Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Eur J Haematol ; 105(3): 247-254, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32311143

ABSTRACT

BACKGROUND: Androgens function through DNA and non-DNA binding-dependent signalling of the androgen receptor (AR). How androgens promote erythropoiesis is not fully understood. DESIGN AND METHODS: To identify the androgen signalling pathway, we treated male mice lacking the second zinc finger of the DNA-binding domain of the AR (ARΔZF2 ) with non-aromatizable 5α-dihydrotestosterone (5α-DHT) or aromatizable testosterone. To distinguish direct hematopoietic and non-hematopoietic mechanisms, we performed bone marrow reconstitution experiments. RESULTS: In wild-type mice, 5α-DHT had greater erythroid activity than testosterone, which can be aromatized to estradiol. The erythroid response in wild-type mice following 5α-DHT treatment was associated with increased serum erythropoietin (EPO) and its downstream target erythroferrone, and hepcidin suppression. 5α-DHT had no erythroid activity in ARΔZF2 mice, proving the importance of DNA binding by the AR. Paradoxically, testosterone, but not 5α-DHT, suppressed EPO levels in ARΔZF2 mice, suggesting testosterone following aromatization may oppose the erythroid-stimulating effects of androgens. Female wild-type mice reconstituted with ARΔZF2 bone marrow cells remained responsive to 5α-DHT. In contrast, ARΔZF2 mice reconstituted with female wild-type bone marrow cells showed no response to 5α-DHT. CONCLUSION: Erythroid promoting effects of androgens are mediated through DNA binding-dependent actions of the AR in non-hematopoietic cells, including stimulating EPO expression.


Subject(s)
Androgens/metabolism , DNA-Binding Proteins/metabolism , Erythropoiesis , Receptors, Androgen/metabolism , Androgens/pharmacology , Animals , Biomarkers , Erythropoiesis/drug effects , Erythropoietin/blood , Female , Gene Expression Regulation , Iron/metabolism , Male , Mice , Mice, Transgenic , Protein Binding , Receptors, Androgen/genetics , Signal Transduction
2.
J Mol Endocrinol ; 57(2): 125-38, 2016 08.
Article in English | MEDLINE | ID: mdl-27402875

ABSTRACT

The aim of this study was to investigate the direct muscle cell-mediated actions of androgens by comparing two different mouse lines. The cre-loxP system was used to delete the DNA-binding activity of the androgen receptor (AR) in mature myofibers (MCK mAR(ΔZF2)) in one model and the DNA-binding activity of the AR in both proliferating myoblasts and myofibers (α-actin mAR(ΔZF2)) in another model. We found that hind-limb muscle mass was normal in MCK mAR(ΔZF2) mice and that relative mass of only some hind-limb muscles was reduced in α-actin mAR(ΔZF2) mice. This suggests that myoblasts and myofibers are not the major cellular targets mediating the anabolic actions of androgens on male muscle during growth and development. Levator ani muscle mass was decreased in both mouse lines, demonstrating that there is a myofiber-specific effect in this unique androgen-dependent muscle. We found that the pattern of expression of genes including c-myc, Fzd4 and Igf2 is associated with androgen-dependent changes in muscle mass; therefore, these genes are likely to be mediators of anabolic actions of androgens. Further research is required to identify the major targets of androgen actions in muscle, which are likely to include indirect actions via other tissues.


Subject(s)
Gene Deletion , Muscles/metabolism , Myoblasts/metabolism , Myofibrils/metabolism , Receptors, Androgen/genetics , Animals , Biomarkers , Gene Expression Regulation , Humans , Male , Mice , Mice, Knockout , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Organ Size , Organ Specificity/genetics , Physical Conditioning, Animal , Receptors, Androgen/metabolism
3.
Asian J Androl ; 16(5): 675-83, 2014.
Article in English | MEDLINE | ID: mdl-24713826

ABSTRACT

We aimed to determine the mechanisms of the anabolic actions of androgens in skeletal muscle by investigating potential androgen receptor (AR)-regulated genes in in vitro and in vivo models. The expression of the myogenic regulatory factor myogenin was significantly decreased in skeletal muscle from testosterone-treated orchidectomized male mice compared to control orchidectomized males, and was increased in muscle from male AR knockout mice that lacked DNA binding activity (AR(ΔZF2)) versus wildtype mice, demonstrating that myogenin is repressed by the androgen/AR pathway. The ubiquitin ligase Fbxo32 was repressed by 12 h dihydrotestosterone treatment in human skeletal muscle cell myoblasts, and c-Myc expression was decreased in testosterone-treated orchidectomized male muscle compared to control orchidectomized male muscle, and increased in AR(∆ZF2) muscle. The expression of a group of genes that regulate the transition from myoblast proliferation to differentiation, Tceal7 , p57(Kip2), Igf2 and calcineurin Aa, was increased in AR(∆ZF2) muscle, and the expression of all but p57(Kip2) was also decreased in testosterone-treated orchidectomized male muscle compared to control orchidectomized male muscle. We conclude that in males, androgens act via the AR in part to promote peak muscle mass by maintaining myoblasts in the proliferative state and delaying the transition to differentiation during muscle growth and development, and by suppressing ubiquitin ligase-mediated atrophy pathways to preserve muscle mass in adult muscle.


Subject(s)
Muscle Proteins/genetics , Myoblasts, Skeletal/metabolism , Myogenin/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , SKP Cullin F-Box Protein Ligases/genetics , Animals , Calcineurin/drug effects , Calcineurin/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p57/drug effects , Cyclin-Dependent Kinase Inhibitor p57/genetics , Dihydrotestosterone/pharmacology , Gene Expression/drug effects , Humans , In Vitro Techniques , Insulin-Like Growth Factor II/drug effects , Insulin-Like Growth Factor II/genetics , Male , Mice , Mice, Knockout , Muscle Proteins/drug effects , Muscle, Skeletal , Myoblasts, Skeletal/drug effects , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Orchiectomy , Proto-Oncogene Proteins c-myc/drug effects , RNA, Messenger/drug effects , SKP Cullin F-Box Protein Ligases/drug effects , Testosterone/pharmacology
4.
Endocr Res ; 39(3): 130-5, 2014.
Article in English | MEDLINE | ID: mdl-24467187

ABSTRACT

Androgens (testosterone and dihydrotestosterone) acting via the androgen receptor (AR) are required for male sexual differentiation, and also regulate the development of many other tissues including muscle, fat and bone. We previously generated an AR(lox) mouse line with exon 3 of the AR gene targeted by loxP sites. The deletion of exon 3 is in-frame, so only the DNA binding-dependent actions of the AR are deleted, but non-DNA binding-dependent actions are retained. This line also contained an antibiotic resistance selection cassette, neomycin (neo) in intron 3, which was also flanked by loxP sites. Hemizygous AR(lox) male mice demonstrated a phenotype of hyperandrogenization, with increased mass of androgen-dependent tissues. We hypothesized that this hyperandrogenization was likely to be due to the presence of the neo cassette. In this study, we have generated an AR(lox) neo-negative mouse line, using the EIIa-cre deleter mouse line to remove the neo cassette. Hemizygous AR(lox) neo-negative male mice have a normal phenotype, with normal body mass and normal mass of androgen-dependent tissues including the testis, seminal vesicles, kidney, spleen, heart and retroperitoneal fat. This neo-negative exon 3-targeted mouse line is the only floxed AR mouse line available to study the DNA binding-dependent actions of the AR in a tissue-specific manner, and is suitable for investigation in all tissues. This study demonstrates the importance of removing the selection cassette, which can potentially alter the phenotype of floxed mouse lines even in the absence of detectable effects on target gene expression.


Subject(s)
Gene Knockout Techniques/methods , Receptors, Androgen/genetics , Animals , DNA-Binding Proteins/metabolism , Female , Gene Deletion , Male , Mice, Inbred C57BL , Mice, Transgenic , Neomycin/metabolism , Phenotype , Receptors, Androgen/metabolism
5.
Gen Comp Endocrinol ; 193: 1-9, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23871650

ABSTRACT

Jawed vertebrates (Gnasthostomes) are broadly separated into cartilaginous fishes (Chondricthyes) and bony vertebrates (Osteichthyes). Cartilaginous fishes are divided into chimaeras (e.g. ratfish, rabbit fish and elephant shark) and elasmobranchs (e.g. sharks, rays and skates). Both cartilaginous fish and bony vertebrates are believed to have a common armoured bony ancestor (Class Placodermi), however cartilaginous fish are believed to have lost bone. This study has identified and investigated genes involved in skeletal development in vertebrates, in the cartilaginous fish, elephant shark (Callorhinchus milii). Ctnnb1 (ß-catenin), Sfrp (secreted frizzled protein) and a single Sost or Sostdc1 gene (sclerostin or sclerostin domain-containing protein 1) were identified in the elephant shark genome and found to be expressed in a number of tissues, including cartilage. ß-catenin was also localized in several elephant shark tissues. The expression of these genes, which belong to the Wnt/ß-catenin pathway, is required for normal bone formation in mammals. These findings in the cartilaginous skeleton of elephant shark support the hypothesis that the common ancestor of cartilaginous fishes and bony vertebrates had the potential for making bone.


Subject(s)
Fish Proteins/genetics , Gene Expression Regulation, Developmental , Sharks/growth & development , Sharks/genetics , Wnt Signaling Pathway/physiology , Animals , Cartilage/metabolism , Female , Fish Proteins/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Intracellular Signaling Peptides and Proteins , Male , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
6.
Mol Cell Endocrinol ; 348(1): 189-97, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-21872641

ABSTRACT

We tested the hypothesis that androgens have physiological actions via non-DNA binding-dependent androgen receptor (AR) signaling pathways in males, using our genetically modified mice that express a mutant AR with deletion of the 2nd zinc finger of the DNA binding domain (AR(ΔZF2)) that cannot bind DNA. In cultured genital skin fibroblasts, the mutant AR(ΔZF2) has normal ligand binding ability, phosphorylates ERK-1/2 in response to 1 min DHT treatment (blocked by the AR antagonist bicalutamide), but has reduced androgen-dependent nuclear localization compared to wildtype (WT). AR(ΔZF2) males have normal baseline ERK-1/2 phosphorylation, with a 1.5-fold increase in Akt phosphorylation in AR(ΔZF2) muscle vs WT. To identify physiological actions of non-DNA binding-dependent AR signaling, AR(ΔZF2) males were treated for 6 weeks with dihydrotestosterone (DHT). Cortical bone growth was suppressed by DHT in AR(ΔZF2) mice (6% decrease in periosteal and 7% decrease in medullary circumference vs untreated AR(ΔZF2) males). In conclusion, these data suggest that non-DNA binding dependent AR actions suppress cortical bone growth, which may provide a mechanism to fine-tune the response to androgens in bone.


Subject(s)
Androgens/physiology , DNA/metabolism , Gene Expression Regulation , Receptors, Androgen/metabolism , Androgens/pharmacology , Animals , Cell Nucleus/metabolism , Dihydrotestosterone/pharmacology , Femur/anatomy & histology , Femur/drug effects , Femur/metabolism , Gene Expression , Kidney/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Models, Biological , Muscle, Skeletal/metabolism , Organ Specificity , Protein Binding , Protein Transport , Receptors, Androgen/genetics , Response Elements , Sequence Deletion , Subcutaneous Fat/metabolism , Testis/metabolism
7.
Am J Physiol Endocrinol Metab ; 301(5): E767-78, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21712531

ABSTRACT

In men, as testosterone levels decrease, fat mass increases and muscle mass decreases. Increased fat mass in men, in particular central obesity, is a major risk factor for type 2 diabetes, cardiovascular disease, and all-cause mortality. Testosterone treatment has been shown to decrease fat mass and increase fat-free mass. We hypothesize that androgens act directly via the DNA binding-dependent actions of the androgen receptor (AR) to regulate genes controlling fat mass and metabolism. The aim of this study was to determine the effect of a global DNA binding-dependent (DBD) AR knockout (DBD-ARKO) on the metabolic phenotype in male mice by measuring body mass, fat mass, food intake, voluntary physical activity, resting energy expenditure, substrate oxidation rates, serum glucose, insulin, lipid, and hormone levels, and metabolic gene expression levels and second messenger protein levels. DBD-ARKO males have increased adiposity despite a decreased total body mass compared with wild-type (WT) males. DBD-ARKO males showed reduced voluntary activity, decreased food intake, increased serum leptin and adiponectin levels, an altered lipid metabolism gene profile, and increased phosphorylated CREB levels compared with WT males. This study demonstrates that androgens acting via the DNA binding-dependent actions of the AR regulate fat mass and metabolism in males and that the increased adiposity in DBD-ARKO male mice is associated with decreased voluntary activity, hyperleptinemia and hyperadiponectinemia and not with insulin resistance, increased food intake, or decreased resting energy expenditure.


Subject(s)
Adiposity/genetics , Insulin Resistance/genetics , Motivation/genetics , Motor Activity/genetics , Receptors, Androgen/genetics , Adiponectin/blood , Animals , DNA/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Eating/genetics , Eating/physiology , Insulin Resistance/physiology , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motivation/physiology , Protein Interaction Domains and Motifs/genetics , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Up-Regulation/genetics
8.
Am J Physiol Endocrinol Metab ; 301(1): E172-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21505150

ABSTRACT

The aim of this study is to determine if the Odc1 gene, which encodes ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine biosynthesis, is directly regulated by the androgen receptor (AR) in skeletal muscle myoblasts and if Odc1 regulates myoblast proliferation and differentiation. We previously showed that expression of Odc1 is decreased in muscle from AR knockout male mice. In this study, we show in vivo that Odc1 expression is also decreased >60% in muscle from male muscle-specific AR knockout mice. In normal muscle homeostasis, Odc1 expression is regulated by age and sex, reflecting testosterone levels, as muscle of adult male mice expresses high levels of Odc1 compared with age-matched females and younger males. In vitro, expression of Odc1 is 10- and 1.5-fold higher in proliferating mouse C(2)C(12) and human skeletal muscle myoblasts, respectively, than in differentiated myotubes. Dihydrotestosterone increases Odc1 levels 2.7- and 1.6-fold in skeletal muscle cell myoblasts after 12 and 24 h of treatment, respectively. Inhibition of ODC activity in C(2)C(12) myoblasts by α-difluoromethylornithine decreases myoblast number by 40% and 66% following 48 and 72 h of treatment, respectively. In contrast, overexpression of Odc1 in C(2)C(12) myoblasts results in a 27% increase in cell number vs. control when cells are grown under differentiation conditions for 96 h. This prolonged proliferation is associated with delayed differentiation, with reduced expression of the differentiation markers myogenin and Myf6 in Odc1-overexpressing cells. In conclusion, androgens act via the AR to upregulate Odc1 in skeletal muscle myoblasts, and Odc1 promotes myoblast proliferation and delays differentiation.


Subject(s)
Cell Proliferation , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/physiology , Ornithine Decarboxylase/genetics , Receptors, Androgen/physiology , Androgens/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Embryo, Mammalian , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Ornithine Decarboxylase/metabolism , Pregnancy , Receptors, Androgen/metabolism , Up-Regulation/drug effects
9.
J Cell Physiol ; 226(6): 1453-60, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21413019

ABSTRACT

The naturally occurring polyamines, spermidine, spermine, and their precursor putrescine, play indispensible roles in both prokaryotic and eukaryotic cells, from basic DNA synthesis to regulation of cell proliferation and differentiation. The rate-limiting polyamine biosynthetic enzymes, ornithine decarboxylase (ODC) and S-adenosylmethionine decarboxylase, are essential for mammalian development, with knockout of the genes encoding these enzymes, Odc1 and Amd1, causing early embryonic lethality in mice. In muscle, the involvement of polyamines in muscle hypertrophy is suggested by the concomitant increase in cardiac and skeletal muscle mass and polyamine levels in response to anabolic agents including ß-agonists. In addition to ß-agonists, androgens, which increase skeletal mass and strength, have also been shown to stimulate polyamine accumulation in a number of tissues. In muscle, androgens act via the androgen receptor to regulate expression of polyamine biosynthetic enzyme genes, including Odc1 and Amd1, which may be one mechanism via which androgens promote muscle growth. This review outlines the role of polyamines in proliferation and hypertrophy, and explores their possible actions in mediating the anabolic actions of androgens in muscle.


Subject(s)
Androgens/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Polyamines/metabolism , Animals , Cell Proliferation , Humans , Hypertrophy , Polyamines/chemistry
10.
J Endocrinol ; 206(1): 93-103, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20395380

ABSTRACT

We used our genomic androgen receptor (AR) knockout (ARKO) mouse model, in which the AR is unable to bind DNA to: 1) document gender differences between males and females; 2) identify the genomic (DNA-binding-dependent) AR-mediated actions in males; 3) determine the contribution of genomic AR-mediated actions to these gender differences; and 4) identify physiological genomic AR-mediated actions in females. At 9 weeks of age, control males had higher body, heart and kidney mass, lower spleen mass, and longer and larger bones compared to control females. Compared to control males, ARKO males had lower body and kidney mass, higher splenic mass, and reductions in cortical and trabecular bone. Deletion of the AR in ARKO males abolished the gender differences in heart and cortical bone. Compared with control females, ARKO females had normal body weight, but 14% lower heart mass and heart weight/body weight ratio. Relative kidney mass was also reduced, and relative spleen mass was increased. ARKO females had a significant reduction in cortical bone growth and changes in trabecular architecture, although with no net change in trabecular bone volume. In conclusion, we have shown that androgens acting via the genomic AR signaling pathway mediate, at least in part, the gender differences in body mass, heart, kidney, spleen, and bone, and play a physiological role in the regulation of cardiac, kidney and splenic size, cortical bone growth, and trabecular bone architecture in females.


Subject(s)
DNA/metabolism , Receptors, Androgen/physiology , Sex Characteristics , Signal Transduction/physiology , Androgens/physiology , Animals , Body Weight , Bone Development , Bone and Bones/anatomy & histology , Calcification, Physiologic , Female , Heart/anatomy & histology , Kidney/anatomy & histology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Receptors, Androgen/deficiency , Spleen/anatomy & histology
12.
Reprod Fertil Dev ; 20(8): 861-70, 2008.
Article in English | MEDLINE | ID: mdl-19007549

ABSTRACT

Spermatogenesis requires androgen but, paradoxically, oestradiol (E2) treatment stimulates spermatogenic development in gonadotrophin- and androgen-deficient hypogonadal (hpg) mice. The mechanisms of E2-induced spermatogenesis were investigated by determining intratesticular E2 levels and testis cell populations in E2-treated hpg male mice, and E2 spermatogenic actions were determined in androgen receptor-knockout (ARKO) mice. Despite increased serum E2 concentrations (150-300 pmol L(-1)), intratesticular E2 concentrations declined fivefold (P < 0.001) in E2-treated v. untreated hpg male mice. Serum FSH reached 40% of normal and total testicular numbers of known FSH-responsive Sertoli, spermatogonia and meiotic spermatocyte populations were significantly (P < 0.001) elevated 1.7-, 4- and 13-fold, respectively. However, E2 administration also increased androgen-dependent pachytene spermatocytes and post-meiotic spermatids to levels comparable with testosterone-treated hpg testes. Selective investigation of androgen receptor involvement used E2-treated ARKO mice, which were found to exhibit increased (1.6-fold; P < 0.05) intratesticular E2 concentrations and suppression of the elevated serum gonadotrophins, although FSH remained twofold higher than normal. However, testis size and total Sertoli, spermatogonia and spermatocyte numbers were not increased in E2-treated ARKO male mice. Therefore, E2-stimulated murine spermatogenic development occurs with markedly suppressed and not elevated intratesticular E2 levels and displays an absolute requirement for functional androgen receptors. We propose that this paradoxical E2 spermatogenic response is explained by predominantly extratesticular E2 actions, increasing FSH to combine with residual androgen activity in hpg testes to stimulate pre- to post-meiotic development.


Subject(s)
Estradiol/pharmacology , Receptors, Androgen/physiology , Spermatogenesis/drug effects , Testis/drug effects , Animals , Estradiol/metabolism , Follicle Stimulating Hormone/blood , Hypogonadism/physiopathology , Male , Mice , Mice, Knockout , Organ Size/drug effects , Receptors, Androgen/genetics , Sertoli Cells/cytology , Sertoli Cells/drug effects , Signal Transduction/physiology , Spermatogenesis/physiology , Spermatozoa/cytology , Spermatozoa/drug effects , Testis/cytology , Testis/metabolism , Testosterone/metabolism
13.
J Bone Miner Res ; 23(8): 1182-93, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18627265

ABSTRACT

It is well established that calcitonin is a potent inhibitor of bone resorption; however, a physiological role for calcitonin acting through its cognate receptor, the calcitonin receptor (CTR), has not been identified. Data from previous genetically modified animal models have recognized a possible role for calcitonin and the CTR in controlling bone formation; however, interpretation of these data are complicated, in part because of their mixed genetic background. Therefore, to elucidate the physiological role of the CTR in calcium and bone metabolism, we generated a viable global CTR knockout (KO) mouse model using the Cre/loxP system, in which the CTR is globally deleted by >94% but <100%. Global CTRKOs displayed normal serum ultrafiltrable calcium levels and a mild increase in bone formation in males, showing that the CTR plays a modest physiological role in the regulation of bone and calcium homeostasis in the basal state in mice. Furthermore, the peak in serum total calcium after calcitriol [1,25(OH)(2)D(3)]-induced hypercalcemia was substantially greater in global CTRKOs compared with controls. These data provide strong evidence for a biological role of the CTR in regulating calcium homeostasis in states of calcium stress.


Subject(s)
Hypercalcemia/prevention & control , Receptors, Calcitonin/metabolism , Acid Phosphatase/metabolism , Actins/metabolism , Animals , Calcitonin/blood , Calcitriol/pharmacology , Calcium/blood , Female , Femur/anatomy & histology , Femur/pathology , Gene Deletion , Gene Targeting , Hypercalcemia/metabolism , Integrases/metabolism , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Phenotype , Tartrate-Resistant Acid Phosphatase
14.
Bone ; 42(6): 1164-74, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18387351

ABSTRACT

Human parathyroid hormone (hPTH) is currently the only treatment for osteoporosis that forms new bone. Previously we described a fish equivalent, Fugu parathyroid hormone 1 (fPth1) which has hPTH-like biological activity in vitro despite fPth1(1-34) sharing only 53% identity with hPTH(1-34). Here we demonstrate the in vivo actions of fPth1(1-34) on bone. In study 1, young male rats were injected intermittently for 30 days with fPth1 [30 microg-1,000 microg/kg body weight (b.w.), (30fPth1-1,000fPth1)] or hPTH [30 microg-100 microg/kg b.w. (30hPTH-100hPTH)]. In proximal tibiae at low doses, the fPth1 was positively correlated with trabecular bone volume/total volume (TbBV/TV) while hPTH increased TbBV/TV, trabecular thickness (TbTh) and trabecular number (TbN). 500fPth1 and 1000fPth1 increased TbBV/TV, TbTh, TbN, mineral apposition rate (MAR) and bone formation rate/bone surface (BFR/BS) with a concomitant decrease in osteoclast surface and number. In study 2 ovariectomized (OVX), osteopenic rats and sham operated (SHAM) rats were injected intermittently with 500 microg/kg b.w. of fPth1 (500fPth1) for 11 weeks. 500fPth1 treatment resulted in increased TbBV/TV (151%) and TbTh (96%) in the proximal tibiae due to increased bone formation as assessed by BFR/BS (490%) and MAR (131%). The effect was restoration of TbBV/TV to SHAM levels without any effect on bone resorption. 500fPth1 also increased TbBV/TV and TbTh in the vertebrae (L6) and cortical thickness in the mid-femora increasing bone strength at these sites. fPth1 was similarly effective in SHAM rats. Notwithstanding the low amino acid sequence homology with hPTH (1-34), we have clearly established the efficacy of fPth1 (1-34) as an anabolic bone agent.


Subject(s)
Anabolic Agents/pharmacology , Bone Diseases, Metabolic/metabolism , Bone and Bones/drug effects , Ovariectomy , Parathyroid Hormone/pharmacology , Peptide Fragments/pharmacology , Takifugu , Anabolic Agents/therapeutic use , Animals , Biomarkers/metabolism , Bone Diseases, Metabolic/drug therapy , Bone and Bones/anatomy & histology , Bone and Bones/physiology , Female , Humans , Male , Osteogenesis/drug effects , Osteoporosis/drug therapy , Parathyroid Hormone/therapeutic use , Peptide Fragments/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Stress, Mechanical
15.
FASEB J ; 22(8): 2676-89, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18390925

ABSTRACT

To identify mechanisms of anabolic androgen action in muscle, we generated male and female genomic androgen receptor (AR) knockout (ARKO) mice, and characterized muscle mass, contractile function, and gene expression. Muscle mass is decreased in ARKO males, but normal in ARKO females. The levator ani muscle, which fails to develop in normal females, is also absent in ARKO males. Force production is decreased from fast-twitch ARKO male muscle, and slow-twitch muscle has increased fatigue resistance. Microarray analysis shows up-regulation of genes encoding slow-twitch muscle contractile proteins. Real-time PCR confirms that expression of genes encoding polyamine biosynthetic enzymes, ornithine decarboxylase (Odc1), and S-adenosylmethionine decarboxylase (Amd1), is reduced in ARKO muscle, suggesting androgens act through regulation of polyamine biosynthesis. Altered expression of regulators of myoblast progression from proliferation to terminal differentiation suggests androgens also promote muscle growth by maintaining myoblasts in the proliferate state and delaying differentiation (increased Cdkn1c and Igf2, decreased Itg1bp3). A similar pattern of gene expression is observed in orchidectomized male mice, during androgen withdrawal-dependent muscle atrophy. In conclusion, androgens are not required for peak muscle mass in females. In males, androgens act through the AR to regulate multiple gene pathways that control muscle mass, strength, and fatigue resistance.


Subject(s)
Muscle, Skeletal/growth & development , Muscle, Skeletal/physiopathology , Receptors, Androgen/deficiency , Androgens/physiology , Animals , Cell Differentiation , Cell Proliferation , Female , Gene Expression , Gene Regulatory Networks , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/pathology , Myoblasts, Skeletal/pathology , Myoblasts, Skeletal/physiology , Orchiectomy , Organ Size , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Sex Characteristics , Testis/physiology
16.
Physiol Genomics ; 33(1): 133-7, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18171720

ABSTRACT

We previously generated a conditional floxed mouse line to study androgen action, in which exon 3 of the androgen receptor (AR) gene is flanked by loxP sites, with the neomycin resistance gene present in intron 3. Deletion of exon 3 in global AR knockout mice causes androgen insensitivity syndrome, characterized by genotypic males lacking normal masculinization. We now report that male mice carrying the floxed allele (AR(lox)) have the reverse phenotype, termed hyperandrogenization. AR(lox) mice have increased mass of androgen-dependent tissues, including kidney, (P < 0.001), seminal vesicle (P < 0.001), levator ani muscle (P = 0.001), and heart (P < 0.05). Serum testosterone is not significantly different. Testis mass is normal, histology shows normal spermatogenesis, and AR(lox) males are fertile. AR(lox) males also have normal AR mRNA levels in kidney, brain, levator ani, liver, and testis. This study reaffirms the need to investigate the potential phenotypic effects of floxed alleles in the absence of cre in tissue-specific knockout studies. In addition, this androgen hypersensitivity model may be useful to further investigate the effects of subtle perturbations of androgen action in a range of androgen-responsive systems in the male.


Subject(s)
Hyperandrogenism/genetics , Loss of Heterozygosity/physiology , Receptors, Androgen/genetics , Animals , Body Weight/genetics , Crosses, Genetic , Female , Gene Expression Regulation/physiology , Heart/anatomy & histology , Integrases/genetics , Integrases/metabolism , Kidney/anatomy & histology , Liver/anatomy & histology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Size/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Testis/anatomy & histology , Testis/cytology , Testosterone/blood
17.
Am J Physiol Endocrinol Metab ; 291(3): E429-38, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16684850

ABSTRACT

Genetically modified mouse models have been used widely to advance our knowledge in the field of endocrinology and metabolism. A number of different approaches to generate genetically modified mice are now available, which provide the power to analyze the role of individual proteins in vivo. However, there are a number of points to be considered in the use and interpretation of these models. This review discusses the advantages and disadvantages involved in the generation and use of different genetically modified mouse models in endocrine research, including conventional techniques (e.g., overexpression, knockout, and knock-in models), tissue- and/or time-specific deletion of target genes [e.g., Cre-loxP and short interfering (si)RNA transgenic approaches], and gene-trap approaches to undertake functional genomics. This review also highlights the many factors that should be considered when assessing the phenotype of these mouse models, many of which are relevant to all murine physiological studies. These approaches are a powerful means by which to dissect the function of genes and are revolutionizing our understanding of endocrine physiology and metabolism.


Subject(s)
Endocrinology/methods , Models, Animal , Research Design , Animals , Endocrinology/trends , Forecasting , Mice , Mice, Knockout , Mice, Transgenic , Research/trends
18.
Am J Physiol Endocrinol Metab ; 291(3): E506-16, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16621900

ABSTRACT

Androgens promote anabolism in skeletal muscle; however, effects on subsequent muscle function are less well defined because of a lack of reliable experimental models. We established a rigorous model of androgen withdrawal and administration in male mice and assessed androgen regulation of muscle mass, structure, and function. Adult C57Bl/6J male mice were orchidectomized (Orx) or sham-operated (Sham) and received 10 wk of continuous testosterone (T) or control treatment (C) via intraperitoneal implants. Mass, fiber cross-sectional area (CSA), and in vitro contractile function were assessed for fast-twitch extensor digitorum longus (EDL) and slow-twitch soleus (SOL) muscles. After 10 wk, Orx+C mice had reduced body weight gain (P < 0.05), seminal vesicle mass (P < 0.01), and levator ani muscle mass (P < 0.001) compared with Sham+C mice, and these effects were prevented with testosterone treatment. Orx+T mice had greater EDL (P < 0.01) and SOL (P < 0.01) muscle mass compared with Orx+C mice; however, median fiber CSA was not significantly altered in these muscles. EDL and SOL muscle force was greater in Sham+T compared with Orx+C mice (P < 0.05) in proportion to muscle mass. Unexpectedly, Orx+T mice had increased fatigue resistance of SOL muscle compared with Orx+C mice (P < 0.001). We used a rigorous model of androgen withdrawal and administration in male mice to demonstrate an essential role of androgens in the maintenance of muscle mass and force. In addition, we showed that testosterone treatment increases resistance to fatigue of slow- but not fast-twitch muscle.


Subject(s)
Muscle Fatigue/drug effects , Muscle, Skeletal/drug effects , Muscular Atrophy/prevention & control , Orchiectomy , Testosterone/pharmacology , Animals , Body Weight/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Seminal Vesicles/drug effects , Seminal Vesicles/pathology , Testis/drug effects , Testis/pathology , Testosterone/blood
19.
Hum Mutat ; 27(5): 483-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16619235

ABSTRACT

We have characterized an unusual family with two different androgen receptor (AR) gene deletions, in which we propose a novel mechanism of deletion formation has occurred. Affected individuals have the X-linked disorder androgen insensitivity syndrome, and we previously showed that different family members have deletions of different exons of the AR gene. We have now fully sequenced the deletions from affected individuals, and confirmed the presence of different deletions in different affected family members. Most affected and heterozygote individuals have a 4,430-bp deletion of exon 5 that occurred between repeated GTGGCAT motifs in introns 4 and 5. One affected hemizygous individual has a 4,033-bp deletion of exons 6 and 7 that occurred between repeated CCTC motifs in introns 5 and 7. The intron 5 breakpoint junctions of the two deletions are only 11 bp apart. Surprisingly, the maternal grandmother of the original index case was found to be mosaic for both deletional events, as well as having the normal AR gene. Karyotyping ruled out 47,XXX trisomy, indicating triple mosaicism for the two different deleted AR alleles and a normal AR allele. This triple mosaicism must have occurred early in embryonic development, as both deletions were passed on to different children. Based on these findings, we propose a novel mechanism of deletion formation. We suggest that during AR gene replication, a double strand DNA break occurred in intron 5, and that a variant of replication slippage occurred on both newly synthesized strands between the repeat motifs of microhomology, leading to the formation of the two different AR gene deletions.


Subject(s)
DNA Repair , DNA Replication/physiology , Gene Deletion , Receptors, Androgen/genetics , Androgen-Insensitivity Syndrome/genetics , Chromosomes, Human, X , DNA Mutational Analysis , Exons , Female , Genetic Linkage , Heterozygote , Humans , Karyotyping , Male , Molecular Sequence Data , Mosaicism , Pedigree
20.
J Endocrinol ; 186(1): 21-31, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16002532

ABSTRACT

Androgen treatment can enhance the size and strength of muscle. However, the mechanisms of androgen action in skeletal muscle are poorly understood. This review discusses potential mechanisms by which androgens regulate satellite cell activation and function. Studies have demonstrated that androgen administration increases satellite cell numbers in animals and humans in a dose-dependent manner. Moreover, androgens increase androgen receptor levels in satellite cells. In vitro, the results are contradictory as to whether androgens regulate satellite cell proliferation or differentiation. IGF-I is one major target of androgen action in satellite cells. In addition, the possibility of non-genomic actions of androgens on satellite cells is discussed. In summary, this review focuses on exploring potential mechanisms through which androgens regulate satellite cells, by analyzing developments from research in this area.


Subject(s)
Androgens/physiology , Satellite Cells, Skeletal Muscle/metabolism , Aging/drug effects , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Humans , Hypogonadism/drug therapy , Hypogonadism/metabolism , Hypogonadism/pathology , Insulin-Like Growth Factor I/metabolism , Myoblasts/cytology , Receptors, Androgen/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...