. Such detrimental effects of androgen on osteoblast function may underlie the generally disappointing results of androgen therapy.


Subject(s)
Androgens/physiology , Bone and Bones/physiology , Signal Transduction/physiology , Animals , Bone and Bones/cytology , Calcification, Physiologic/physiology , Cell Differentiation/physiology , Cells, Cultured , Male , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/physiology , Receptors, Androgen/biosynthesis
4.
Bone ; 43(3): 440-51, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18595795

ABSTRACT

Androgens are anabolic hormones that affect many tissues, including bone. However, an anabolic effect of androgen treatment on bone in eugonadal subjects has not been observed and clinical trials have been disappointing. The androgen receptor (AR) mediates biological responses to androgens. In bone tissue, both AR and the estrogen receptor (ER) are expressed. Since androgens can be converted into estrogen, the specific role of the AR in maintenance of skeletal homoeostasis remains controversial. The goal of this study was to use skeletally targeted overexpression of AR in differentiated osteoblasts as a means of elucidating the specific role(s) for AR transactivation in the mature bone compartment. Transgenic mice overexpressing AR under the control of the 2.3-kb alpha1(I)-collagen promoter fragment showed no difference in body composition, testosterone, or 17ss-estradiol levels. However, transgenic males have reduced serum osteocalcin, CTx and TRAPC5b levels, and a bone phenotype was observed. In cortical bone, high-resolution micro-computed tomography revealed no difference in periosteal perimeter but a significant reduction in cortical bone area due to an enlarged marrow cavity. Endocortical bone formation rate was also significantly inhibited. Biomechanical analyses showed decreased whole bone strength and quality, with significant reductions in all parameters tested. Trabecular morphology was altered, with increased bone volume comprised of more trabeculae that were closer together but not thicker. Expression of genes involved in bone formation and bone resorption was significantly reduced. The consequences of androgen action are compartment-specific; anabolic effects are exhibited exclusively at periosteal surfaces, but in mature osteoblasts androgens inhibited osteogenesis with detrimental effects on matrix quality, bone fragility and whole bone strength. Thus, the present data demonstrate that enhanced androgen signaling targeted to bone results in low bone turnover and inhibition of bone formation by differentiated osteoblasts. These results indicate that direct androgen action in mature osteoblasts is not anabolic, and raise concerns regarding anabolic steroid abuse in the developing skeleton or high-dose treatment in eugonadal adults.


Subject(s)
Androgens/metabolism , Bone and Bones/metabolism , Osteogenesis/physiology , Receptors, Androgen/metabolism , Animals , Cloning, Molecular , Collagen/metabolism , Female , Male , Mice , Mice, Transgenic , Osteoblasts/metabolism , Plasmids/metabolism , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Signal Transduction
5.
Bone ; 38(5): 637-51, 2006 May.
Article in English | MEDLINE | ID: mdl-16413235

ABSTRACT

Both the number and the activity of osteoblasts are critical for normal bone growth and maintenance. Although a potential role for estrogen in protection of bone mass through inhibition of osteoblast apoptosis has been proposed, a function for androgen is much less clear. The aim of this study was to establish a direct role for androgen to influence osteoblast apoptosis both in vitro and in vivo. AR-MC3T3-E1 cells, with androgen receptor (AR) overexpression controlled by the type I collagen promoter, were treated with the non-aromatizable androgen 5alpha-dihydrotestosterone (DHT). Apoptosis was assessed by three different techniques including DNA fragmentation, caspase-3 activation, and changes in mitochondrial membrane potential. Transactivation of AR by DHT enhanced apoptosis while 17beta-estradiol (E(2)) treatment reduced apoptosis in both proliferating preosteoblasts and mature osteocyte-like cells. To explore mechanism, the apoptosis regulators Bcl-2 (antiapoptotic) and Bax (proapoptotic) were evaluated. Western analysis revealed that DHT decreased Bcl-2 resulting in a significantly increased Bax/Bcl-2 ratio. Regulation of Bcl-2 was post-transcriptional since bcl-2 mRNA levels were unaffected by DHT treatment. Furthermore, ubiquitination of Bcl-2 was increased and serine phosphorylation was reduced, consistent with inhibition of MAP kinase signaling by DHT. Increased Bax/Bcl-2 ratio was essential since either Bcl-2 overexpression or Bax downregulation by RNA interference (RNAi) partially abrogated or reversed DHT-enhanced osteoblastic apoptosis. In order to establish physiologic significance in vivo, AR-transgenic mice with AR overexpression in the osteoblast lineage and thus enhanced androgen sensitivity were characterized. In male AR-transgenic mice, increased osteoblast apoptosis was observed in vivo even in association with new bone formation. Thus, although estrogen can be antiapoptotic, androgen stimulates osteoblast and osteocyte apoptosis through an increased Bax/Bcl-2 ratio even in anabolic settings. These results identify a new mechanism for androgen regulation of osteoblast activity distinct from estrogen, and suggest that enhanced apoptosis can be associated with anabolic stimulation of new bone growth. Androgens thus play a distinct role in skeletal homeostasis.


Subject(s)
Androgens/pharmacology , Apoptosis , Osteoblasts/drug effects , Osteocytes/drug effects , Proto-Oncogene Proteins/metabolism , bcl-2-Associated X Protein/metabolism , Androgens/physiology , Animals , Bone and Bones/cytology , Bone and Bones/drug effects , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Estradiol/physiology , Male , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/physiology , Osteocytes/cytology , Osteocytes/physiology , Phosphorylation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , RNA Interference , Receptors, Androgen/genetics , Serine/metabolism , Transcriptional Activation , Up-Regulation , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...