Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 356
Filtrar
1.
Oncogene ; 37(2): 185-196, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-28892047

RESUMO

Tumor complexity and intratumor heterogeneity contribute to subclonal diversity. Despite advances in next-generation sequencing (NGS) and bioinformatics, detecting rare mutations in primary tumors and metastases contributing to subclonal diversity is a challenge for precision genomics. Here, in order to identify rare mutations, we adapted a recently described epithelial reprograming assay for short-term propagation of epithelial cells from primary and metastatic tumors. Using this approach, we expanded minor clones and obtained epithelial cell-specific DNA/RNA for quantitative NGS analysis. Comparative Ampliseq Comprehensive Cancer Panel sequence analyses were performed on DNA from unprocessed breast tumor and tumor cells propagated from the same tumor. We identified previously uncharacterized mutations present only in the cultured tumor cells, a subset of which has been reported in brain metastatic but not primary breast tumors. In addition, whole-genome sequencing identified mutations enriched in liver metastases of various cancers, including Notch pathway mutations/chromosomal inversions in 5/5 liver metastases, irrespective of cancer types. Mutations/rearrangements in FHIT, involved in purine metabolism, were detected in 4/5 liver metastases, and the same four liver metastases shared mutations in 32 genes, including mutations of different HLA-DR family members affecting OX40 signaling pathway, which could impact the immune response to metastatic cells. Pathway analyses of all mutated genes in liver metastases showed aberrant tumor necrosis factor and transforming growth factor signaling in metastatic cells. Epigenetic regulators including KMT2C/MLL3 and ARID1B, which are mutated in >50% of hepatocellular carcinomas, were also mutated in liver metastases. Thus, irrespective of cancer types, organ-specific metastases may share common genomic aberrations. Since recent studies show independent evolution of primary tumors and metastases and in most cases mutation burden is higher in metastases than primary tumors, the method described here may allow early detection of subclonal somatic alterations associated with metastatic progression and potentially identify therapeutically actionable, metastasis-specific genomic aberrations.


Assuntos
Análise Mutacional de DNA/métodos , Regulação Neoplásica da Expressão Gênica/genética , Genômica/métodos , Neoplasias/genética , Animais , Biópsia , Epigênese Genética/genética , Células Epiteliais/patologia , Estudos de Viabilidade , Fibroblastos , Redes Reguladoras de Genes/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Camundongos , Mutação , Neoplasias/patologia , Cultura Primária de Células , Transdução de Sinais/genética , Células Tumorais Cultivadas , Sequenciamento Completo do Genoma
2.
Nat Commun ; 8: 15461, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28580940

RESUMO

Free-electron lasers providing ultra-short high-brightness pulses of X-ray radiation have great potential for a wide impact on science, and are a critical element for unravelling the structural dynamics of matter. To fully harness this potential, we must accurately know the X-ray properties: intensity, spectrum and temporal profile. Owing to the inherent fluctuations in free-electron lasers, this mandates a full characterization of the properties for each and every pulse. While diagnostics of these properties exist, they are often invasive and many cannot operate at a high-repetition rate. Here, we present a technique for circumventing this limitation. Employing a machine learning strategy, we can accurately predict X-ray properties for every shot using only parameters that are easily recorded at high-repetition rate, by training a model on a small set of fully diagnosed pulses. This opens the door to fully realizing the promise of next-generation high-repetition rate X-ray lasers.

3.
Int J Obes (Lond) ; 36(7): 982-5, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21876548

RESUMO

OBJECTIVE: In the adipose tissue, activation of AMP-activated protein kinase (AMPK) by phosphorylation favours local fatty acid oxidation and inhibition of lipogenesis. We have previously shown that the potent androgen dihydrotestosterone (DHT) can inhibit phosphorylation of AMPK in adipose tissue and 3T3-L1 adipocytes in a dose-dependent manner. This negative effect of DHT was reversed by oestrogen treatment. The purpose of this current study was to determine the underlying mechanisms whereby androgens and oestrogens can regulate AMPK phosphorylation in adipocytes, and whether this mechanism is receptor dependent. RESULTS: Phosphorylation of AMPK was assessed by western blot in cells treated for 24 h with testosterone or DHT (1-1000 nM). Testosterone and DHT significantly inhibited basal phosphorylation of AMPK. Addition of the androgen receptor antagonist Flutamide (1 µM) to the media reversed the negative effect of testosterone and DHT by returning AMPK phosphorylation levels to those of basal. To further dissect the mechanism underlying AMPK inhibition by testosterone or DHT, we examined the mRNA expression of the upstream activator of AMPK, namely LKB1. Testosterone and DHT treatment of murine 3T3-L1 or human SGBS adipocytes for 24 h significantly decreased the mRNA expression of LKB1. In contrast, 17ß-estradiol treatment increased LKB1 mRNA, an effect mediated by oestrogen receptor alpha. CONCLUSION: We conclude that regulation of AMPK phosphorylation by androgens and oestrogens is receptor-dependent, and demonstrate for the first time that LKB1 is regulated by sex hormones in adipocytes.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Lipogênese/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Western Blotting , Células Cultivadas , Di-Hidrotestosterona/farmacologia , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , Hormônios Esteroides Gonadais/farmacologia , Humanos , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Testosterona/metabolismo
4.
Handb Exp Pharmacol ; (198): 29-35, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20839084

RESUMO

Estrogens influence fertility and infertility in animals. This chapter reviews the use of estrogen as a contraceptive through the regulation of its production and action. It is concluded that the use of specific agonists and antagonists of estrogen action that avoid the global and unwanted side effects of estrogen offers new potential methods of contraception.


Assuntos
Estrogênios/fisiologia , Reprodução/fisiologia , Transdução de Sinais/fisiologia , Animais , Anticoncepcionais Orais Hormonais/efeitos adversos , Anticoncepcionais Orais Hormonais/farmacologia , Estrogênios/biossíntese , Feminino , Humanos
5.
Br J Ophthalmol ; 93(9): 1172-6, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19414441

RESUMO

AIMS: To report the treatment outcomes and complication rates of stereotactic radiotherapy in the management of patients with juxtapapillary choroidal melanoma. METHODS: A retrospective review of 64 consecutive patients with juxtapapillary choroidal melanoma, located within 2 mm of the optic disc, treated with stereotactic radiotherapy at Princess Margaret Hospital between October 1998 and January 2006. RESULTS: The median age was 63 years. The median tumour height was 4.2 mm, and median largest basal diameter was 9.8 mm. The prescribed radiation dose was 70 Gy in five fractions over 10 days, and the median follow-up was 37 months. Post-treatment, the actuarial rates of local tumour control, metastases and survival at 37 months were 94%, 15% and 90%, respectively. Actuarial rates of radiation-induced complications at 37 months were neovascular glaucoma 42%, cataract 53%, retinopathy 81% and optic neuropathy 64%. Secondary enucleation was necessary for 10 patients (16%), in four patients for tumour recurrence and in six for painful neovascular glaucoma. CONCLUSIONS: Stereotactic radiotherapy offers a non-invasive alternative to enucleation and brachytherapy in the management of juxtapapillary choroidal melanoma with a high tumour control rate, however, at the expense of a significant rate of long-term ocular complications.


Assuntos
Neoplasias da Coroide/cirurgia , Melanoma/cirurgia , Recidiva Local de Neoplasia/cirurgia , Radiocirurgia/métodos , Neoplasias da Coroide/patologia , Fracionamento da Dose de Radiação , Feminino , Seguimentos , Humanos , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Radiocirurgia/efeitos adversos , Estudos Retrospectivos , Resultado do Tratamento , Acuidade Visual/fisiologia
6.
Endocr Rev ; 30(4): 343-75, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19389994

RESUMO

Aromatase is the enzyme that catalyzes the conversion of androgens to estrogens. Initial studies of its enzymatic activity and function took place in an environment focused on estrogen as a component of the birth control pill. At an early stage, investigators recognized that inhibition of this enzyme could have major practical applications for treatment of hormone-dependent breast cancer, alterations of ovarian and endometrial function, and treatment of benign disorders such as gynecomastia. Two general approaches ultimately led to the development of potent and selective aromatase inhibitors. One targeted the enzyme using analogs of natural steroidal substrates to work out the relationships between structure and function. The other approach initially sought to block adrenal function as a treatment for breast cancer but led to the serendipitous finding that a nonsteroidal P450 steroidogenesis inhibitor, aminoglutethimide, served as a potent but nonselective aromatase inhibitor. Proof of the therapeutic concept of aromatase inhibition involved a variety of studies with aminoglutethimide and the selective steroidal inhibitor, formestane. The requirement for even more potent and selective inhibitors led to intensive molecular studies to identify the structure of aromatase, to development of high-sensitivity estrogen assays, and to "mega" clinical trials of the third-generation aromatase inhibitors, letrozole, anastrozole, and exemestane, which are now in clinical use in breast cancer. During these studies, unexpected findings led investigators to appreciate the important role of estrogens in males as well as in females and in multiple organs, particularly the bone and brain. These studies identified the important regulatory properties of aromatase acting in an autocrine, paracrine, intracrine, neurocrine, and juxtacrine fashion and the organ-specific enhancers and promoters controlling its transcription. The saga of these studies of aromatase and the ultimate utilization of inhibitors as highly effective treatments of breast cancer and for use in reproductive disorders serves as the basis for this first Endocrine Reviews history manuscript.


Assuntos
Inibidores da Aromatase/uso terapêutico , Aromatase/fisiologia , Neoplasias da Mama/tratamento farmacológico , Aromatase/química , Osso e Ossos/fisiologia , Encéfalo/fisiologia , Doenças do Sistema Endócrino/tratamento farmacológico , Feminino , Humanos , Masculino , Reprodução/fisiologia
7.
Int J Impot Res ; 20(3): 315-23, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18273027

RESUMO

The current study describes the presence of sexually dimorphic cell groups within the MPO of the 129SvEv, but not the C57BL/6J strain of mice. We detected galanin-positive clusters of cells located in the MPO of 129SvEv and C57BL/6J mice, with sex differences found only in the 129SvEv strain. Aromatase-positive and dense Nissl-counterstained clusters of cells were identified only in the MPO of 129SvEv mice, but not in the C57BL/6J strain. Furthermore, this study has demonstrated that the volume of these sexually dimorphic cell groups is regulated by estrogen, but not testosterone, as male aromatase knockout mice (which have high levels of testosterone, but no estrogen) show reduced cell group volumes. These structural changes, which occur in an estrogen-deficient state may influence male sexual behaviors.


Assuntos
Aromatase/biossíntese , Estrogênios/fisiologia , Área Pré-Óptica/citologia , Área Pré-Óptica/fisiologia , Caracteres Sexuais , Animais , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL
8.
J Steroid Biochem Mol Biol ; 106(1-5): 3-7, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17643292

RESUMO

Estrogen has an important role to play in energy homeostasis in both men and mice. Lack of estrogen results in the development of a metabolic syndrome in humans and rodents, including excess adiposity, hepatic steatosis (in male but not female aromatase knockout (ArKO) mice) and insulin resistance. Estrogen replacement results in a prompt reversal of the energy imbalance symptoms associated with estrogen deficiency. A corollary to the perturbed energy balance observed in the ArKO mouse is the death by apoptosis of dopaminergic neurons in the hypothalamic arcuate nucleus of male ArKO mice, an area of the brain pivotal to the regulation of energy uptake, storage, and mobilisation. An extension of our work exploring the relationship between estrogen and adiposity has been to examine the role played by androgens in energy balance. We have demonstrated that an increased androgen to estrogen ratio can promote visceral fat accumulation in the rodent by inhibiting AMPK activation and stimulating lipogenesis. Therefore, understanding the regulation of energy homeostasis is becoming an increasingly fascinating challenge, as the number of contributors, their communications, and the complexity of their interactions, involved in the preservation of this equilibrium continues to increase. Models of aromatase deficiency, both naturally occurring and engineered, will continue to provide valuable insights into energy homeostasis.


Assuntos
Adiposidade , Aromatase/deficiência , Aromatase/metabolismo , Estrogênios/metabolismo , Modelos Biológicos , Animais , Aromatase/genética , Homeostase , Humanos
9.
J Neuroendocrinol ; 18(8): 567-76, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16867177

RESUMO

The effect of circulating oestrogen deficiency on sleep regulation and locomotor activity was investigated in aromatase cytochrome P450 deficient mice (ArKO) and wild-type (WT) controls. Sleep was recorded in 3-month old mice during a 24-h baseline day, 6-h sleep deprivation (SD) and 18-h recovery, and activity was recorded at the age of 3, 9 and 12 months. In mice deficient of oestrogen, the total amount of sleep per 24 h was the same as in WT controls. However, in ArKO mice, sleep was enhanced in the dark period at the expense of sleep in the light phase, and was more fragmented than sleep in WT mice. This redistribution of sleep resulted in a damped amplitude of slow-wave activity (SWA; power between 0.75-4.0 Hz) in non-rapid eye movement sleep across 24 h. After SD, the rebound of sleep and SWA was similar between the genotypes, suggesting that oestrogen deficiency does not affect the mechanisms maintaining the homeostatic balance between the amount of sleep and its intensity. Motor activity decreased with age in both genotypes and was lower in ArKO mice compared to WT at all three ages. After SD, the amount of rest in 3-month old WT mice increased above baseline and was more consolidated. Both effects were less pronounced in ArKO mice, reflecting the baseline differences between the genotypes. The results indicate that despite the pronounced redistribution of sleep and motor activity in oestrogen deficient mice, the basic homeostatic mechanisms of sleep regulation in ArKO mice remain intact.


Assuntos
Aromatase/fisiologia , Estrogênios/fisiologia , Atividade Motora/fisiologia , Fases do Sono/fisiologia , Vigília/fisiologia , Fatores Etários , Animais , Aromatase/deficiência , Ritmo Circadiano/fisiologia , Eletroencefalografia , Estrogênios/deficiência , Estrogênios/metabolismo , Feminino , Homeostase/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estatísticas não Paramétricas
10.
Artigo em Inglês | MEDLINE | ID: mdl-17824171

RESUMO

Models of estrogen insufficiency have revealed new and unexpected roles for estrogens in males as well as females. These models include natural mutations in the aromatase gene in humans, as well as mouse knock-outs of aromatase and the estrogen receptors, and one man with a mutation in the ERa gene. These mutations, both natural and experimental, have revealed that estrogen deficiency results in a spectrum of symptoms. These include loss of fertility and libido in both males and females; loss of bone in both males and females; a cardiovascular and cerebrovascular phenotype; development of a metabolic syndrome in both males and females, with truncal adiposity and male-specific hepatic steatosis. Most of these symptoms can be reversed or attenuated by estradiol therapy. Thus estrogen is involved in the maintenance of general physiological homeostasis in both sexes.


Assuntos
Aromatase/fisiologia , Estrogênios/fisiologia , Adiposidade , Animais , Aromatase/genética , Inibidores da Aromatase/uso terapêutico , Desenvolvimento Ósseo , Metabolismo Energético , Feminino , Humanos , Infertilidade Masculina/etiologia , Resistência à Insulina , Masculino , Camundongos , Camundongos Knockout , Área Pré-Óptica/fisiologia
12.
Horm Metab Res ; 37(1): 26-31, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15702435

RESUMO

The aromatase (ArKO) knockout mouse develops obesity marked by increased gonadal fat depots. This obesity is characterized by pronounced hypertrophy and hyperplasia in adipocytes with corresponding increases in transcripts involved in fat development. Aromatase deficiency in mice and humans with natural mutations of the aromatase gene also leads to metabolic syndrome, particularly hepatic steatosis. In ArKO mice, this hepatic steatosis, the increased body weight and serum triglycerides are surprisingly prevented by cholesterol feeding. We sought to investigate whether the reduction in body weight upon cholesterol feeding is reflected in gonadal fat depots, which account for a large percentage of body weight in the ArKO mouse. Indeed, gonadal fat depots in female ArKO mice were significantly reduced after cholesterol feeding. Concomitantly, adipocyte hyperplasia and hypertrophy were dramatically reduced upon cholesterol feeding in ArKO mice. Real-time PCR analysis revealed concurrent changes with adipocyte volume in the levels of lipoprotein lipase, caveolin-1 and CD59 transcripts. Little change was observed in levels of transcripts involved in de novo fatty acid synthesis, beta-oxidation, lipolysis, differentiation and cholesterol metabolism, suggesting that cholesterol feeding prevents hyperplasia and hypertrophy of ArKO adipocytes, possibly as a consequence of changes in transcript levels of lipoprotein lipase and therefore fatty acid uptake.


Assuntos
Adipócitos/patologia , Tecido Adiposo/enzimologia , Aromatase/fisiologia , Colesterol/administração & dosagem , Colesterol/metabolismo , Gorduras na Dieta/metabolismo , Tecido Adiposo/patologia , Administração Oral , Animais , Aromatase/deficiência , Composição Corporal/genética , Composição Corporal/fisiologia , Peso Corporal , Contagem de Células , Tamanho Celular , Gorduras na Dieta/administração & dosagem , Metabolismo Energético/fisiologia , Estrogênios/deficiência , Estrogênios/fisiologia , Feminino , Gônadas , Hiperplasia/metabolismo , Hiperplasia/prevenção & controle , Hipertrofia/metabolismo , Hipertrofia/prevenção & controle , Camundongos , Camundongos Knockout , PPAR gama/metabolismo , Distribuição Aleatória , Estatísticas não Paramétricas
14.
J Mol Endocrinol ; 32(2): 533-45, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15072557

RESUMO

A number of clinical studies have highlighted the importance of estrogen in bone growth and maintenance in men and postmenopausal women. In these instances, estrogen is synthesized locally within bone tissue by aromatase, encoded by the CYP19 gene. The mechanisms regulating aromatase expression in bone, however, are unclear. In this work we characterized the expression of aromatase activity and gene transcripts in the human fetal osteoblastic cell line, SV-HFO. Aromatase activity and gene transcript expression were stimulated by dexamethasone. Oncostatin M strongly stimulated aromatase expression in synergy with dexamethasone. These factors induced CYP19 transcripts that included the sequence of exon I.4 in their 5'UTR. Consistent with this, a reporter construct harboring the genomic sequence of the promoter region of exon I.4 (promoter I.4) was also activated by dexamethasone and oncostatin M. 5' deletion and mutation analysis revealed important roles for a glucocorticoid response element, an interferon gamma activating sequence and a putative binding site for Sp1. Transfection of exogenous glucocorticoid receptor, STAT3 or Sp1 increased promoter activity, indicating a potential role for these transcription factors in regulating aromatase expression in SV-HFO cells. These data suggest that the SV-HFO cell line is a valuable model with which to elucidate the mechanisms regulating local estrogen synthesis in osteoblasts.


Assuntos
Aromatase/genética , Aromatase/metabolismo , Osteoblastos/enzimologia , Regiões 5' não Traduzidas , Aromatase/efeitos dos fármacos , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA , Dexametasona/farmacologia , Embrião de Mamíferos/citologia , Éxons , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Oncostatina M , Peptídeos/farmacologia , Regiões Promotoras Genéticas , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Elementos de Resposta , Fator de Transcrição STAT3 , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Transativadores
15.
J Steroid Biochem Mol Biol ; 86(3-5): 225-30, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14623515

RESUMO

In premenopausal women, the ovaries are the principle source of estradiol, which functions as a circulating hormone to act on distal target tissues. However, in postmenopausal women when the ovaries cease to produce estrogen, and in men, this is no longer the case, because estradiol is no longer solely an endocrine factor. Instead, it is produced in a number of extragonadal sites and acts locally at these sites as a paracrine or even intracrine factor. These sites include the mesenchymal cells of adipose tissue including that of the breast, osteoblasts and chondrocytes of bone, the vascular endothelium and aortic smooth muscle cells, and numerous sites in the brain. Thus, circulating levels of estrogens in postmenopausal women and in men are not the drivers of estrogen action, they are reactive rather than proactive. This is because in these cases circulating estrogen originates in the extragonadal sites where it acts locally, and if it escapes local metabolism then it enters the circulation. Therefore, circulating levels reflect rather than direct estrogen action in postmenopausal women and in men. Tissue-specific regulation of CYP19 expression is achieved through the use of distinct promoters, each of which is regulated by different hormonal factors and second messenger signaling pathways. Thus, in the ovary, CYP19 expression is regulated by FSH which acts through cyclic AMP via the proximal promoter II, whereas in placenta the distal promoter I.1 regulates CYP19 expression in response to retinoids. In adipose tissue and bone by contrast, another distal promoter--promoter I.4--drives CYP19 expression under the control of glucocorticoids, class 1 cytokines and TNFalpha. The importance of this unique aspect of the tissue-specific regulation of aromatase expression lies in the fact that the low circulating levels of estrogens which are observed in postmenopausal women have little bearing on the concentrations of estrogen in, for example, a breast tumor, which can reach levels at least one order of magnitude greater than those present in the circulation, due to local synthesis within the breast. Thus, the estrogen which is responsible for breast cancer development, for the maintenance of bone mineralization and for the maintenance of cognitive function is not circulating estrogen but rather that which is produced locally at these specific sites within the breast, bone and brain. In breast adipose of breast cancer patients, aromatase activity and CYP19 expression are elevated. This occurs in response to tumor-derived factors such as prostaglandin E2 produced by breast tumor fibroblasts and epithelium as well as infiltrating macrophages. This increased CYP19 expression is associated with a switch in promoter usage from the normal adipose-specific promoter I.4 to the cyclic AMP responsive promoter, promoter II. Since these two promoters are regulated by different cohorts of transcription factors and coactivators, it follows that the differential regulation of CYP19 expression via alternative promoters in disease-free and cancerous breast adipose tissue may permit the development of selective aromatase modulators (SAMs) that target the aberrant overexpression of aromatase in cancerous breast, whilst sparing estrogen synthesis in other sites such as normal adipose tissue, bone and brain.


Assuntos
Aromatase/metabolismo , Estrogênios/biossíntese , Aromatase/genética , Estrogênios/metabolismo , Humanos , Especificidade de Órgãos , Precursores de Proteínas/metabolismo , Esteroides/metabolismo
16.
J Steroid Biochem Mol Biol ; 86(3-5): 239-44, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14623517

RESUMO

Intratumoral aromatase is a potential therapeutic target for the treatment of postmenopausal estrogen-dependent breast cancers. Therefore, reliable methods should be developed for routine application for the detection of intratumoral aromatase. A multi-center collaborative group has been established to generate and validate new aromatase monoclonal antibodies (MAbs). A recombinant GST-aromatase fusion protein was expressed in baculovirus and the purified protein was used for immunization of mice either as a native or formalin-fixed antigen. Hybridomas were generated using standard techniques and screened biochemically prior to immunohistochemistry (IHC) evaluation in human placenta, ovary and breast cancer tissues. Twenty-three MAbs selected by biochemical assays were further evaluated by IHC of paraffin-embedded tissue sections including normal ovary, and placenta, and a small series of 10 breast carcinomas. Of the 23 MAbs, 2 (clones 677 and F2) were determined to specifically stain cell types known to express aromatase in normal tissues. In breast carcinomas staining of malignant epithelium, adipose tissue, normal/benign and stromal compartments was detected. IHC was performed and independently evaluated by three pathologists (HS, TJA and SGS), each using the same evaluation criteria for staining intensity and proportion of immunopositive cells. With these two MAbs, interpathologist and intralaboratory variations were minimal in comparison with differences which could be detected between tissue specimens and antibodies.


Assuntos
Anticorpos Monoclonais/imunologia , Aromatase/imunologia , Aromatase/metabolismo , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/metabolismo , Aromatase/análise , Aromatase/genética , Western Blotting , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica/métodos , Camundongos , Ovário/enzimologia , Placenta/enzimologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo
17.
Genes Brain Behav ; 2(2): 93-102, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12884966

RESUMO

Estrogen has been suggested to play a neuromodulatory and neuroprotective role on the brain dopamine system. We used aromatase knockout (ArKO) mice that lack a functional aromatase enzyme and are unable to convert testosterone into estrogen, and assessed prepulse inhibition of acoustic startle, locomotor hyperactivity to amphetamine treatment and rotarod performance. Mice were tested at either 1 month, 4-5 months or 12-18 months of age. In male, but not female ArKO mice, there was an age-related reduction of prepulse inhibition. The 12-18 months old male ArKO mice also showed significantly greater amphetamine-induced hyperactivity. Mice heterozygous for the mutation showed no deficits or were in-between wildtype mice and ArKO mice. We postulate that these data indicate a neuroprotective role of estrogen, particularly in male mice, on ageing of brain mechanisms involved in pre-pulse inhibition and locomotor activity regulation. It is likely that these brain mechanisms are or include dopaminergic activity.


Assuntos
Envelhecimento/fisiologia , Aromatase/fisiologia , Reflexo de Sobressalto/fisiologia , Caracteres Sexuais , Estimulação Acústica , Animais , Aromatase/deficiência , Feminino , Genótipo , Habituação Psicofisiológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Inibição Neural
18.
Mol Cell Endocrinol ; 193(1-2): 7-12, 2002 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-12160996

RESUMO

Aromatase, the enzyme responsible for the conversion of androgens to estrogens, is present in the mouse gonads, brain, adipose tissue and bone. Depletion of endogenous estrogens in the aromatase deficient mouse (ArKO) caused by the targeted disruption of the Cyp19 gene resulted in an impairment of sexual behaviour and an age-dependent disruption of spermatogenesis. This disruption occurred during early spermiogenesis, due possibly to increased number of apoptotic round spermatids. Development of obesity was associated with ageing, decrease in lean mass, hypercholesterolemia, hyperleptinemia, and insulin resistance and hepatic steatosis. However, it was not correlated with hyperphagia but to decreased physically-active behaviour. ArKO mice also developed osteoporosis. Thus, studies using the ArKO mice model has led to several insights into the multiple roles played by estrogens in the development and maintenance of fertility, sexual behaviour, lipid metabolism and bone remodelling.


Assuntos
Aromatase/genética , Estrogênios/deficiência , Estrogênios/fisiologia , Animais , Aromatase/deficiência , Humanos , Metabolismo dos Lipídeos , Camundongos , Camundongos Knockout , Modelos Animais , Mutação , Osteoporose , Espermatogênese
19.
Endocrinology ; 142(11): 4589-94, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11606422

RESUMO

There is a growing awareness that androgens and estrogens have general metabolic roles that are not directly involved in reproductive processes. These include actions on vascular function, lipid and carbohydrate metabolism, as well as bone mineralization and epiphyseal closure, in both sexes. In postmenopausal women, as in men, estrogen is no longer solely an endocrine factor, but instead is produced in a number of extragonadal sites and acts locally at these sites in a paracrine and intracrine fashion. These sites include breast, bone, vasculature, and brain. Within these sites, aromatase action can generate high levels of E2 locally without significantly affecting circulating levels. Circulating C(19) steroid precursors are essential substrates for extragonadal estrogen synthesis. The levels of these androgenic precursors decline markedly with advancing age in women, possibly from the mid to late reproductive years. This may be a fundamental reason why women are at increased risk for bone mineral loss and fracture and possibly decline of cognitive function, compared with men. Aromatase expression in these various sites is under the control of tissue-specific promoters regulated by different cohorts of transcription factors. Thus, in principle, it should be possible to develop selective aromatase modulators that block aromatase expression, for example, in breast, but allow unimpaired estrogen synthesis in other tissues such as bone.


Assuntos
Aromatase/fisiologia , Estrogênios/biossíntese , Envelhecimento/metabolismo , Animais , Aromatase/genética , Humanos , Precursores de Proteínas/metabolismo
20.
J Androl ; 22(5): 825-30, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11545296

RESUMO

Previous studies employing the male aromatase knockout (ArKO) mouse have indicated that local expression of estrogens appears to be important for the progression of spermatogenesis. In the absence of estrogen biosynthesis round spermatids are observed to undergo apoptosis and thus fail to differentiate into mature, elongated spermatids. This lesion appears to arise between the ages of 18 weeks and 1 year. To ultimately determine if the disruption to spermatogenesis arises earlier than 18 weeks, we performed an intensive study to examine the fertility of younger male ArKO mice. This involved an analysis of their mating capacity together with an extensive stereological analysis, determination of the in vitro potential of mature sperm, and sexual behavior. ArKO and wild-type (w/t) males at 7 weeks of age were placed with w/t females for 7 weeks. At age 14 weeks, the males were killed and the testes removed. ArKO mice were observed to sire significantly fewer litters than the w/t mice; 5 out of the 10 sired no litters at all. Stereological analysis performed on the removed testes found a significant decrease in round spermatid numbers between w/t and ArKO mice at this age; however, there were no differences in all other germ cells and Sertoli cell numbers. When mature spermatozoa were analyzed, sperm from 15-week-old ArKO mice had a significant reduction in motility. This was further reduced by 1 year of age with a decrease in concentration. A preliminary examination of sexual behavior found that ArKO mice did not attempt to mount the females, in contrast to the w/t mice, which mounted consistently during the time period. In conclusion, we observed that ArKO mice have reduced fertility at age 14 weeks. This may be due in part to a disruption in spermatogenesis because the phenotype does appear to arise earlier than 18 weeks, possibly leading to abnormalities in the mature spermatozoa. Or, in part, this may be attributable to an impairment in the development of copulatory behavior, which is consistent with the available evidence that points to a crucial role for estrogens in the neural development and initiation of male sexual behavior.


Assuntos
Aromatase/fisiologia , Fertilidade/genética , Animais , Aromatase/genética , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Comportamento Sexual Animal , Espermatogênese/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA