Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 273
Filtrar
1.
J Pharmacol Exp Ther ; 365(1): 84-93, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29339455

RESUMEN

N-acetyltransferase 1 (NAT1) is an enzyme that metabolizes carcinogens, which suggests a potential role in breast carcinogenesis. High NAT1 expression in breast tumors is associated with estrogen receptor α (ERα+) and the luminal subtype. We report that NAT1 mRNA transcript, protein, and enzyme activity were higher in human breast tumors with high expression of ERα/ESR1 compared with normal breast tissue. There was a strong correlation between NATb promoter and NAT1 protein expression/enzyme activity. High NAT1 expression in tumors was not the result of adipocytes, as evidenced by low perilipin (PLIN) expression. ESR1, NAT1, and XBP1 expression were associated in tumor biopsies. Direct regulation of NAT1 transcription by estradiol (E2) was investigated in ERα (+) MCF-7 and T47D breast cancer cells. E2 did not increase NAT1 transcript expression but increased progesterone receptor expression in a dose-dependent manner. Likewise, NAT1 transcript levels were not increased by dihydrotestosterone (DHT) or 5α-androstane-3ß, (3ß-adiol) 17ß-diol. Dithiothreitol increased levels of the activated, spliced XBP1 in ERα (+) MCF-7 and T47D breast cancer cells but did not affect NAT1 or ESR1 expression. We conclude that NAT1 expression is not directly regulated by E2, DHT, 3ß-adiol, or dithiothreitol despite high NAT1 and ESR1 expression in luminal A breast cancer cells, suggesting that ESR1, XBP1, and NAT1 expression may share a common transcriptional network arising from the luminal epithelium associated with better survival in breast cancer. Clusters of high-expression genes, including NAT1, in breast tumors might serve as potential targets for novel therapeutic drug development.


Asunto(s)
Arilamina N-Acetiltransferasa/genética , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Isoenzimas/genética , Androstano-3,17-diol/farmacología , Neoplasias de la Mama/genética , Dihidrotestosterona/farmacología , Estradiol/farmacología , Humanos , Células MCF-7 , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
2.
J Steroid Biochem Mol Biol ; 178: 89-98, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29155210

RESUMEN

Androgen ablation is the standard of care prescribed to patients with advanced or metastatic prostate cancer (PCa) to slow down disease progression. Unfortunately, a majority of PCa patients under androgen ablation progress to castration-resistant prostate cancer (CRPC). Several mechanisms including alternative intra-prostatic androgen production and androgen-independent androgen receptor (AR) activation have been proposed for CRPC progression. Aldo-keto reductase family 1 member C3 (AKR1C3), a multi-functional steroid metabolizing enzyme, is specifically expressed in the cytoplasm of PCa cells; and positive immunoreactivity of the type A γ-aminobutyric acid receptor (GABAAR), an ionotropic receptor and ligand-gated ion channel, is detected on the membrane of PCa cells. We studied a total of 72 radical prostatectomy cases by immunohistochemistry, and identified that 21 cases exhibited positive immunoreactivities for both AKR1C3 and GABAAR. In the dual positive cancer cases, AKR1C3 and GABAAR subunit α1 were either expressed in the same cells or in neighboring cells. Among several possible substrates, AKR1C3 reduces 5α-dihydrotesterone (DHT) to form 5α-androstane-3α, 17ß-diol (3α-diol). 3α-diol is a neurosteroid that acts as a positive allosteric modulator of the GABAAR in the central nervous system (CNS). We examined the hypothesis that 3α-diol-regulated pathological effects in the prostate are GABAAR-dependent, but are independent of the AR. In GABAAR-positive, AR-negative human PCa PC-3 cells, 3α-diol significantly stimulated cell growth in culture and the in ovo chorioallantoic membrane (CAM) xenograft model. 3α-diol also up-regulated expression of the epidermal growth factor (EGF) family of growth factors and activation of EGF receptor (EGFR) and Src as measured by quantitative polymerase chain reaction and immunoblotting, respectively. Inclusion of GABAAR antagonists reversed 3α-diol-stimulated tumor cell growth, expression of EGF family members, and activation of EGFR and Src to the level observed in untreated cells. Results from the present study suggest that 3α-diol may act as an alternative intra-prostatic neurosteroid that activates AR-independent PCa progression. The involvement of AKR1C3-mediated steroid metabolisms in modulating GABAAR activation and promoting PCa progression requires continued studies.


Asunto(s)
Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/metabolismo , Anabolizantes/farmacología , Androstano-3,17-diol/farmacología , Neoplasias de la Próstata/patología , Receptores de GABA-A/metabolismo , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/genética , Proliferación Celular , Progresión de la Enfermedad , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores de GABA-A/genética , Transducción de Señal , Células Tumorales Cultivadas
3.
BMC Cancer ; 17(1): 319, 2017 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-28482871

RESUMEN

BACKGROUND: Estrogen receptor (ER) ß has been suggested to affect ovarian carcinogenesis. We examined the effects of four ERß agonists on proliferation and gene expression of two ovarian cancer cell lines. METHODS: OVCAR-3 and OAW-42 ovarian cancer cells were treated with the ERß agonists ERB-041, WAY200070, Liquiritigenin and 3ß-Adiol and cell growth was measured by means of the Cell Titer Blue Assay (Promega). ERß expression was knocked down by transfection with specific siRNA. Additionally, transcriptome analyses were performed by means of Affymetrix GeneChip arrays. To confirm the results of DNA microarray analysis, Western blot experiments were performed. RESULTS: All ERß agonists tested significantly decreased proliferation of OVCAR-3 and OAW-42 cells at a concentration of 10 nM. Maximum antiproliferative effects were induced by flavonoid Liquiritigenin, which inhibited growth of OVCAR-3 cells by 31.2% after 5 days of treatment, and ERB-041 suppressing proliferation of the same cell line by 29.1%. In OAW-42 cells, maximum effects were observed after treatment with the ERß agonist WAY200070, inhibiting cell growth by 26.8%, whereas ERB-041 decreased proliferation by 24.4%. In turn, knockdown of ERß with specific siRNA increased cell growth of OAW-42 cells about 1.9-fold. Transcriptome analyses revealed a set of genes regulated by ERß agonists including ND6, LCN1 and PTCH2, providing possible molecular mechanisms underlying the observed antiproliferative effects. CONCLUSION: In conclusion, the observed growth-inhibitory effects of all ERß agonists on ovarian cancer cell lines in vitro encourage further studies to test their possible use in the clinical setting.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Receptor beta de Estrógeno/agonistas , Estrógenos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Androstano-3,17-diol/farmacología , Androstano-3,17-diol/uso terapéutico , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Estrógenos/uso terapéutico , Femenino , Flavanonas/farmacología , Flavanonas/uso terapéutico , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/fisiopatología , Oxazoles/farmacología , Oxazoles/uso terapéutico , Fenoles/farmacología , Fenoles/uso terapéutico
4.
Prostate ; 77(7): 803-811, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28181685

RESUMEN

BACKGROUND: There is increasing evidence showing that chronic non-bacterial prostatic inflammation is involved in the pathogenesis of benign prostatic hyperplasia (BPH) and male lower urinary tract symptoms (LUTS). It has also been reported that estrogen receptor ß (ERß) could have an immunoprotective role in prostatic tissue. Therefore, we investigated the effect of ERß-activation on not only prostatic inflammation, but also bladder overactive conditions in a rat model with nonbacterial prostatic inflammation. METHODS: Male Sprague-Dawley rats (8 weeks, n = 15) were divided into three groups: sham-saline group (n = 5), formalin-vehicle group (n = 5), and formalin-treatment group (n = 5). The sham-saline group had sham operation and 50 µl normal saline injected into each ventral lobe of the prostate. The formalin-vehicle group had 50 µl 5% formalin injection into bilateral ventral lobes of the prostate. The formalin-treatment group was treated with 3α-Adiol (a selective ERß agonist precursor) at a dose of 3 mg/kg daily from 2 days before induction of prostatic inflammation, whereas formalin-vehicle rats received vehicle (olive oil). In each group, conscious cystometry was performed on day 28 after intraprostatic formalin injection or sham treatment. After cystometry, the bladder and prostate were harvested for evaluation of mRNA expression and histological analysis. RESULTS: In cystometric investigation, the mean number of non-voiding contractions was significantly greater and voiding intervals were significantly shorter in formalin-vehicle rats than those in sham-saline rats (P < 0.05). In RT-qPCR analysis, mRNA expression of NGF, P2X2, and TRPA1 receptors was significantly increased in the bladder mucosa, and mRNA expression of TNF-α, iNOS and COX2 in the ventral lobes of prostate was significantly increased in formalin-vehicle rats compared with sham-saline rats (P < 0.05). In addition, relative mRNA expression ratio of ERß to ERα (ERß/ERα) in the ventral lobes of prostate was significantly decreased in formalin-vehicle rats compared with sham-saline rats (P < 0.05). These changes were ameliorated by 3α-Adiol administration in formalin-treatment rats. CONCLUSIONS: These results indicate that ERß activation by 3α-Adiol administration, which normalized the ERß/ERα expression ratio in the prostate, can improve not only prostatic inflammation, but also bladder overactivity. Therefore, ERß agonists might be useful for treating irritative bladder symptoms in patients with symptomatic BPH associated with prostatic inflammation. Prostate 77:803-811, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Androstano-3,17-diol/farmacología , Receptor beta de Estrógeno , Síntomas del Sistema Urinario Inferior , Hiperplasia Prostática , Prostatitis , Vejiga Urinaria/metabolismo , Animales , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/antagonistas & inhibidores , Estrógenos/farmacología , Síntomas del Sistema Urinario Inferior/diagnóstico , Síntomas del Sistema Urinario Inferior/inmunología , Síntomas del Sistema Urinario Inferior/fisiopatología , Masculino , Próstata/inmunología , Próstata/patología , Hiperplasia Prostática/inmunología , Hiperplasia Prostática/patología , Hiperplasia Prostática/fisiopatología , Prostatitis/diagnóstico , Prostatitis/inmunología , Prostatitis/fisiopatología , Factores Protectores , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/metabolismo , Vejiga Urinaria/patología , Urodinámica
5.
Endocrinology ; 157(12): 4570-4578, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27754784

RESUMEN

Low free T levels in men are associated with age-related cognitive decline and increased risk for neurotoxicity, resulting in disease. The mechanisms underlying these observations remain poorly defined. Although rapid, androgen receptor-dependent activation of ERK has been postulated as a neurotrophic and neuroprotective mechanism, actions of T metabolites such as 5α-androstane-3α,17ß-diol (3α-diol) may also be involved. We investigated the influence of 3α-diol on the induction of ERK phosphorylation in SH-SY5Y human female neuroblastoma cells and primary cortical neurons from male and female mice. In SH-SY5Y cells, ERK phosphorylation was induced by 10 nM DHT, epidermal growth factor, hydrogen peroxide (H2O2), and acetylcholine. The addition of 10 nM 3α-diol, which did not itself activate ERK, significantly inhibited ERK phosphorylation induced by DHT, epidermal growth factor, or H2O2, but not acetylcholine. In both SH-SY5Y cells and primary cortical neurons, prolonged ERK phosphorylation and caspase-3 cleavage resulting from amyloid ß-peptide 1-42 (Aß42) exposure were inhibited by cotreatment with 3α-diol. 3α-diol also reduced the loss in cellular viability induced by Aß42 or H2O2 in SH-SY5Y cells. These data suggest that T-mediated neuroprotection may occur via two distinct but complementary mechanisms: an initial rapid activation of ERK phosphorylation, followed by modulation via 3α-diol of the potentially adverse consequences of prolonged ERK activation.


Asunto(s)
Androstano-3,17-diol/farmacología , Corteza Cerebral/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuronas/efectos de los fármacos , Acetilcolina/farmacología , Péptidos beta-Amiloides/farmacología , Animales , Caspasa 3/metabolismo , Línea Celular Tumoral , Corteza Cerebral/metabolismo , Dihidrotestosterona/farmacología , Factor de Crecimiento Epidérmico/farmacología , Femenino , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Neuroblastoma/metabolismo , Neuronas/metabolismo , Fosforilación/efectos de los fármacos
6.
J Neurosci ; 35(2): 666-77, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25589761

RESUMEN

Maternal social stress during late pregnancy programs hypothalamo-pituitary-adrenal (HPA) axis hyper-responsiveness to stressors, such that adult prenatally stressed (PNS) offspring display exaggerated HPA axis responses to a physical stressor (systemic interleukin-1ß; IL-1ß) in adulthood, compared with controls. IL-1ß acts via a noradrenergic relay from the nucleus tractus solitarii (NTS) to corticotropin releasing hormone neurons in the paraventricular nucleus (PVN). Neurosteroids can reduce HPA axis responses, so allopregnanolone and 3ß-androstanediol (3ß-diol; 5α-reduced metabolites of progesterone and testosterone, respectively) were given subacutely (over 24 h) to PNS rats to seek reversal of the "programmed" hyper-responsive HPA phenotype. Allopregnanolone attenuated ACTH responses to IL-1ß (500 ng/kg, i.v.) in PNS females, but not in PNS males. However, 3ß-diol normalized HPA axis responses to IL-1ß in PNS males. Impaired testosterone and progesterone metabolism or increased secretion in PNS rats was indicated by greater plasma testosterone and progesterone concentrations in male and female PNS rats, respectively. Deficits in central neurosteroid production were indicated by reduced 5α-reductase mRNA levels in both male and female PNS offspring in the NTS, and in the PVN in males. In PNS females, adenovirus-mediated gene transfer was used to upregulate expression of 5α-reductase and 3α-hydroxysteroid dehydrogenase mRNAs in the NTS, and this normalized hyperactive HPA axis responses to IL-1ß. Thus, downregulation of neurosteroid production in the brain may underlie HPA axis hyper-responsiveness in prenatally programmed offspring, and administration of 5α-reduced steroids acutely to PNS rats overrides programming of hyperactive HPA axis responses to immune challenge in a sex-dependent manner.


Asunto(s)
Androstano-3,17-diol/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Pregnanolona/farmacología , Estrés Psicológico/tratamiento farmacológico , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/genética , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/metabolismo , Androstano-3,17-diol/uso terapéutico , Animales , Femenino , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Interleucina-1beta/farmacología , Masculino , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/crecimiento & desarrollo , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/crecimiento & desarrollo , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Pregnanolona/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/crecimiento & desarrollo , Núcleo Solitario/metabolismo
7.
Biochim Biophys Acta ; 1842(12 Pt A): 2427-38, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25281013

RESUMEN

The brain has high energy requirements to maintain neuronal activity. Consequently impaired mitochondrial function will lead to disease. Normal aging is associated with several alterations in neurosteroid production and secretion. Decreases in neurosteroid levels might contribute to brain aging and loss of important nervous functions, such as memory. Up to now, extensive studies only focused on estradiol as a promising neurosteroid compound that is able to ameliorate cellular bioenergetics, while the effects of other steroids on brain mitochondria are poorly understood or not investigated at all. Thus, we aimed to characterize the bioenergetic modulating profile of a panel of seven structurally diverse neurosteroids (progesterone, estradiol, estrone, testosterone, 3α-androstanediol, DHEA and allopregnanolone), known to be involved in brain function regulation. Of note, most of the steroids tested were able to improve bioenergetic activity in neuronal cells by increasing ATP levels, mitochondrial membrane potential and basal mitochondrial respiration. In parallel, they modulated redox homeostasis by increasing antioxidant activity, probably as a compensatory mechanism to a slight enhancement of ROS which might result from the rise in oxygen consumption. Thereby, neurosteroids appeared to act via their corresponding receptors and exhibited specific bioenergetic profiles. Taken together, our results indicate that the ability to boost mitochondria is not unique to estradiol, but seems to be a rather common mechanism of different steroids in the brain. Thus, neurosteroids may act upon neuronal bioenergetics in a delicate balance and an age-related steroid disturbance might be involved in mitochondrial dysfunction underlying neurodegenerative disorders.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Adenosina Trifosfato/metabolismo , Envejecimiento/metabolismo , Androstano-3,17-diol/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Deshidroepiandrosterona/farmacología , Relación Dosis-Respuesta a Droga , Estradiol/farmacología , Estrona/farmacología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Mitocondrias/fisiología , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Neurotransmisores/metabolismo , Consumo de Oxígeno , Pregnanolona/farmacología , Progesterona/farmacología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Testosterona/farmacología
8.
Biomed Res Int ; 2014: 801473, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24877132

RESUMEN

Prostate cancer (PC) progression from androgen-dependent (AD) to castration-resistant (CR) disease is a process caused by modifications of different signal transduction pathways within tumor microenvironment. Reducing cell proliferation, estrogen receptor beta (ERbeta) is emerging as a potential target in PC chemoprevention. Among the known selective ERbeta ligands, 3beta-Adiol, the endogenous ligand in the prostate, has been proved to counteract PC progression. This study compares the effects of chronic exposure (1-12 weeks) to different ERbeta selective ligands (DPN, 8beta-VE2, 3beta-Adiol) on proliferation of human androgen-responsive CWR22Rv1 cells, representing an intermediate phenotype between the AD- and CR-PC. 3beta-Adiol (10 nM) is the sole ligand decreasing cell proliferation and increasing p21 levels. In vitro transcriptional activity assays were performed to elucidate different behavior between 3beta-Adiol and the other ligands; in these experiments the endogenous and the main ERbeta subtype activation were considered. It is concluded that ERbeta activation has positive effects also in androgen-responsive PC. The underlying mechanisms are still to be clarified and may include the interplay among different ERbeta subtypes and the specific PC microenvironment. ERbeta agonists might be useful in counteracting PC progression, although the final outcome may depend upon the molecular pattern specific to each PC lesion.


Asunto(s)
Androstano-3,17-diol/análogos & derivados , Androstano-3,17-diol/farmacología , Receptor beta de Estrógeno/agonistas , Neoplasias de la Próstata/tratamiento farmacológico , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Receptor beta de Estrógeno/metabolismo , Células HEK293 , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
9.
J Steroid Biochem Mol Biol ; 143: 115-21, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24607810

RESUMEN

Diabetic peripheral neuropathy causes a decrease in the levels of dihydroprogesterone and 5α-androstane-3α,17ß-diol (3α-diol) in the peripheral nerves. These two neuroactive steroids exert protective effects, by mechanisms that still remain elusive. We have previously shown that the activation of Liver X Receptors improves the peripheral neuropathic phenotype in diabetic rats. This protective effect is accompanied by the restoration to control values of the levels of dihydroprogesterone and 3α-diol in peripheral nerves. In addition, activation of these receptors decreases peripheral myelin abnormalities by improving the lipid desaturation capacity, which is strongly blunted by diabetes, and ultimately restores the myelin lipid profile to non-diabetic values. On this basis, we here investigate whether dihydroprogesterone or 3α-diol may exert their protective effects by modulating the myelin lipid profile. We report that both neuroactive steroids act on the lipogenic gene expression profile in the sciatic nerve of diabetic rats, reducing the accumulation of myelin saturated fatty acids and promoting desaturation. These changes were associated with a reduction in myelin structural alterations. These findings provide evidence that dihydroprogesterone and 3α-diol are protective agents against diabetic peripheral neuropathy by regulating the de novo lipogenesis pathway, which positively influences myelin lipid profile.


Asunto(s)
20-alfa-Dihidroprogesterona/farmacología , Androstano-3,17-diol/farmacología , Neuropatías Diabéticas/metabolismo , Lípidos/análisis , Vaina de Mielina/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Nervio Ciático/metabolismo , Anabolizantes/farmacología , Animales , Biomarcadores/análisis , Cromatografía Liquida , Diabetes Mellitus Experimental/complicaciones , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/etiología , Masculino , Vaina de Mielina/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/etiología , Progestinas/farmacología , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nervio Ciático/efectos de los fármacos , Espectrometría de Masas en Tándem
10.
PLoS One ; 8(11): e80915, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260511

RESUMEN

Painful peripheral neuropathy belongs to major side-effects limiting cancer chemotherapy. Paclitaxel, widely used to treat several cancers, induces neurological symptoms including burning pain, allodynia, hyperalgesia and numbness. Therefore, identification of drugs that may effectively counteract paclitaxel-induced neuropathic symptoms is crucial. Here, we combined histopathological, neurochemical, behavioral and electrophysiological methods to investigate the natural neurosteroid 3α-androstanediol (3α-DIOL) ability to counteract paclitaxel-evoked peripheral nerve tissue damages and neurological symptoms. Prophylactic or corrective 3α-DIOL treatment (4 mg/kg/2 days) prevented or suppressed PAC-evoked heat-thermal hyperalgesia, cold-allodynia and mechanical allodynia/hyperalgesia, by reversing to normal, decreased thermal and mechanical pain thresholds of PAC-treated rats. Electrophysiological studies demonstrated that 3α-DIOL restored control values of nerve conduction velocity and action potential peak amplitude significantly altered by PAC-treatment. 3α-DIOL also repaired PAC-induced nerve damages by restoring normal neurofilament-200 level in peripheral axons and control amount of 2',3'-cyclic-nucleotide-3'-phosphodiesterase in myelin sheaths. Decreased density of intraepidermal nerve fibers evoked by PAC-therapy was also counteracted by 3α-DIOL treatment. More importantly, 3α-DIOL beneficial effects were not sedation-dependent but resulted from its neuroprotective ability, nerve tissue repairing capacity and long-term analgesic action. Altogether, our results showing that 3α-DIOL efficiently counteracted PAC-evoked painful symptoms, also offer interesting possibilities to develop neurosteroid-based strategies against chemotherapy-induced peripheral neuropathy. This article shows that the prophylactic or corrective treatment with 3α-androstanediol prevents or suppresses PAC-evoked painful symptoms and peripheral nerve dysfunctions in rats. The data suggest that 3α-androstanediol-based therapy may constitute an efficient strategy to explore in humans for the eradication of chemotherapy-induced peripheral neuropathy.


Asunto(s)
Androstano-3,17-diol/farmacología , Antineoplásicos Fitogénicos/efectos adversos , Hiperalgesia/prevención & control , Neuralgia/prevención & control , Fármacos Neuroprotectores/farmacología , Paclitaxel/efectos adversos , Dolor/prevención & control , Enfermedades del Sistema Nervioso Periférico/prevención & control , Potenciales de Acción/efectos de los fármacos , Androstano-3,17-diol/análogos & derivados , Animales , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Masculino , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/patología , Conducción Nerviosa/efectos de los fármacos , Neuralgia/inducido químicamente , Neuralgia/fisiopatología , Dolor/inducido químicamente , Dolor/fisiopatología , Dimensión del Dolor , Nervios Periféricos/efectos de los fármacos , Nervios Periféricos/patología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Ratas , Ratas Sprague-Dawley
11.
Breast Cancer Res Treat ; 139(3): 731-40, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23780684

RESUMEN

Aromatase inhibitors (AIs) have been reported to exert their antiproliferative effects in postmenopausal women with hormone receptor-positive breast cancer not only by reducing estrogen production but also by unmasking the inhibitory effects of androgens such as testosterone (TS) and dihydrotestosterone (DHT). However, the role of androgens in AI-resistance mechanisms is not sufficiently understood. 5α-Androstane-3ß,17ß-diol (3ß-diol) generated from DHT by 3ß-hydroxysteroid dehydrogenase type 1 (HSD3B1) shows androgenic and substantial estrogenic activities, representing a potential mechanism of AI resistance. Estrogen response element (ERE)-green fluorescent protein (GFP)-transfected MCF-7 breast cancer cells (E10 cells) were cultured for 3 months under steroid-depleted, TS-supplemented conditions. Among the surviving cells, two stable variants showing androgen metabolite-dependent ER activity were selected by monitoring GFP expression. We investigated the process of adaptation to androgen-abundant conditions and the role of androgens in AI-resistance mechanisms in these variant cell lines. The variant cell lines showed increased growth and induction of estrogen-responsive genes rather than androgen-responsive genes after stimulation with androgens or 3ß-diol. Further analysis suggested that increased expression of HSD3B1 and reduced expression of androgen receptor (AR) promoted adaptation to androgen-abundant conditions, as indicated by the increased conversion of DHT into 3ß-diol by HSD3B1 and AR signal reduction. Furthermore, in parental E10 cells, ectopic expression of HSD3B1 or inhibition of AR resulted in adaptation to androgen-abundant conditions. Coculture with stromal cells to mimic local estrogen production from androgens reduced cell sensitivity to AIs compared with parental E10 cells. These results suggest that increased expression of HSD3B1 and reduced expression of AR might reduce the sensitivity to AIs as demonstrated by enhanced androgen metabolite-induced ER activation and growth mechanisms. Androgen metabolite-dependent growth of breast cancer cells may therefore play a role in AI-resistance.


Asunto(s)
Andrógenos/metabolismo , Inhibidores de la Aromatasa/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Receptores de Estrógenos/metabolismo , Androstano-3,17-diol/metabolismo , Androstano-3,17-diol/farmacología , Neoplasias de la Mama/metabolismo , Dihidrotestosterona/metabolismo , Dihidrotestosterona/farmacología , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Letrozol , Células MCF-7/efectos de los fármacos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Nitrilos/farmacología , Progesterona Reductasa/genética , Progesterona Reductasa/metabolismo , Receptores Androgénicos/metabolismo , Elementos de Respuesta/genética , Transducción de Señal , Esteroide Isomerasas/genética , Esteroide Isomerasas/metabolismo , Testosterona/metabolismo , Testosterona/farmacología , Triazoles/farmacología
12.
Endocrinology ; 154(5): 1802-12, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23515287

RESUMEN

Testosterone has been shown to suppress the acute stress-induced activation of the hypothalamic-pituitary-adrenal axis; however, the mechanisms underlying this response remain unclear. The hypothalamic-pituitary-adrenal axis is regulated by a neuroendocrine subpopulation of medial parvocellular neurons in the paraventricular nucleus of the hypothalamus (PVN). These neurons are devoid of androgen receptors (ARs). Therefore, a possibility is that the PVN target neurons respond to a metabolite in the testosterone catabolic pathway via an AR-independent mechanism. The dihydrotestosterone metabolite, 5α-androstane-3ß,17ß-diol (3ß-diol), binds and activates estrogen receptor-ß (ER-ß), the predominant ER in the PVN. In the PVN, ER-ß is coexpressed with oxytocin (OT). Therefore, we tested the hypothesis that 3ß-diol regulates OT expression through ER-ß activation. Treatment of ovariectomized rats with estradiol benzoate or 3ß-diol for 4 days increased OT mRNA selectively in the midcaudal, but not rostral PVN compared with vehicle-treated controls. 3ß-Diol treatment also increased OT mRNA in the hypothalamic N38 cell line in vitro. The functional interactions between 3ß-diol and ER-ß with the human OT promoter were examined using an OT promoter-luciferase reporter construct (OT-luc). In a dose-dependent manner, 3ß-diol treatment increased OT-luc activity when cells were cotransfected with ER-ß, but not ER-α. The 3ß-diol-induced OT-luc activity was reduced by deletion of the promoter region containing the composite hormone response element (cHRE). Point mutations of the cHRE also prevented OT-luc activation by 3ß-diol. These results indicate that 3ß-diol induces OT promoter activity via ER-ß-cHRE interactions.


Asunto(s)
Androstano-3,17-diol/farmacología , Receptor beta de Estrógeno/fisiología , Oxitocina/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Andrógenos/metabolismo , Androstano-3,17-diol/metabolismo , Animales , Células Cultivadas , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Ratones , Ovariectomía , Oxitocina/metabolismo , Regiones Promotoras Genéticas/fisiología , Ratas , Ratas Sprague-Dawley , Elementos de Respuesta/efectos de los fármacos , Elementos de Respuesta/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Activación Transcripcional/genética
13.
Steroids ; 78(2): 195-202, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23153457

RESUMEN

Expression of estrogen receptor ß (ERß) has been described to reduce growth of cancer cell lines derived from hormone-dependent tumors, like breast cancer. In this study we tested to what extent two ERß agonists, androgen derivative 3ß-Adiol and flavonoid Liquiritigenin, would affect growth and gene expression of different ERß-positive human breast cancer cell lines. Under standard cell culture conditions, we observed 3ß-Adiol to inhibit growth of MCF-7 cells in a dose-dependent manner, whereas growth of BT-474 and MCF-10A cells was suppressed by the maximum concentration (100 nM) only. When treated in serum-free medium, all cell lines except of MDA-MB-231 were responsive to 1 nM 3ß-Adiol, and ZR75-1 cells exhibited a dose-dependent antiproliferative response. Providing putative mechanisms underlying the observed growth-inhibitory effect, expression of Ki-67 or cyclins A2 and B1 was downregulated after 3ß-Adiol treatment in all responsive lines. In contrast, treatment with lower doses of Liquiritigenin did not affect growth. In MCF-7 cells, the highest dose of this flavonoid exerted proliferative effects accompanied by increased expression of cyclin B1, PR and PS2, indicating unspecific activation of ERα. In conclusion, the ERß agonists tested exerted distinct concentration-dependent and cell line-specific effects on growth and gene expression. The observed inhibitory effects of 3ß-Adiol on breast cancer cell growth encourage further studies on the potential of this and other ERß agonists as targeted drugs for breast cancer therapy.


Asunto(s)
Androstano-3,17-diol/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Receptor beta de Estrógeno/agonistas , Flavanonas/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina B1/metabolismo , Relación Dosis-Respuesta a Droga , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Factores de Tiempo
14.
Sheng Li Xue Bao ; 64(6): 633-8, 2012 Dec 25.
Artículo en Chino | MEDLINE | ID: mdl-23258325

RESUMEN

The influence of 3α-androstanediol (3α-diol) on twitch and electroencephalogram (EEG) of the epileptic rats induced by pentylenetetrazole (PTZ) has been observed in this experiment in order to comprehensively explore the role of 3α-diol on epileptic attack from the aspects of behavior and EEG. Thirty-two male Sprague-Dawley rats were evenly and randomly divided into 4 groups: the normal and supplied with oil epileptic (N+oil+PTZ) group, the normal and supplied with 3α-diol epileptic (N+3α-diol+PTZ) group, the gonadectomized and supplied with oil epileptic (GDX+oil+PTZ) group and the gonadectomized and supplied with 3α-diol epileptic (GDX+3α-diol+PTZ) group. The changes of the behavior and EEG of epileptic rats in every group were recorded and analyzed. The results of behavior observation showed that the latency to clonic seizure and tonic-clonic seizure was shortened and the number of tonic-clonic seizure was increased significantly in the GDX+oil+PTZ group in comparison with N+oil+PTZ group (P < 0.05); comparing GDX+3α-diol+PTZ group with GDX+oil+PTZ group, or N+3α-diol+PTZ group with N+oil+PTZ group, we found that the latency to clonic seizure and tonic-clonic seizure became prolonged significantly, and the number of clonic seizure and tonic-clonic seizure was decreased significantly (P < 0.05). The results of EEG showed that the latency to epileptic waves was cut and the number of epileptic waves was augmented significantly in the GDX+oil+PTZ group in comparison with N+oil+PTZ group (P < 0.05); comparing GDX+3α-diol+PTZ group with GDX+oil+PTZ group, or N+3α-diol+PTZ group with N+oil+PTZ group, we found that the latency to epileptic waves became lengthened significantly, the number of epileptic waves was reduced significantly and the percentage of change of TP (total power of spectrum) was lessened significantly (P < 0.05). These results indicate that 3α-diol has an antiepileptic activity in the gonadectomized and normal epileptic rats.


Asunto(s)
Androstano-3,17-diol/análogos & derivados , Anticonvulsivantes/farmacología , Epilepsia/tratamiento farmacológico , Pentilenotetrazol/efectos adversos , Convulsiones/tratamiento farmacológico , Androstano-3,17-diol/farmacología , Animales , Electroencefalografía , Epilepsia/inducido químicamente , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
15.
Endocrinology ; 153(12): 5949-60, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23117931

RESUMEN

P-glycoprotein (Pgp), a multiple drug resistance transporter expressed by vascular endothelial cells, is a key component of the blood-brain barrier and has been shown to increase after inflammation. The nonaromatizable androgen, dihydrotestosterone (DHT), decreases inflammatory markers in vascular smooth muscle cells, independent of androgen receptor (AR) stimulation. The principal metabolite of DHT, 5α-androstane-3ß,17ß-diol (3ß-diol), activates estrogen receptor (ER)ß and similarly decreases inflammatory markers in vascular cells. Therefore, we tested the hypothesis that either DHT or 3ß-diol decrease cytokine-induced proinflammatory mediators, vascular cell adhesion molecule-1 (VCAM-1) and cyclooxygenase-2 (COX-2), to regulate Pgp expression in male primary human brain microvascular endothelial cells (HBMECs). Using RT-qPCR, the mRNAs for AR, ERα, and ERß and steroid metabolizing enzymes necessary for DHT conversion to 3ß-diol were detected in male HBMECs demonstrating that the enzymes and receptors for production of and responsiveness to 3ß-diol are present. Western analysis showed that 3ß-diol reduced COX-2 and Pgp expression; the effect on Pgp was inhibited by the ER antagonist, ICI-182,780. IL-1ß-caused an increase in COX-2 and VCAM-1 that was reduced by either DHT or 3ß-diol. 3ß-diol also decreased cytokine-induced Pgp expression. ICI-182,780 blocked the effect of 3ß-diol on COX-2 and VCAM-1, but not Pgp expression. Therefore, in cytokine-stimulated male HBMECs, the effect of 3ß-diol on proinflammatory mediator expression is ER dependent, whereas its effect on Pgp expression is ER independent. These studies suggest a novel role of 3ß-diol in regulating blood-brain barrier function and support the concept that 3ß-diol can be protective against proinflammatory mediator stimulation.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Androstano-3,17-diol/metabolismo , Encéfalo/metabolismo , Ciclooxigenasa 2/metabolismo , Regulación de la Expresión Génica , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Androstano-3,17-diol/farmacología , Barrera Hematoencefálica , Células Cultivadas , Dihidrotestosterona/farmacología , Receptor beta de Estrógeno/metabolismo , Humanos , Inflamación , Masculino , ARN Mensajero/metabolismo
16.
Neuroscience ; 226: 397-410, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22989919

RESUMEN

Anabolic androgenic steroids (AAS), synthetic testosterone derivatives that are used for ergogenic purposes, alter neurotransmission and behaviors mediated by GABA(A) receptors. Some of these effects may reflect direct and rapid action of these synthetic steroids at the receptor. The ability of other natural allosteric steroid modulators to alter GABA(A) receptor-mediated currents is dependent upon the phosphorylation state of the receptor complex. Here we show that phosphorylation of the GABA(A) receptor complex immunoprecipitated by ß(2)/ß(3) subunit-specific antibodies from the medial preoptic area (mPOA) of the mouse varies across the estrous cycle; with levels being significantly lower in estrus. Acute exposure to the AAS, 17α-methyltestosterone (17α-MeT), had no effect on the amplitude or kinetics of inhibitory postsynaptic currents in the mPOA of estrous mice when phosphorylation was low, but increased the amplitude of these currents from mice in diestrus, when it was high. Inclusion of the protein kinase C (PKC) inhibitor, calphostin, in the recording pipette eliminated the ability of 17α-MeT to enhance currents from diestrous animals, suggesting that PKC-receptor phosphorylation is critical for the allosteric modulation elicited by AAS during this phase. In addition, a single injection of 17α-MeT was found to impair an mPOA-mediated behavior (nest building) in diestrus, but not in estrus. PKC is known to target specific serine residues in the ß(3) subunit of the GABA(A) receptor. Although phosphorylation of these ß(3) serine residues showed a similar profile across the cycle, as did phosphoserine in mPOA lysates immunoprecipitated with ß2/ß3 antibody (lower in estrus than in diestrus or proestrus), the differences were not significant. These data suggest that the phosphorylation state of the receptor complex regulates both the ability of AAS to modulate receptor function in the mPOA and the expression of a simple mPOA-dependent behavior through a PKC-dependent mechanism that involves the ß(3) subunit and other sites within the GABA(A) receptor complex.


Asunto(s)
Anabolizantes/farmacología , Andrógenos/farmacología , Ciclo Estral/fisiología , Metiltestosterona/farmacología , Área Preóptica/efectos de los fármacos , Receptores de GABA-A/metabolismo , Androstano-3,17-diol/farmacología , Animales , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Western Blotting , Femenino , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/aislamiento & purificación , Proteínas del Tejido Nervioso/metabolismo , Comportamiento de Nidificación/efectos de los fármacos , Fosforilación , Fosfoserina/metabolismo , Área Preóptica/metabolismo , Proteína Quinasa C/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Maduración Sexual
17.
Endocrinology ; 153(5): 2353-61, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22434086

RESUMEN

The endocrine component of the stress response is regulated by glucocorticoids and sex steroids. Testosterone down-regulates hypothalamic-pituitary-adrenal (HPA) axis activity; however, the mechanisms by which it does so are poorly understood. A candidate testosterone target is the oxytocin gene (Oxt), given that it too inhibits HPA activity. Within the paraventricular nucleus of the hypothalamus, oxytocinergic neurons involved in regulating the stress response do not express androgen receptors but do express estrogen receptor-ß (ERß), which binds the dihydrotestosterone metabolite 3ß,17ß-diol (3ß-diol). Testosterone regulation of the HPA axis thus appears to involve the conversion to the ERß-selective ligand 5α-androstane, 3ß-diol. To study mechanisms by which 3ß-diol could regulate Oxt expression, we used a hypothalamic neuronal cell line derived from embryonic mice that expresses Oxt constitutively and compared 3ß-diol with estradiol (E2) effects. E2 and 3ß-diol elicited a phasic response in Oxt mRNA levels. In the presence of either ligand, Oxt mRNA levels were increased for at least 60 min and returned to baseline by 2 h. ERß occupancy preceded an increase in Oxt mRNA levels in the presence of 3ß-diol but not E2. In tandem with ERß occupancy, 3ß-diol increased occupancy of the Oxt promoter by cAMP response element-binding protein and steroid receptor coactivator-1 at 30 min. At the same time, 3ß-diol led to the increased acetylation of histone H4 but not H3. Taken together, the data suggest that in the presence of 3ß-diol, ERß associates with cAMP response element-binding protein and steroid receptor coactivator-1 to form a functional complex that drives Oxt gene expression.


Asunto(s)
Androstano-3,17-diol/farmacología , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Oxitocina/genética , Androstano-3,17-diol/metabolismo , Animales , Línea Celular , Células Cultivadas , Estradiol/metabolismo , Estradiol/farmacología , Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Oxitocina/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos
18.
Anticancer Drugs ; 23(8): 803-14, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22361842

RESUMEN

This study investigated the antineoplasic potential of a new family of aminosteroids. The antiproliferative activity of seven 5α-androstane-3α,17ß-diol derivatives selected from a screening study was measured on nine cancerous cell lines (HL-60, K-562, LNCaP, PC-3, Shionogi, MCF-7, MDA-MB-231, BT-20, and OVCAR-3) and two normal cell lines (peripheral blood lymphocytes and WI-38). The aminosteroids efficiently inhibited the cell growth of seven cancer cell lines [inhibitory concentration (IC(50)) values=0.2-6.4 µmol/l] and showed weak toxicity on normal cell lines. Two representative aminosteroids were tested and found to induce apoptosis and a G0/G1 cell cycle block in HL-60-treated cells, but not terminal myeloid differentiation. By a nuclear morphology analysis with fluorescence microscopy, typical apoptotic morphological changes were exhibited by treated cells. One aminosteroid tested in vivo (xenograft model) reduced the breast cancer (MCF-7 cells) tumor growth induced in nude mice. Furthermore, the information gathered suggests that this family of aminosteroids induced growth inhibition cells by arresting the cell cycle and triggering apoptosis.


Asunto(s)
Androstano-3,17-diol/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Androstano-3,17-diol/química , Animales , Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Femenino , Fase G1/efectos de los fármacos , Células HL-60 , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Fluorescente , Neoplasias/patología , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Invest New Drugs ; 30(1): 176-85, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20890632

RESUMEN

RM, a novel aminosteroid synthesized by our research group, shows a broad spectrum of antitumor activity against nine cancer cell lines and limited toxicity against two normal cell lines. However, its related mechanism of action has not yet been elucidated. In this study, we investigated the cellular and molecular events underlying the cytotoxicity of RM in human acute promyelocytic leukemia HL-60 cells. RM was found to induce a G0/G1 cell cycle block of HL-60 cells but not terminal myeloid differentiation. Interestingly, typical apoptotic morphological changes were exhibited by HL-60 cells treated with RM stained with Hoechst 33342 and examined by fluorescence microscopy. Apoptotic death assay using annexin-V/propidium iodide dual staining flow cytometry demonstrated a dose-dependent apoptotic effect of RM on HL-60 cells. In addition, RM induced the cleavage of caspase-3, caspase-8 and PARP, but not the cleavage of caspase-9. Our findings suggest that RM reduces HL-60 cells survival through a caspase-dependent death receptor pathway.


Asunto(s)
Androstano-3,17-diol/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Leucemia Promielocítica Aguda/patología , Caspasas/metabolismo , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Células HL-60 , Humanos , Microscopía Fluorescente , Poli(ADP-Ribosa) Polimerasas/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Factores de Tiempo
20.
Proc Natl Acad Sci U S A ; 108(50): 20090-4, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22114194

RESUMEN

Estrogen receptor ß (ERß) is activated in the prostate by 5α-androstane-3ß,17ß-diol (3ß-Adiol) where it exerts antiproliferative activity. The proliferative action of the androgen receptor is activated by 5α-dihydrotestosterone (DHT). Thus, prostate growth is governed by the balance between androgen receptor and ERß activation. 3ß-Adiol is a high-affinity ligand and agonist of ERß and is derived from DHT by 3-keto reductase/3ß-hydroxysteroid dehydrogenase enzymes. Here, we demonstrate that, when it is expressed in living cells containing an estrogen response element-luciferase reporter, 17ß-hydroxysteroid dehydrogenase type 6 (17ßHSD6) converts the androgen DHT to the estrogen 3ß-Adiol, and this leads to activation of the ERß reporter. This conversion of DHT occurs at concentrations that are in the physiological range of this hormone in the prostate. Immunohistochemical analysis revealed that 17ßHSD6 is expressed in ERß-positive epithelial cells of the human prostate and that, in prostate cancers of Gleason grade higher than 3, both ERß and 17ßHSD6 are undetectable. Both proteins were present in benign prostatic hyperplasia samples. These observations reveal that formation of 3ß-Adiol via 17ßHSD6 from DHT is an important growth regulatory pathway that is lost in prostate cancer.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Racemasas y Epimerasas/metabolismo , Transducción de Señal , Androstano-3,17-diol/farmacología , Proliferación Celular/efectos de los fármacos , Dihidrotestosterona/farmacología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Receptor beta de Estrógeno/genética , Células HEK293 , Humanos , Hiperplasia , Inmunohistoquímica , Masculino , Modelos Biológicos , Próstata/efectos de los fármacos , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA