Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
EBioMedicine ; 97: 104817, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37804569

RESUMO

BACKGROUND: Prostate cancer (PCa) patients treated with androgen deprivation therapy (ADT) have an increased fracture risk. Exploring biomarkers for early bone loss detection is of great interest. METHODS: Pre-planned substudy of the ARNEO-trial (NCT03080116): a double blind, randomised, placebo-controlled phase 2 trial performed in high-risk PCa patients without bone metastases between March 2019 and April 2021. Patients were 1:1 randomised to treatment with gonadotropin-releasing hormone antagonist (degarelix) + androgen receptor signalling inhibitor (ARSI; apalutamide) versus degarelix + matching placebo for 12 weeks prior to prostatectomy. Before and following ADT, serum and 24-h urinary samples were collected. Primary endpoints were changes in calcium-phosphate homeostasis and bone biomarkers. FINDINGS: Of the 89 randomised patients, 43 in the degarelix + apalutamide and 44 patients in the degarelix + placebo group were included in this substudy. Serum corrected calcium levels increased similarly in both treatment arms (mean difference +0.04 mmol/L, 95% confidence interval, 0.02; 0.06), and parathyroid hormone and 1,25-dihydroxyvitamin D3 levels decreased. Bone resorption markers increased, and stable calcium isotope ratios reflecting net bone mineral balance decreased in serum and urine similarly in both groups. INTERPRETATION: This exploratory substudy suggests that 12 weeks of ADT in non-metastatic PCa patients results in early bone loss. Additional treatment with ARSI does not seem to more negatively influence bone loss in the early phase. Future studies should address if these early biomarkers are able to predict fracture risk, and can be implemented in clinical practice for follow-up of bone health in PCa patients under ADT. FUNDING: Research Foundation Flanders; KU Leuven; University-Hospitals-Leuven.


Assuntos
Antagonistas de Androgênios , Neoplasias da Próstata , Masculino , Humanos , Antagonistas de Androgênios/efeitos adversos , Neoplasias da Próstata/patologia , Androgênios , Receptores Androgênicos , Cálcio , Antagonistas de Receptores de Andrógenos/efeitos adversos , Minerais/uso terapêutico , Homeostase , Biomarcadores
2.
Endocrinology ; 163(7)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35640239

RESUMO

Failure of bone mass maintenance in spite of functional loading is an important contributor to osteoporosis and related fractures. While the link between sex steroids and the osteogenic response to loading is well established, the underlying mechanisms are unknown, hampering clinical relevance. Androgens inhibit mechanoresponsiveness in male mice, but the cell type mediating this effect remains unidentified. To evaluate the role of neuronal sex steroid receptor signaling in the male bone's adaptive capacity, we subjected adult male mice with an extrahypothalamic neuron-specific knockout of the androgen receptor (N-ARKO) or the estrogen receptor alpha (N-ERαKO) to in vivo mechanical stimulation of the tibia. Loading increased cortical thickness in the control animals mainly through periosteal expansion, as total cross-sectional tissue area and cortical bone area but not medullary area were higher in the loaded than the unloaded tibia. Trabecular bone volume fraction also increased upon loading in the control group, mostly due to trabecular thickening. N-ARKO and N-ERαKO males displayed a loading response at both the cortical and trabecular bone compartments that was not different from their control littermates. In conclusion, we show that the presence of androgen receptor or estrogen receptor alpha in extrahypothalamic neurons is dispensable for the osteogenic response to mechanical loading in male mice.


Assuntos
Receptor alfa de Estrogênio , Receptores Androgênicos , Animais , Estudos Transversais , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Tíbia
3.
Endocrinology ; 162(6)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33674833

RESUMO

Testosterone (T) reduces male fat mass, but the underlying mechanisms remain elusive, limiting its clinical relevance in hypogonadism-associated obesity. Here, we subjected chemically castrated high-fat diet-induced adult obese male mice to supplementation with T or the nonaromatizable androgen dihydrotestosterone (DHT) for 20 weeks. Both hormones increased lean mass, thereby indirectly increasing oxygen consumption and energy expenditure. In addition, T but not DHT decreased fat mass and increased ambulatory activity, indicating a role for aromatization into estrogens. Investigation of the pattern of aromatase expression in various murine tissues revealed the absence of Cyp19a1 expression in adipose tissue while high levels were observed in brain and gonads. In obese hypogonadal male mice with extrahypothalamic neuronal estrogen receptor alpha deletion (N-ERαKO), T still increased lean mass but was unable to decrease fat mass. The stimulatory effect of T on ambulatory activity was also abolished in N-ERαKO males. In conclusion, our work demonstrates that the fat-burning action of T is dependent on aromatization into estrogens and is at least partially mediated by the stimulation of physical activity via extrahypothalamic ERα signaling. In contrast, the increase in lean mass upon T supplementation is mediated through the androgen receptor and indirectly leads to an increase in energy expenditure, which might also contribute to the fat-burning effects of T.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Receptor alfa de Estrogênio/fisiologia , Atividade Motora/fisiologia , Testosterona/farmacologia , Tecido Adiposo/metabolismo , Animais , Di-Hidrotestosterona/farmacologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Hipogonadismo/genética , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Atividade Motora/efeitos dos fármacos , Obesidade/genética , Obesidade/metabolismo , Condicionamento Físico Animal/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Congêneres da Testosterona/farmacologia
4.
Am J Physiol Endocrinol Metab ; 320(3): E415-E424, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33308013

RESUMO

Sex steroids are critical for skeletal development and maturation during puberty as well as for skeletal maintenance during adult life. However, the exact time during puberty when sex steroids have the highest impact as well as the ability of bone to recover from transient sex steroid deficiency is unclear. Surgical castration is a common technique to study sex steroid effects in rodents, but it is irreversible, invasive, and associated with metabolic and behavioral alterations. Here, we used a low dose (LD) or a high dose (HD) of gonadotropin-releasing hormone antagonist to either temporarily or persistently suppress sex steroid action in male mice, respectively. The LD group, a model for delayed puberty, did not show changes in linear growth or body composition, but displayed reduced trabecular bone volume during puberty, which fully caught up at adult age. In contrast, the HD group, representing complete pubertal suppression, showed a phenotype reminiscent of that observed in surgically castrated rodents. Indeed, HD animals exhibited severely impaired cortical and trabecular bone acquisition, decreased body weight and lean mass, and increased fat mass. In conclusion, we developed a rodent model of chemical castration that can be used as an alternative to surgical castration. Moreover, the transient nature of the intervention enables to study the effects of delayed puberty and reversibility of sex steroid deficiency.NEW & NOTEWORTHY We developed a rodent model of chemical castration, which can be used as an alternative to surgical castration. Moreover, the transient nature of the intervention enables to study the effects of delayed puberty and reversibility of sex steroid deficiency.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/fisiologia , Hormônios Esteroides Gonadais/deficiência , Hipogonadismo/patologia , Animais , Composição Corporal/efeitos dos fármacos , Desenvolvimento Ósseo/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Hormônios Esteroides Gonadais/farmacologia , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/farmacologia , Antagonistas de Hormônios/farmacologia , Hipogonadismo/complicações , Hipogonadismo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Orquiectomia , Maturidade Sexual/fisiologia , Fatores de Tempo
5.
Mol Cell Endocrinol ; 514: 110891, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32533994

RESUMO

Renal calcium and phosphate handling is an important contributor to mineral homeostasis and bone health and the androgen receptor (AR) is highly expressed in the kidney. We investigated the short term effects of androgen deprivation on renal calcium and phosphate reabsorption, independent of their effects on bone. Two weeks following orchidectomy (ORX) of adult mice, bone loss occurred along with hypercalciuria, which was similarly prevented by testosterone and dihydrotestosterone supplementation. Treatment with bisphosphonates prior to ORX also inhibited hypercalciuria, indicating that the calcium flux originated from the bone. Renal calcium and phosphate transporter expression was increased post-ORX, independent of bisphosphonates. Furthermore, androgen deprivation appeared to stimulate local synthesis of 1,25(OH)2D3. When bisphosphonate-treated mice were fed a low calcium diet, bone resorption was no longer blocked and secondary hyperparathyroidism developed, which was more pronounced in ORX mice than sham-operated mice. In conclusion, this study shows that androgen deprivation increased renal calcium and phosphate transporter expression, independent of bone, and underlines the importance of adequate intestinal calcium supply in circumstances of androgen deprivation and bisphosphonate treatment.


Assuntos
Androgênios/farmacologia , Cálcio da Dieta/farmacologia , Cálcio/metabolismo , Difosfonatos/farmacologia , Rim/efeitos dos fármacos , Fosfatos/metabolismo , Animais , Densidade Óssea/efeitos dos fármacos , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Dieta , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Orquiectomia , Urinálise
6.
FASEB J ; 34(5): 7118-7126, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32239553

RESUMO

Sexually dimorphic bone structure emerges largely during puberty. Sex steroids are critical for peak bone mass acquisition in both genders. In particular, the biphasic effects of estrogens mediate the skeletal sexual dimorphism. However, so far the stimulatory vs inhibitory actions of estrogens on bone mass are not fully explained by direct effects on bone cells. Recently, it has become evident that there is possible neuroendocrine action of estrogen receptor alpha (ERα) on the skeleton. Based on these considerations, we hypothesized that neuronal ERα-signaling may contribute to the skeletal growth during puberty. Here, we generated mice with tamoxifen-inducible Thy1-Cre mediated ERα inactivation during late puberty specifically in extrahypothalamic neurons (N-ERαKO). Inactivation of neuronal ERα did not alter the body weight in males, whereas N-ERαKO females exhibited a higher body weight and increased body and bone length compared to their control littermates at 16 weeks of age. Ex vivo microCT analysis showed increased radial bone expansion of the midshaft femur in female N-ERαKO along with higher serum levels of insulin-like growth factor (IGF)-1 as well as IGF-binding protein (IGFBP)-3. Furthermore, the 3-point bending test revealed increased bone strength in female N-ERαKO. In contrast, inactivation of neuronal ERα had no major effect on bone growth in males. In conclusion, we demonstrate that central ERα-signaling limits longitudinal bone growth and radial bone expansion specifically in females potentially by interacting with the GH/IGF-1 axis.


Assuntos
Desenvolvimento Ósseo/fisiologia , Receptor alfa de Estrogênio/metabolismo , Neurônios/metabolismo , Maturidade Sexual/fisiologia , Animais , Fenômenos Biomecânicos , Densidade Óssea/genética , Densidade Óssea/fisiologia , Desenvolvimento Ósseo/genética , Osso e Ossos/anatomia & histologia , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/fisiologia , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Caracteres Sexuais , Maturidade Sexual/genética , Transdução de Sinais , Microtomografia por Raio-X
7.
J Bone Miner Res ; 34(3): 508-519, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30496619

RESUMO

Androgens via the androgen receptor (AR) are required for optimal male bone health. The target cell(s) for the effects of androgens on cortical bone remain(s) incompletely understood. In females, estrogen receptor alpha in neurons is a negative regulator of cortical and trabecular bone. Whether neuronal AR regulates bone mass in males remains unexplored. Here, we inactivated AR in neurons using a tamoxifen-inducible CreERT2 under the control of the neuronal promoter Thy1. Tamoxifen induced a 70% to 80% reduction of AR mRNA levels in Thy1-CreERT2-positive brain regions cerebral cortex and brainstem as well as in the peripheral nervous tissue of male neuronal AR knockout (N-ARKO) mice. Hypothalamic AR mRNA levels were only marginally reduced and the hypothalamic-pituitary-gonadal axis remained unaffected, as determined by normal levels of serum testosterone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH). In contrast to orchidectomy, deletion of neuronal AR did not alter body weight, body composition, hindlimb muscle mass, grip strength, or wheel running. MicroCT analysis of the femur revealed no changes in bone accrual during growth in N-ARKO mice. However, 36- and 46-week-old N-ARKO mice displayed an accelerated age-related cortical involution, namely a more pronounced loss of cortical thickness and strength, which occurred in the setting of androgen sufficiency. Neuronal AR inactivation decreased the cancellous bone volume fraction in L5 vertebra but not in the appendicular skeleton of aging mice. MicroCT findings were corroborated in the tibia and after normalization of hormonal levels. Serum markers of bone turnover and histomorphometry parameters were comparable between genotypes, except for a 30% increase in osteoclast surface in the trabecular compartment of 36-week-old N-ARKO mice. Cortical bone loss in N-ARKO mice was associated with an upregulation of Ucp1 expression in brown adipose tissue, a widely used readout for sympathetic tone. We conclude that androgens preserve cortical integrity in aging male mice via AR in neurons. © 2018 American Society for Bone and Mineral Research.


Assuntos
Envelhecimento/patologia , Osso Cortical/patologia , Neurônios/metabolismo , Receptores Androgênicos/metabolismo , Animais , Composição Corporal , Peso Corporal , Reabsorção Óssea/patologia , Osso Esponjoso/patologia , Fêmur/patologia , Deleção de Genes , Gônadas/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculos/metabolismo , Osteogênese , Proteína Desacopladora 1/metabolismo
8.
Sci Rep ; 8(1): 957, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343749

RESUMO

Low testosterone (T) in men, especially its free fraction, has been associated with loss of energy. In accordance, orchidectomy (ORX) in rodents results in decreased physical activity. Still, the mechanisms through which T stimulates activity remain mostly obscure. Here, we studied voluntary wheel running behavior in three different mouse models of androgen deficiency: ORX, androgen receptor (AR) knock-out (ARKO) and sex hormone binding globulin (SHBG)-transgenic mice, a novel mouse model of "low free T". Our results clearly show a fast and dramatic action of T stimulating wheel running, which is not explained by its action on muscle, as evidenced by neuromuscular studies and in a muscle-specific conditional ARKO mouse model. The action of T occurs via its free fraction, as shown by the results in SHBG-transgenic mice, and it implies both androgenic and estrogenic pathways. Both gene expression and functional studies indicate that T modulates the in vivo sensitivity to dopamine (DA) agonists. Furthermore, the restoration of wheel running by T is inhibited by treatment with DA antagonists. These findings reveal that the free fraction of T, both via AR and indirectly through aromatization into estrogens, stimulates physical activity behavior in male mice by acting on central DA pathways.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Condicionamento Físico Animal/fisiologia , Testosterona/metabolismo , Androgênios/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/metabolismo , Atividade Motora/fisiologia , Orquiectomia/métodos , Receptores Androgênicos/metabolismo , Corrida/fisiologia
9.
Mol Cell Endocrinol ; 452: 57-63, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28504114

RESUMO

The selective estrogen receptor modulator tamoxifen exerts estrogen agonistic or antagonistic actions on several tissues, including bone. The off-target effects of tamoxifen are one of the most widely recognized pitfalls of tamoxifen-inducible Cre recombinases (CreERs), potentially confounding the phenotypic findings. Still, the validation of tamoxifen induction schemes that minimize the side effects of the drug has not been addressed. Here, we compared the side effects on the skeleton and other androgen-responsive targets of a shortened tamoxifen regimen (2 doses of 190 mg/kg body weight by oral gavage) to a standard protocol (4 doses) and determined their efficiency in inducing CreER-mediated gene deletion. In addition, both a vehicle- and a 10-dose group, which served as a positive control for tamoxifen side effects, were also included. For this purpose, we generated male mice with a floxed androgen receptor (AR) and a neuron-specifically expressed CreER. Treatment with two doses of tamoxifen was the only regimen that did not diminish androgenic bioactivity, as assessed by both seminal vesicles and levator ani/bulbocavernosus muscle weights and serum testosterone concentrations. Similarly, trabecular and cortical femoral bone structure were dramatically altered by both the standard and high-dose protocols but not by the shortened version. Serum osteocalcin and bone-gene expression analyses confirmed the absence of effects on bone by 2 doses of tamoxifen. This protocol decreased AR mRNA levels efficiently and specifically in the nervous system. Thus, we optimized a protocol for tamoxifen-induced CreER gene deletion in mice without off-target effects on bone and male reproductive organs.


Assuntos
Osso e Ossos/efeitos dos fármacos , Deleção de Genes , Técnicas de Inativação de Genes , Integrases/metabolismo , Recombinação Genética/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Análise de Variância , Animais , Peso Corporal/efeitos dos fármacos , Osso Esponjoso/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/agonistas , Integrases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteocalcina/sangue , Osteocalcina/genética , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Glândulas Seminais , Tamoxifeno/administração & dosagem , Tamoxifeno/efeitos adversos , Testosterona/sangue , Fatores de Tempo
10.
Mol Cell Endocrinol ; 411: 198-206, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25958043

RESUMO

Androgen deficiency or androgen receptor knockout (ARKO) causes high-turnover osteopenia, but the target cells for this effect remain unclear. To examine whether AR in osteoclasts directly suppresses bone resorption, we crossed AR-floxed with cathepsin K-Cre mice. Osteoclast-specific ARKO (ocl-ARKO) mice showed no changes neither in osteoclast surface nor in bone microarchitecture nor in the response to orchidectomy and androgen replacement, indicating that the AR in osteoclasts is not critical for bone maintenance. In line with the lack of a bone phenotype, the levels of AR were very low in osteoclast-enriched cultures derived from bone marrow (BM) and undetectable in osteoclasts generated from spleen precursors. Since tibiae of ubiquitous ARKO mice displayed increased osteoclast counts, the role of AR was further explored using cell cultures from these animals. Osteoclast generation and activity in vitro were similar between ARKO and wildtype control (WT) mice. In co-culture experiments, BM stromal cells (BMSCs) were essential for the suppressive action of AR on osteoclastogenesis and osteoclast activity. Stimulation with 1,25(OH)2 vitamin D3 increased Rankl and decreased Tnfsf11 (osteoprotegerin, Opg) gene expression in BMSCs more than in osteoblasts. This increase in the Rankl/Opg ratio following 1,25(OH)2D3 stimulation was lower, not higher, in ARKO mice. Runx2 expression in BMSCs was however higher in ARKO vs. WT, suggesting that ARKO mice may more readily commit osteoprogenitor cells to osteoblastogenesis. In conclusion, the AR does not seem to suppress bone resorption through direct actions in osteoclasts. BMSCs may however represent an alternative AR target in the BM milieu.


Assuntos
Reabsorção Óssea/metabolismo , Osteoclastos/metabolismo , Receptores Androgênicos/metabolismo , Fosfatase Ácida/sangue , Animais , Células Cultivadas , Fêmur/citologia , Fêmur/metabolismo , Isoenzimas/sangue , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Knockout , Osteocalcina/sangue , Osteoclastos/citologia , Receptores Androgênicos/genética , Fosfatase Ácida Resistente a Tartarato , Tíbia/citologia , Tíbia/metabolismo
11.
Endocrinology ; 156(4): 1343-53, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25654322

RESUMO

Androgens are well known to enhance exercise-induced muscle hypertrophy; however, whether androgens also influence bone's adaptive response to mechanical loading remains unclear. We studied the adaptive osteogenic response to unilateral in vivo mechanical loading of tibia in adult male mice in both a long- and a short-term experimental set-up. Mice were divided into four groups: sham operated, orchidectomized (ORX), T (ORX+T), or nonaromatizable dihydrotestosterone (ORX+DHT) replacement. Significant interactions between androgen status and osteogenic response to mechanical loading were observed. Cortical thickness increased by T (0.14 vs 0.11 mm sham, P<.05) and DHT (0.17 vs 0.11 mm sham, P<.05). However, T partially (+36%) and DHT completely (+10%) failed to exhibit the loading-related increase observed in sham (+107%) and ORX (+131%, all P<.05) mice. ORX decreased periosteal bone formation, which was restored to sham levels by T and DHT. However, both androgens completely suppressed the loading-related increase in periosteal bone formation. Short-term loading decreased the number of sclerostin-positive osteocytes in sham, whereas in control fibulas, ORX decreased and T increased the number of sclerostin-positive osteocytes. Loading no longer down-regulated sclerostin in the ORX or T groups. In conclusion, both T and DHT suppress the osteogenic response to mechanical loading.


Assuntos
Androgênios/farmacologia , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Di-Hidrotestosterona/farmacologia , Osteogênese/fisiologia , Testosterona/farmacologia , Suporte de Carga/fisiologia , Animais , Densidade Óssea/fisiologia , Osso e Ossos/fisiologia , Masculino , Camundongos , Orquiectomia , Osteogênese/efeitos dos fármacos
12.
J Bone Miner Res ; 27(12): 2535-43, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22836391

RESUMO

Androgens play a key role in the maintenance of male skeletal integrity. The regulation of this integrity by androgen receptor (AR) signaling has been mainly attributed to osteoblasts. Although osteocytes have emerged as key regulators of bone remodeling, the influence of sex steroids on these cells has been poorly studied. We aimed to investigate the role of AR signaling, specifically in osteocytes using the Cre/LoxP system in male mice (driven by dentin matrix protein 1 [ocy-ARKOs]). Osteocyte fractions of control (AR(ex2)/Y) and ocy-ARKO (ARflox(ex2)/Y; DMP1-cre) mice isolated through sequential collagenase digestion showed increasing AR expression toward the mature osteocyte fraction of control males compared with the more immature fractions, whereas this was reduced by >80% in ocy-ARKO osteocytes. The skeletal phenotype of mutant mice was further assessed by histomorphometry and quantitative micro-computed tomography at 12 and 32 weeks of age. Ocy-ARKOs had significantly lower trabecular bone volume and number in femora and tibias at 32 weeks as well as decreased trabecular number in the L(5) vertebra at 12 weeks. Biomechanical testing showed that ocy-ARKO femora were also stiffer and required a lower ultimate force to induce failure at 32 weeks. However, femoral cortical structure was not significantly different at any time point. The absence of AR in osteocyte also did not appear to affect trabecular bone formation nor its response to mechanical loading. In conclusion, selective inactivation of the AR in osteocytes of male mice accelerates age-related deterioration of skeletal integrity. These findings provide evidence for a direct role of androgens in the maintenance of trabecular bone through actions of the AR in osteocytes.


Assuntos
Osteócitos/metabolismo , Receptores Androgênicos/fisiologia , Animais , Fenômenos Biomecânicos , Doenças Ósseas/fisiopatologia , Osso e Ossos/patologia , Proteínas da Matriz Extracelular/genética , Fêmur/patologia , Fêmur/fisiologia , Masculino , Camundongos , Camundongos Knockout , Osteócitos/efeitos dos fármacos , Receptores Androgênicos/deficiência , Tíbia/patologia
13.
Mol Cancer Ther ; 5(9): 2211-7, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16985054

RESUMO

Because of its ability to mimic a low energy status of the cell, the cell-permeable nucleoside 5-aminoimidazole-4-carboxamide (AICA) riboside was proposed as an antineoplastic agent switching off major energy-consuming processes associated with the malignant phenotype (lipid production, DNA synthesis, cell proliferation, cell migration, etc.). Key to the antineoplastic action of AICA riboside is its conversion to ZMP, an AMP mimetic that at high concentrations activates the AMP-activated protein kinase (AMPK). Here, in an attempt to increase the efficacy of AICA riboside, we pretreated cancer cells with methotrexate, an antimetabolite blocking the metabolism of ZMP. Methotrexate enhanced the AICA riboside-induced accumulation of ZMP and led to a decrease in the levels of ATP, which functions as an intrasteric inhibitor of AMPK. Consequently, methotrexate markedly sensitized AMPK for activation by AICA riboside and potentiated the inhibitory effects of AICA riboside on tumor-associated processes. As cotreatment elicited antiproliferative effects already at concentrations of compounds that were only marginally effective when used alone, our findings on the cooperation between methotrexate and AICA riboside provide new opportunities both for the application of classic antimetabolic chemotherapeutics, such as methotrexate, and for the exploitation of the energy-sensing machinery as a target for cancer intervention.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Neoplasias da Mama/tratamento farmacológico , Carcinoma de Células Escamosas/tratamento farmacológico , Metotrexato/farmacologia , Ribonucleosídeos/farmacologia , Proteínas Quinases Ativadas por AMP , Trifosfato de Adenosina/metabolismo , Aminoimidazol Carboxamida/antagonistas & inibidores , Aminoimidazol Carboxamida/metabolismo , Aminoimidazol Carboxamida/farmacocinética , Aminoimidazol Carboxamida/farmacologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , DNA de Neoplasias/antagonistas & inibidores , DNA de Neoplasias/biossíntese , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Humanos , Lipídeos/biossíntese , Complexos Multienzimáticos/metabolismo , Nucleotídeo Desaminases/antagonistas & inibidores , Nucleotídeo Desaminases/genética , Nucleotídeo Desaminases/metabolismo , Fosforribosilaminoimidazolcarboxamida Formiltransferase/antagonistas & inibidores , Fosforribosilaminoimidazolcarboxamida Formiltransferase/genética , Fosforribosilaminoimidazolcarboxamida Formiltransferase/metabolismo , Fosforribosilglicinamido Formiltransferase/antagonistas & inibidores , Fosforribosilglicinamido Formiltransferase/genética , Fosforribosilglicinamido Formiltransferase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Purinas/antagonistas & inibidores , Purinas/biossíntese , Interferência de RNA , Ribonucleosídeos/farmacocinética , Ribonucleotídeos/antagonistas & inibidores , Ribonucleotídeos/metabolismo
14.
J Steroid Biochem Mol Biol ; 98(2-3): 164-73, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16388947

RESUMO

It remains unclear why it has proven so difficult to identify androgen target genes in cultured Sertoli cells. Given the lack of useful endogenous reporter genes, we studied the androgen and glucocorticoid responsiveness of these cells by transfection with three different steroid-responsive reporter constructs. The constructs were driven by the tyrosine aminotransferase steroid-responsive region (TAT-GRE4x-Luc), the mouse mammary tumor virus promoter (MMTV-Luc) and the Pem homeobox gene proximal promoter respectively (Pem-Luc). These constructs can be activated either by both the glucocorticoid receptor (GR) and the androgen receptor (AR) (TAT-GRE4x-Luc and MMTV-Luc) or selectively by the AR (Pem-Luc). Despite high transfection efficiency (30-40%) none of the constructs could be activated by treatment of the Sertoli cells with testosterone, 5alpha-dihydrotestosterone or synthetic androgens. Even pretreatment with follicle-stimulating hormone to raise AR levels (from 31 up to 82fmol/mg protein) did not result in androgen responsiveness. In contrast, treatment with dexamethasone markedly stimulated TAT-GRE4x-Luc and MMTV-Luc activity. GR levels reached a value of 172fmol/mg protein in the cultured cells and both AR and GR displayed homogeneous distribution by immunocytochemical evaluation. Androgen responsiveness was restored and glucocorticoid responsiveness was increased by cotransfection with AR or GR expression constructs. Under cotransfection conditions, 1nM of testosterone (a concentration that is some 100 times lower than that estimated to be present in the testis) was sufficient to stimulate the TAT-GRE4x-Luc maximally. Our data indicate that cultured Sertoli cells respond better to glucocorticoids than to androgens and that one of the factors limiting androgen responsiveness is the availability of AR. Other factors limiting the transactivation capacity of the (endogenous) AR, however, cannot be excluded.


Assuntos
Genes Reporter/efeitos dos fármacos , Receptores Androgênicos/metabolismo , Receptores de Glucocorticoides/metabolismo , Células de Sertoli/metabolismo , Transfecção/métodos , Animais , Dexametasona/metabolismo , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores Androgênicos/genética , Receptores de Glucocorticoides/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células de Sertoli/efeitos dos fármacos , Testosterona/metabolismo , Testosterona/farmacologia
15.
Cancer Res ; 65(6): 2441-8, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15781660

RESUMO

Aggressive cancer cells typically show a high rate of energy-consuming anabolic processes driving the synthesis of lipids, proteins, and DNA. Here, we took advantage of the ability of the cell-permeable nucleoside 5-aminoimidazole-4-carboxamide (AICA) riboside to increase the intracellular levels of AICA ribotide, an AMP analogue, mimicking a low energy status of the cell. Treatment of cancer cells with AICA riboside impeded lipogenesis, decreased protein translation, and blocked DNA synthesis. Cells treated with AICA riboside stopped proliferating and lost their invasive properties and their ability to form colonies. When administered in vivo, AICA riboside attenuated the growth of MDA-MB-231 tumors in nude mice. These findings point toward a central tie between energy, anabolism, and cancer and suggest that the cellular energy sensing machinery in cancer cells is an exploitable target for cancer prevention and/or therapy.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Ribonucleosídeos/farmacologia , Monofosfato de Adenosina/metabolismo , Adenilato Quinase/metabolismo , Aminoimidazol Carboxamida/metabolismo , Animais , Materiais Biomiméticos/metabolismo , Materiais Biomiméticos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Metabolismo Energético , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ribonucleosídeos/metabolismo , Ribonucleotídeos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Prostate ; 53(1): 34-49, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12210478

RESUMO

BACKGROUND: Primary cultures and subcultures of prostate epithelial cells (PEC) proliferate markedly, but rapidly loose secretory differentiated function and androgen responsiveness. Here, we investigated whether differentiation could be restored or preserved by using three-dimensional reaggregation cultures treated with retinoids and/or androgens. METHODS: PEC were cultured as monolayers or as reaggregation cultures on a rotatory shaker. Reaggregation cultures were also developed from freshly isolated cells. Morphology was evaluated microscopically. Expression of cytokeratins (CKbasal for basal cells and CK18 for luminal cells), E-cadherin, alpha- and beta-catenin, androgen receptor (AR), and prostate specific antigen (PSA) was evaluated by immunohistochemistry and/or Western blotting. Differentiated function was further evaluated by measurements of PSA in the medium and by reverse transcriptase-polymerase chain reactions for AR, PSA, prostate specific membrane antigen, beta-microseminoprotein, and zinc-alpha 2-glycoprotein. Proliferation was evaluated by immunohistochemical staining for Ki-67. RESULTS: Monolayer cultures of PEC expressed CKbasal as well as CK18, a combination compatible with an intermediary amplifying population of epithelial cells. No expression of PSA could be detected, and all attempts to re-induce differentiation of PEC in classic two-dimensional culture systems failed. In reaggregation cultures of subcultured PEC, retinoids proved essential to maintain a compact three-dimensional structure. This effect was accompanied by increased levels of E-cadherin and of the catenins and by a shift in the cytokeratin expression pattern toward that typical for secretory differentiated cells (CK18 only). Even in the presence of androgens, however, PSA remained undetectable. Similar effects of retinoids were observed in reaggregation cultures of freshly prepared PEC, and in the latter cultures, the combination of androgens and retinoids maintained a low level of PSA secretion for at least 40 days. CONCLUSIONS: A combination of retinoids and androgens is able to preserve, for a prolonged period of time, some degree of secretory differentiation in freshly isolated PEC maintained in reaggregation culture. The same combination is unable to restore secretory differentiation in subcultured PEC.


Assuntos
Androgênios/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/fisiologia , Hiperplasia Prostática/patologia , Neoplasias da Próstata/patologia , Retinoides/farmacologia , Western Blotting , Caderinas/análise , Caderinas/biossíntese , Técnicas de Cultura de Células , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/biossíntese , Humanos , Imuno-Histoquímica , Queratinas/análise , Queratinas/biossíntese , Masculino , Antígeno Prostático Específico/análise , Antígeno Prostático Específico/biossíntese , Receptores Androgênicos/fisiologia , Transativadores/análise , Transativadores/biossíntese , alfa Catenina , beta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA