Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Lancet Child Adolesc Health ; 8(5): 325-338, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38513681

RESUMEN

BACKGROUND: Sepsis is defined as dysregulated host response to infection that leads to life-threatening organ dysfunction. Biomarkers characterising the dysregulated host response in sepsis are lacking. We aimed to develop host gene expression signatures to predict organ dysfunction in children with bacterial or viral infection. METHODS: This cohort study was done in emergency departments and intensive care units of four hospitals in Queensland, Australia, and recruited children aged 1 month to 17 years who, upon admission, underwent a diagnostic test, including blood cultures, for suspected sepsis. Whole-blood RNA sequencing of blood was performed with Illumina NovaSeq (San Diego, CA, USA). Samples with completed phenotyping, monitoring, and RNA extraction by March 31, 2020, were included in the discovery cohort; samples collected or completed thereafter and by Oct 27, 2021, constituted the Rapid Paediatric Infection Diagnosis in Sepsis (RAPIDS) internal validation cohort. An external validation cohort was assembled from RNA sequencing gene expression count data from the observational European Childhood Life-threatening Infectious Disease Study (EUCLIDS), which recruited children with severe infection in nine European countries between 2012 and 2016. Feature selection approaches were applied to derive novel gene signatures for disease class (bacterial vs viral infection) and disease severity (presence vs absence of organ dysfunction 24 h post-sampling). The primary endpoint was the presence of organ dysfunction 24 h after blood sampling in the presence of confirmed bacterial versus viral infection. Gene signature performance is reported as area under the receiver operating characteristic curves (AUCs) and 95% CI. FINDINGS: Between Sept 25, 2017, and Oct 27, 2021, 907 patients were enrolled. Blood samples from 595 patients were included in the discovery cohort, and samples from 312 children were included in the RAPIDS validation cohort. We derived a ten-gene disease class signature that achieved an AUC of 94·1% (95% CI 90·6-97·7) in distinguishing bacterial from viral infections in the RAPIDS validation cohort. A ten-gene disease severity signature achieved an AUC of 82·2% (95% CI 76·3-88·1) in predicting organ dysfunction within 24 h of sampling in the RAPIDS validation cohort. Used in tandem, the disease class and disease severity signatures predicted organ dysfunction within 24 h of sampling with an AUC of 90·5% (95% CI 83·3-97·6) for patients with predicted bacterial infection and 94·7% (87·8-100·0) for patients with predicted viral infection. In the external EUCLIDS validation dataset (n=362), the disease class and disease severity predicted organ dysfunction at time of sampling with an AUC of 70·1% (95% CI 44·1-96·2) for patients with predicted bacterial infection and 69·6% (53·1-86·0) for patients with predicted viral infection. INTERPRETATION: In children evaluated for sepsis, novel host transcriptomic signatures specific for bacterial and viral infection can identify dysregulated host response leading to organ dysfunction. FUNDING: Australian Government Medical Research Future Fund Genomic Health Futures Mission, Children's Hospital Foundation Queensland, Brisbane Diamantina Health Partners, Emergency Medicine Foundation, Gold Coast Hospital Foundation, Far North Queensland Foundation, Townsville Hospital and Health Services SERTA Grant, and Australian Infectious Diseases Research Centre.


Asunto(s)
Infecciones Bacterianas , Sepsis , Virosis , Humanos , Niño , Estudios de Cohortes , Transcriptoma , Insuficiencia Multiorgánica/diagnóstico , Insuficiencia Multiorgánica/genética , Estudios Prospectivos , Australia , Sepsis/diagnóstico , Sepsis/genética
2.
Curr Oncol ; 30(11): 9437-9447, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37999103

RESUMEN

Patient-derived xenograft (PDX) models have been established as important preclinical cancer models, overcoming some of the limitations associated with the use of cancer cell lines. The utility of prostate cancer PDX models has been limited by an inability to genetically manipulate them in vivo and difficulties sustaining PDX-derived cancer cells in culture. Viable, short-term propagation of PDX models would allow in vitro transfection with traceable reporters or manipulation of gene expression relevant to different studies within the prostate cancer field. Here, we report an organoid culture system that supports the growth of prostate cancer PDX cells in vitro and permits genetic manipulation, substantially increasing the scope to use PDXs to study the pathobiology of prostate cancer and define potential therapeutic targets. We have established a short-term PDX-derived in vitro cell culture system which enables genetic manipulation of prostate cancer PDXs LuCaP35 and BM18. Genetically manipulated cells could be re-established as viable xenografts when re-implanted subcutaneously in immunocompromised mice and were able to be serially passaged. Tumor growth of the androgen-dependent LuCaP35 PDX was significantly inhibited following depletion of the androgen receptor (AR) in vivo. Taken together, this system provides a method to generate novel preclinical models to assess the impact of controlled genetic perturbations and allows for targeting specific genes of interest in the complex biological setting of solid tumors.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Animales , Humanos , Masculino , Ratones , Línea Celular Tumoral , Xenoinjertos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Immunology ; 169(1): 13-26, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36370035

RESUMEN

Granulomas are key histopathological features of Mycobacterium tuberculosis (Mtb) infection, with complex roles in pathogen control and dissemination. Thus, understanding drivers and regulators of granuloma formation is important for improving tuberculosis diagnosis, treatment, and prevention. Yet, molecular mechanisms underpinning granuloma formation and dynamics remain poorly understood. Here we used low-dose Mtb infection of C57BL/6 mice, which elicits structured lung granulomas composed of central macrophage clusters encased by a lymphocyte mantle, alongside the disorganized lymphocyte and macrophage clusters commonly observed in Mtb-infected mice. Using gene-deficient mice, we observed that Toll-like receptor (TLR) 2 and the TLR-related Radioprotective 105 kDa protein (RP105) contributed to the extent and spatial positioning of pathology in infected lung tissues, consistent with functional cooperation between TLR2 and RP105 in the innate immune recognition of Mtb. In mice infected with the highly virulent Mtb clinical isolate HN878, TLR2, but not RP105, positively regulated the extent of central macrophage regions within structured granulomas. Moreover, RP105, but not TLR2, promoted the formation of structured lung granulomas, suggesting that the functions of RP105 as an innate immune sensor for Mtb reach beyond its roles as TLR2 co-receptor. TLR2 and RP105 contributions to lung pathology are governed by Mtb biology, as neither receptor affected the frequency or architecture of structured granulomas in mice infected with the reference strain Mtb H37Rv. Thus, by revealing distinctive as well as cooperative functions of TLR2 and RP105 in lung pathology, our data identify TLRs as molecular determinants of TB granuloma formation and architecture, and expand understanding of how interactions between innate immune receptors and Mtb shape TB disease manifestation.


Asunto(s)
Mycobacterium tuberculosis , Animales , Ratones , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Ratones Endogámicos C57BL , Receptores Toll-Like , Pulmón , Receptores Inmunológicos , Granuloma , Inmunidad Innata
4.
Endocr Relat Cancer ; 28(5): 353-375, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33794502

RESUMEN

Hyperleptinaemia is a well-established therapeutic side effect of drugs inhibiting the androgen axis in prostate cancer (PCa), including main stay androgen deprivation therapy (ADT) and androgen targeted therapies (ATT). Given significant crossover between the adipokine hormone signalling of leptin and multiple cancer-promoting hallmark pathways, including growth, proliferation, migration, angiogenesis, metabolism and inflammation, targeting the leptin axis is therapeutically appealing, especially in advanced PCa where current therapies fail to be curative. In this study, we uncover leptin as a novel universal target in PCa and are the first to highlight increased intratumoural leptin and leptin receptor (LEPR) expression in PCa cells and patients' tumours exposed to androgen deprivation, as is observed in patients' tumours of metastatic and castrate resistant (CRPC) PCa. We also reveal the world-first preclinical evidence that demonstrates marked efficacy of targeted leptin-signalling blockade, using Allo-aca, a potent, specific, and safe LEPR peptide antagonist. Allo-aca-suppressed tumour growth and delayed progression to CRPC in mice bearing LNCaP xenografts, with reduced tumour vascularity and altered pathways of apoptosis, transcription/translation, and energetics in tumours determined as potential mechanisms underpinning anti-tumour efficacy. We highlight LEPR blockade in combination with androgen axis inhibition represents a promising new therapeutic strategy vital in advanced PCa treatment.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Antagonistas de Andrógenos/uso terapéutico , Andrógenos/metabolismo , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Leptina , Masculino , Ratones , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/metabolismo
5.
Mol Cancer Res ; 17(5): 1166-1179, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30808729

RESUMEN

De novo lipogenesis is a well-described androgen receptor (AR)-regulated metabolic pathway that supports prostate cancer tumor growth by providing fuel, membrane material, and steroid hormone precursor. In contrast, our current understanding of lipid supply from uptake of exogenous lipids and its regulation by AR is limited, and exogenous lipids may play a much more significant role in prostate cancer and disease progression than previously thought. By applying advanced automated quantitative fluorescence microscopy, we provide the most comprehensive functional analysis of lipid uptake in cancer cells to date and demonstrate that treatment of AR-positive prostate cancer cell lines with androgens results in significantly increased cellular uptake of fatty acids, cholesterol, and low-density lipoprotein particles. Consistent with a direct, regulatory role of AR in this process, androgen-enhanced lipid uptake can be blocked by the AR-antagonist enzalutamide, but is independent of proliferation and cell-cycle progression. This work for the first time comprehensively delineates the lipid transporter landscape in prostate cancer cell lines and patient samples by analysis of transcriptomics and proteomics data, including the plasma membrane proteome. We show that androgen exposure or deprivation regulates the expression of multiple lipid transporters in prostate cancer cell lines and tumor xenografts and that mRNA and protein expression of lipid transporters is enhanced in bone metastatic disease when compared with primary, localized prostate cancer. Our findings provide a strong rationale to investigate lipid uptake as a therapeutic cotarget in the fight against advanced prostate cancer in combination with inhibitors of lipogenesis to delay disease progression and metastasis. IMPLICATIONS: Prostate cancer exhibits metabolic plasticity in acquiring lipids from uptake and lipogenesis at different disease stages, indicating potential therapeutic benefit by cotargeting lipid supply.


Asunto(s)
Andrógenos/farmacología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Metabolismo de los Lípidos/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Neoplasias Óseas/genética , Línea Celular Tumoral , Colesterol/metabolismo , Progresión de la Enfermedad , Ácidos Grasos/metabolismo , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Microscopía Fluorescente , Neoplasias de la Próstata/genética , Receptores Androgénicos/metabolismo , Transducción de Señal
6.
Oncotarget ; 8(32): 52237-52255, 2017 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-28881726

RESUMEN

Proliferation and maintenance of both normal and prostate cancer (PCa) cells is highly regulated by steroid hormones, particularly androgens, and the extracellular environment. Herein, we identify the secretion of CD9 positive extracellular vesicles (EV) by LNCaP and DUCaP PCa cells in response to dihydrotestosterone (DHT) and use nano-LC-MS/MS to identify the proteins present in these EV. Subsequent bioinformatic and pathway analyses of the mass spectrometry data identified pathologically relevant pathways that may be altered by EV contents. Western blot and CD9 EV TR-FIA assay confirmed a specific increase in the amount of CD9 positive EV in DHT-treated LNCaP and DUCaP cells and treatment of cells with EV enriched with CD9 after DHT exposure can induce proliferation in androgen-deprived conditions. siRNA knockdown of endogenous CD9 in LNCaPs reduced cellular proliferation and expression of AR and prostate specific antigen (PSA) however knockdown of AR did not alter CD9 expression, also implicating CD9 as an upstream regulator of AR. Moreover CD9 positive EV were also found to be significantly higher in plasma from prostate cancer patients in comparison with benign prostatic hyperplasia patients. We conclude that CD9 positive EV are involved in mediating paracrine signalling and contributing toward prostate cancer progression.

7.
Mol Cell Endocrinol ; 420: 159-68, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26499396

RESUMEN

Simultaneous expression of highly homologous RLN1 and RLN2 genes in prostate impairs their accurate delineation. We used PacBio SMRT sequencing and RNA-Seq in LNCaP cells in order to dissect the expression of RLN1 and RLN2 variants. We identified a novel fusion transcript comprising the RLN1 and RLN2 genes and found evidence of its expression in the normal and prostate cancer tissues. The RLN1-RLN2 fusion putatively encodes RLN2 isoform with the deleted secretory signal peptide. The identification of the fusion transcript provided information to determine unique RLN1-RLN2 fusion and RLN1 regions. The RLN1-RLN2 fusion was co-expressed with RLN1 in LNCaP cells, but the two gene products were inversely regulated by androgens. We showed that RLN1 is underrepresented in common PCa cell lines in comparison to normal and PCa tissue. The current study brings a highly relevant update to the relaxin field, and will encourage further studies of RLN1 and RLN2 in PCa and broader.


Asunto(s)
Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/genética , Relaxina/genética , Andrógenos/farmacología , Línea Celular Tumoral , Exones/genética , Humanos , Masculino , Proteínas de Fusión Oncogénica/metabolismo , Sistemas de Lectura Abierta/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Relaxina/metabolismo , Análisis de Secuencia de ARN
8.
Am J Physiol Endocrinol Metab ; 301(4): E727-35, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21750267

RESUMEN

Androgens influence prostate growth and development, so androgen withdrawal can control progression of prostate diseases. Although estrogen treatment was originally used to induce androgen withdrawal, more recently direct estrogen effects on the prostate have been recognized, but the nature of androgen-estrogen interactions within the prostate remain poorly understood. To characterize androgen effects on estrogen sensitivity in the mouse prostate, we contrasted models of castration-induced androgen withdrawal in the prostate stromal and epithelial compartments with a prostate epithelial androgen receptor (AR) knockout (PEARKO) mouse model of selective epithelial AR inactivation. Castration markedly increased prostate epithelial estrogen receptor (ER)α immunoreactivity compared with very low ERα expression in intact males. Similarly, strong basal and luminal ERα expression was detected in PEARKO prostate of intact males, suggesting that epithelial AR activity regulated epithelial ERα expression. ERß was strongly expressed in intact, castrated, and PEARKO prostate. However, strong clusters of epithelial ERß positivity coincided with epithelial stratification in PEARKO prostate. In vivo estrogen sensitivity was increased in PEARKO males, with greater estradiol-induced prostate growth and epithelial proliferation leading to squamous metaplasia, featuring markedly increased epithelial proliferation, thickening, and keratinization compared with littermate controls. Our results suggest that ERα expression in the prostate epithelial cells is regulated by local, epithelia-specific, androgen-dependent mechanisms, and this imbalance in the AR- and ER-mediated signaling sensitizes the mature prostate to exogenous estrogens.


Asunto(s)
Células Epiteliales/metabolismo , Estradiol/farmacología , Estrógenos/farmacología , Próstata/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Células Epiteliales/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Próstata/efectos de los fármacos , Receptores Androgénicos/genética , Receptores de Estrógenos/genética
9.
J Endocrinol ; 207(1): 27-34, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20675302

RESUMEN

Aromatase inhibitors have been increasingly used in boys with growth retardation to prolong the duration of growth and increase final height. Multiple important roles of oestrogen in males point to potential adverse effects of this strategy. Although the deleterious effects of aromatase deficiency in early childhood and adulthood are well documented, there is limited information about the potential long-term adverse effects of peripubertal aromatase inhibition. To address this issue, we evaluated short-term and long-term effects of peripubertal aromatase inhibition in an animal model. Peripubertal male Wistar rats were treated with aromatase inhibitor letrozole or placebo and followed until adulthood. Letrozole treatment caused sustained reduction in bone strength and alteration in skeletal geometry, lowering of IGF1 levels, inhibition of growth resulting in significantly lower weight and length of treated animals and development of focal prostatic hyperplasia. Our observation of adverse long-term effects after peripubertal male rats were exposed to aromatase inhibitors highlights the need for further characterisation of long-term adverse effects of aromatase inhibitors in peripubertal boys before further widespread use is accepted. Furthermore, this suggests the need to develop more selective oestrogen inhibition strategies in order to inhibit oestrogen action on the growth plate, while beneficial effects in other tissues are preserved.


Asunto(s)
Inhibidores de la Aromatasa/efectos adversos , Desarrollo Óseo/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/fisiopatología , Hiperplasia Prostática/etiología , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/fisiología , Desarrollo Óseo/fisiología , Huesos/patología , Niño , Trastornos del Crecimiento/tratamiento farmacológico , Trastornos del Crecimiento/patología , Trastornos del Crecimiento/fisiopatología , Humanos , Letrozol , Hormona Luteinizante/sangre , Masculino , Nitrilos/efectos adversos , Próstata/efectos de los fármacos , Próstata/patología , Hiperplasia Prostática/patología , Ratas , Ratas Wistar , Maduración Sexual/fisiología , Testículo/efectos de los fármacos , Testículo/patología , Triazoles/efectos adversos
10.
Proc Natl Acad Sci U S A ; 107(7): 3123-8, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20133657

RESUMEN

Prostate cancer (PCa) and benign prostatic hyperplasia (BPH) are androgen-dependent diseases commonly treated by inhibiting androgen action. However, androgen ablation or castration fail to target androgen-independent cells implicated in disease etiology and recurrence. Mechanistically different to castration, this study shows beneficial proapoptotic actions of estrogen receptor-beta (ERbeta) in BPH and PCa. ERbeta agonist induces apoptosis in prostatic stromal, luminal and castrate-resistant basal epithelial cells of estrogen-deficient aromatase knock-out mice. This occurs via extrinsic (caspase-8) pathways, without reducing serum hormones, and perturbs the regenerative capacity of the epithelium. TNFalpha knock-out mice fail to respond to ERbeta agonist, demonstrating the requirement for TNFalpha signaling. In human tissues, ERbeta agonist induces apoptosis in stroma and epithelium of xenografted BPH specimens, including in the CD133(+) enriched putative stem/progenitor cells isolated from BPH-1 cells in vitro. In PCa, ERbeta causes apoptosis in Gleason Grade 7 xenografted tissues and androgen-independent cells lines (PC3 and DU145) via caspase-8. These data provide evidence of the beneficial effects of ERbeta agonist on epithelium and stroma of BPH, as well as androgen-independent tumor cells implicated in recurrent disease. Our data are indicative of the therapeutic potential of ERbeta agonist for treatment of PCa and/or BPH with or without androgen withdrawal.


Asunto(s)
Apoptosis/fisiología , Receptor beta de Estrógeno/metabolismo , Hiperplasia/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Análisis de Varianza , Andrógenos/metabolismo , Animales , Línea Celular Tumoral , Receptor beta de Estrógeno/agonistas , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Próstata/metabolismo , Factor de Necrosis Tumoral alfa/genética
11.
Environ Health Perspect ; 116(7): 923-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18629315

RESUMEN

BACKGROUND: Androgens are critical for specifying prostate development, with the fetal prostate sensitive to altered hormone levels and endocrine-disrupting chemicals (EDCs) that exhibit estrogenic or antiandrogenic properties. Prostatic inflammation (prostatitis) affects 9% of men of all ages, and > 90% of cases are of unknown etiology. OBJECTIVES: In this study we aimed to evaluate effects of in utero exposure to the antiandrogenic EDC vinclozolin, during the period of male reproductive tract development, on neonatal, prepubertal, and postpubertal prostate gland function of male offspring. METHODS: Fetal rats were exposed to vinclozolin (100 mg/kg body weight) or vehicle control (2.5 mL/kg body weight) in utero from gestational day 14 (GD14) to GD19 via oral administration to pregnant dams. Tissue analysis was carried out when male offspring were 0, 4, or 8 weeks of age. RESULTS: In utero exposure to vinclozolin was insufficient to perturb prostatic development and branching, although expression of androgen receptor and mesenchymal fibroblast growth factor-10 was down-regulated. Prostate histology remained normal until puberty, but 100% of animals displayed prostatitis postpubertally (56 days of age). Prostatic inflammation was associated with phosphorylation and nuclear translocation of nuclear factor-kappa B (NFkappaB) and postpubertal activation of proinflammatory NFkappaB-dependent genes, including the chemokine interleukin-8 and the cytokine transforming growth factor-beta1. Significantly, inflammation arising from vinclozolin exposure was not associated with the emergence of premalignant lesions, such as prostatic intra-epithelial neoplasia or proliferative inflammatory atrophy, and hence mimics nonbacterial early-onset prostatitis that commonly occurs in young men. CONCLUSIONS: These data are the first to unequivocally implicate EDCs as a causative factor and fill an important knowledge gap on the etiology of prostatitis.


Asunto(s)
Antagonistas de Andrógenos/toxicidad , Intercambio Materno-Fetal , Oxazoles/toxicidad , Efectos Tardíos de la Exposición Prenatal , Prostatitis/inducido químicamente , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , Epitelio/crecimiento & desarrollo , Epitelio/patología , Femenino , Humanos , Interleucina-8/metabolismo , Masculino , Morfogénesis , FN-kappa B/metabolismo , Embarazo , Próstata/crecimiento & desarrollo , Próstata/patología , Prostatitis/metabolismo , Prostatitis/patología , Ratas , Ratas Sprague-Dawley , Maduración Sexual , Factor de Crecimiento Transformador beta1/metabolismo
12.
Differentiation ; 76(6): 660-70, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18557760

RESUMEN

Both androgens and estrogens play a significant role in the prostate and are critical for normal prostate growth and development, as well as the maintenance of adult prostatic homeostasis throughout life. It is the balance of these two hormones, rather than each individually, that is important for prostatic development and differentiation. Estrogen action is mediated by the estrogen receptors, ERalpha and ERbeta. ERalpha is expressed throughout the prostatic tissue during fetal and early neonatal life, and if activated inappropriately, produces late-life disease, including inflammation and emergence of pre-malignant pathologies. In contrast, ERbeta expression is initiated after ERalpha, is localized primarily to the epithelium, and appears to be important during later periods of development such as puberty and adulthood, acting to regulate cellular proliferation and differentiation in the adult tissue. Therefore, there is also a spatial and temporal balance between ERalpha and ERbeta that is critical for development. Together with the shifting balance between androgens and estrogens themselves, the subtle, yet critical, balance between the activity of ERalpha and ERbeta is what ultimately determines the response of the prostate to estrogen, and is crucial for prostate health.


Asunto(s)
Estrógenos/farmacología , Próstata/crecimiento & desarrollo , Animales , Diferenciación Celular , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/efectos de los fármacos , Humanos , Masculino , Próstata/efectos de los fármacos
13.
FASEB J ; 22(5): 1512-20, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18055862

RESUMEN

It was recently demonstrated that antiestrogens prevented prostate cancer (PRCA) in men. The source of estradiol (E2) that contributes to carcinogenesis, as well as the selected estrogen receptor (ER) signaling pathway, is unknown. To evaluate estrogen's effects in carcinogenesis, we developed a new model of PRCA utilizing testosterone and E2 to stimulate PRCA. To determine whether local in situ production of E2 affected incidence of PRCA, aromatase-knockout (ArKO) mice were evaluated. In contrast to the wild-type mice, ArKO mice had reduced incidences of PRCA, which implicates in situ production of E2 as an important determinant of PRCA. To determine whether E2-mediated responses were due to ER alpha or ER beta signaling, ER alpha-knockout (alphaERKO) or ERbeta-knockout (betaERKO) mice were used. Prostates from betaERKO mice underwent biochemical and histological carcinogenesis similar to wild-type mice, whereas prostates from alphaERKO mice remained free of pathology. These data suggest that effective prevention of carcinogenesis will require antagonism of ER alpha but not ER beta. This mouse model provides a means to examine genetic gain and loss of function and determine the efficacy of therapeutics on prostatic carcinogenesis.


Asunto(s)
Aromatasa/fisiología , Estradiol/biosíntesis , Receptor alfa de Estrógeno/fisiología , Neoplasias de la Próstata/inducido químicamente , Animales , Aromatasa/deficiencia , Receptor alfa de Estrógeno/deficiencia , Receptor beta de Estrógeno/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Próstata/efectos de los fármacos , Próstata/patología , Neoplasias de la Próstata/patología , Testosterona
14.
J Mol Endocrinol ; 39(3): 183-8, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17766643

RESUMEN

Although modern biotechnology has provided us with a greater understanding of the molecular events in endocrine-related diseases, such as benign prostatic hyperplasia and prostate cancer, these conditions continue to be a significant healthcare problem world-wide. As the number of men afflicted by these diseases will only continue to grow with the aging population, finding new strategies and new therapeutic options for the treatment of both of these diseases is crucial. A better knowledge of the mechanisms of hormone action is pivotal to making progress in the development of new hormone-based therapies. This is fundamental to increasing our understanding of the endocrine, paracrine, and autocrine signaling mechanisms in the prostate and in prostate disease, distinguishing the effects and role of each, and identifying where and how this communication goes wrong.


Asunto(s)
Comunicación Autocrina/fisiología , Estrógenos/metabolismo , Comunicación Paracrina/fisiología , Próstata/fisiología , Andrógenos/metabolismo , Animales , Proliferación Celular , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Inflamación/metabolismo , Masculino , Próstata/patología , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo
15.
Endocrinology ; 148(5): 2264-72, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17317769

RESUMEN

Prostate development and maturation requires stromal-epithelial interactions and androgen action via the androgen receptor (AR) within these compartments. However, the specific roles of epithelial and stromal AR in postnatal prostate differentiation are unclear. We used Cre-LoxP technology to determine the prostate phenotype in mice with epithelial-selective genetic inactivation of the AR leaving the stromal AR functionally intact. We find that prostate development abolished in mice globally lacking a functional AR can be rescued by restricting the AR knockout to the postnatal prostate epithelium. We show that, at 8 wk of age, prostate epithelial AR knockout (PEARKO) mice exhibit prostate development with normal branching morphogenesis but lobe-specific decrease in prostate weight and hindered structural and functional differentiation of the mature prostate epithelium. No change was observed in PEARKO testis weight or serum testosterone compared with littermate controls. The most striking change was increased proliferation and abnormal lesions of epithelial cells predominantly in the anterior lobe of PEARKO mice. These findings highlight the vital role of stromal AR in postnatal prostate growth and structural differentiation and emphasize the requirement of epithelial AR in maintaining functional differentiation and restraining proliferation of epithelial cells in a lobe-specific manner. This unique PEARKO mouse provides a new paradigm with which to define the molecular mechanisms of the androgen signaling in mature prostate lobes in vivo and provides insight into the identification of better targets for treatment of prostate cancer and hyperplasia.


Asunto(s)
Células Epiteliales/patología , Próstata/crecimiento & desarrollo , Próstata/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Células Epiteliales/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Integrasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Fenotipo , Próstata/fisiología , ARN Mensajero/metabolismo , Células del Estroma/metabolismo , Células del Estroma/fisiología , Transgenes
16.
Endocrinology ; 148(2): 566-74, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17068134

RESUMEN

Estrogens, acting via estrogen receptors (ER) alpha and beta, exert direct and indirect actions on prostate growth and differentiation. Previous studies using animal models to determine the role of ERbeta in the prostate have been problematic because the centrally mediated response to estrogen results in reduced androgen levels and prostatic epithelial regression, potentially masking any direct effects via ERbeta. This study overcomes this problem by using the estrogen-deficient aromatase knockout mouse and tissue recombination to provide new insight into estrogen action on prostate growth and pathology. Homo- and heterotypic aromatase knockout tissue recombinants revealed stromal aromatase deficiency induced hyperplasia in normal prostatic epithelium due to disruption of paracrine interaction between stroma and epithelia. Treatment of tissue recombinants with an ERbeta-specific agonist demonstrated that stimulation of ERbeta elicits antiproliferative responses in epithelium that are not influenced by alterations to systemic androgen levels or the activation of ERalpha. Additionally, work performed with intact aromatase knockout mice demonstrated that the administration of an ERbeta-specific agonist ablated preexisting prostatic epithelial hyperplasia, whereas an ERalpha-specific agonist did not. Therefore, failed activation of ERbeta, resulting from local stromal aromatase deficiency, in conjunction with increased androgen levels, results in increased epithelial cell proliferation and prostatic hyperplasia. These data demonstrate essential and beneficial effects of estrogens that are necessary for normal growth of the prostate and distinguishes them from those that adversely alter prostate growth and differentiation. This highlights the potential of selective estrogen-receptor modulators, rather than aromatase inhibitors, for the management of dysregulated prostate growth.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Próstata/fisiopatología , Hiperplasia Prostática/fisiopatología , Animales , Animales Recién Nacidos , Aromatasa/deficiencia , Proliferación Celular , Epitelio/metabolismo , Epitelio/trasplante , Receptor beta de Estrógeno/agonistas , Estrógenos/deficiencia , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Ratones SCID , Próstata/metabolismo , Próstata/patología , Hiperplasia Prostática/etiología , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Receptores de Superficie Celular/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Transducción de Señal , Células del Estroma/metabolismo , Células del Estroma/trasplante , Distribución Tisular , Trasplante Heterotópico
17.
Am J Pathol ; 168(6): 1869-78, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16723702

RESUMEN

Exposure of newborn male mice to estrogens is associated with age-related changes in prostate size and induction of epithelial hyperplasia and dysplasia. Whether these changes directly result from systemic estrogen administration or indirect effects of estrogens on systemic testosterone levels is unclear. We have addressed this question using aromatase-knockout (ArKO) mice that are estrogen-deficient during their lifespan but have elevated androgen levels and develop prostate enlargement and hyperplasia (McPherson SJ, Wang H, Jones ME, Pedersen J, Iismaa TP, Wreford N, Simpson ER, Risbridger GP: Endocrinology 2001, 142:2458-2467). Circulating testosterone and dihydrotestosterone levels were significantly decreased by neonatal diethylstilbestrol treatment, remained suppressed in adult wild-type mice, but rapidly returned to control levels in ArKO animals. However, adult prostate weight and luminal size were reduced in both wild-type and ArKO animals. Because both wild-type and ArKO mice developed epithelial hyperplasia and inflammation following neonatal diethylstilbestrol treatment, this validates that estrogens directly cause prostatic inflammation and epithelial hyperplasia. Furthermore, because ArKO mice are estrogen-deficient, this study demonstrates the sensitivity of the neonatal period to estrogen exposure and the long range and permanent nature of the prostatic responses that occur. Finally, this study establishes the ArKO mouse model of estrogen deficiency as a unique approach to study the effects of estrogens, estrogenic factors, and endocrine disruptors on prostate development.


Asunto(s)
Aromatasa/genética , Dietilestilbestrol/farmacología , Estrógenos no Esteroides/farmacología , Inflamación , Próstata/patología , Animales , Peso Corporal/efectos de los fármacos , Estrógenos/metabolismo , Femenino , Masculino , Exposición Materna , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Próstata/efectos de los fármacos , Próstata/metabolismo
18.
Biol Reprod ; 71(5): 1712-23, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15269096

RESUMEN

Estradiol-17beta (E(2)) affects late follicular development, whereas primordial follicle differentiation and early activation are believed to be independent of E(2). To test this hypothesis we compared numbers of primordial and primary follicles in wild-type and E(2)-deficient, aromatase knockout (ArKO) mice, and the immunohistochemical staining or mRNA expression of Mullerian inhibiting substance (MIS), Wilms tumor 1 (WT-1), and growth differentiation factor (GDF9), which are known to effect early follicular differentiation. Proliferating cell nuclear antigen (PCNA) staining was a marker of proliferative index. The effects of E(2) replacement for 3 wk in 7-wk-old ArKO and wild-type mice on these parameters were also tested. ArKO mice had reduced numbers of primordial and primary follicles compared with wild-type mice (63%, P < 0.001 and 60%, P = 0.062, respectively). This reduction was not corrected by E(2) treatment, suggesting that E(2) affects the initial formation or activation of primordial follicles. There was a significant increase in the diameters of the oocytes in primordial follicles of ArKO mice compared with mice of the wild type. There were no differences in the immunostaining of MIS, WT-1, and PCNA in primordial and primary follicles between wild-type and ArKO mice. The only difference was as a consequence of Sertoli and Leydig cells that develop in ovaries of ArKO mice. GDF9 mRNA expression was markedly increased in ArKO ovaries. E(2) treatment restored the ovarian follicular morphology in ArKO mice, and consequently the immunostaining patterns, but had no effect on early follicle numbers. In conclusion, E(2) has a role in controlling the size of the oocyte and primordial follicle pool in mice.


Asunto(s)
Estradiol/fisiología , Folículo Ovárico/fisiología , Animales , Hormona Antimülleriana , Aromatasa/deficiencia , Aromatasa/genética , Proteína Morfogenética Ósea 15 , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Tamaño de la Célula , Proteínas de Unión al ADN/metabolismo , Estradiol/deficiencia , Estradiol/farmacología , Femenino , Glicoproteínas/metabolismo , Factor 9 de Diferenciación de Crecimiento , Inmunohistoquímica/métodos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados/genética , Proteínas Nucleares/metabolismo , Oocitos/citología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Empalme de ARN , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Coloración y Etiquetado , Hormonas Testiculares/metabolismo
19.
Prostate ; 56(1): 54-64, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12746847

RESUMEN

BACKGROUND: Red clover (RC)-derived dietary isoflavones have been implicated as potential preventative agents for the development and prevalence of non-malignant prostate diseases. This study investigated whether dietary isoflavones inhibit prostate growth in vivo in the aromatase knock-out (ArKO) mouse that exhibits lifelong elevation of androgens leading to prostate enlargement. METHODS: Adult (11-week-old) wild-type (WT) and ArKO mice were fed on protein matched isoflavones free (IF) and RC (isoflavone rich) diets for 28 days. Individual prostate lobes and testes were weighed and collected for histological analysis and serum androgens were measured. Responses were compared to castration and estrogen administration to ArKO mice to determine the mechanism of action. RESULTS: ArKO mice fed on IF diet exhibited enlarged prostate lobes and elevated serum androgens compared to WT mice. Following 28 days of RC diet, ArKO VP, AP, and SV weights were reduced to WT weights, although testis and body weights remained unaltered. Stereological analysis of VPs revealed a reduction in all components of the tissue, particularly the lumen. The RC diet reduced ArKO serum testosterone and dihydrotestosterone to WT levels. In comparison to castration and estrogen administration, the dietary isoflavones were shown to be anti-androgenic rather than weakly estrogenic, mimicking responses observed in the castrated ArKO, rather than estrogen treated ArKOs. CONCLUSIONS: This study demonstrates that RC-derived isoflavones have a significant effect on prostatic growth, and are capable of reducing the enlarged non-malignant prostate phenotype of the adult ArKO mouse, by acting as anti-androgenic agents rather than weak estrogenic substances.


Asunto(s)
Aromatasa/genética , Isoflavonas/farmacología , Hiperplasia Prostática/tratamiento farmacológico , Trifolium , Alimentación Animal , Animales , Peso Corporal/efectos de los fármacos , Dihidrotestosterona/sangre , Estrógenos/farmacología , Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Orquiectomía , Tamaño de los Órganos/efectos de los fármacos , Fitoterapia , Preparaciones de Plantas/farmacología , Próstata/patología , Hiperplasia Prostática/sangre , Testículo/patología , Testosterona/sangre
20.
Cancer Epidemiol Biomarkers Prev ; 11(12): 1689-96, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12496063

RESUMEN

Epidemiological evidence suggests a geographical basis for the incidence of prostate cancer and dietary factors, including isoflavone consumption, may be linked to this phenomenon. This paper reports a nonrandomized, nonblinded trial with historically matched controls from archival tissue designed to determine the effects of acute exposure to a dietary supplement of isoflavones in men with clinically significant prostate cancer before radical prostatectomy. Thirty-eight patients were recruited to the study upon diagnosis of prostate cancer. Before surgery, 20 men consumed 160 mg/day of red clover-derived dietary isoflavones, containing a mixture of genistein, daidzein, formononetin, and biochanin A. Serum PSA, testosterone, and biochemical factors were measured, and clinical and pathological parameters were recorded. The incidence of apoptosis in prostate tumor cells from radical prostatectomy specimens was compared between 18 treated and 18 untreated control tissues. There were no significant differences between pre- and posttreatment serum PSA, Gleason score, serum testosterone, or biochemical factors in the treated patients (P > 0.05). Apoptosis in radical prostatectomy specimens from treated patients was significantly higher than in control subjects (P = 0.0018), specifically in regions of low to moderate-grade cancer (Gleason grade 1-3). No adverse events related to the treatment were reported. This report suggests that dietary isoflavones may halt the progression of prostate cancer by inducing apoptosis in low to moderate-grade tumors, potentially contributing to the lower incidence of clinically significant disease in Asian men. The assessment of new prostatic therapies aimed at increasing apoptosis should control for intake of dietary isoflavones.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Apoptosis/fisiología , Isoflavonas/administración & dosificación , Fitoterapia/métodos , Antígeno Prostático Específico/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Trifolium , Adenocarcinoma/cirugía , Anciano , Apoptosis/efectos de los fármacos , Biopsia con Aguja , Suplementos Dietéticos , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Extractos Vegetales/uso terapéutico , Cuidados Preoperatorios , Estudios Prospectivos , Antígeno Prostático Específico/análisis , Prostatectomía/métodos , Neoplasias de la Próstata/cirugía , Valores de Referencia , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...